Sie sind auf Seite 1von 353

Introduction to Diffusion Tensor

Imaging 2
Louise Emsell, Wim Van Hecke,
and Jacques-Donald Tournier

are the (mean) apparent diffusion coefficient


Learning Points (ADC) and fractional anisotropy (FA).
• The primary application of DTI is in (pre)
• Diffusion tensor imaging (DTI) is one of sev- clinical research. It is also used in clinical
eral diffusion MRI-based techniques that can practice for assessing the evolution of stroke
be used to noninvasively, indirectly assess tis- and for neurosurgical planning.
sue macro- and microstructure. • Acquiring and analyzing DTI data is not triv-
• DTI can be used as a visualization tool to dis- ial and requires careful consideration of many
tinguish between large oriented macromolec- choices throughout the acquisition and analy-
ular structures, such as white matter fiber sis pipeline.
bundles in the brain. • DTI is highly sensitive to changes in diffu-
• Several scalar quantitative parameters can be sion, but lacks specificity, which confounds
derived from DTI, the most common of which the interpretation of underlying biological or
pathological processes.
• DTI suffers from a number of limitations that
L. Emsell, PhD (*)
confound the interpretation of quantitative
Translational MRI, Department of Imaging and
Pathology, KU Leuven and Radiology, University measures and the accuracy of fiber tracking.
Hospitals Leuven, Herestraat 49, 3000 Leuven,
Belgium It is arguably one of the most beautiful imag-
Universitair Psychiatrisch Centrum (UPC), KU ing techniques presently available in the field of
Leuven, Leuven, Belgium radiology, and has captured the imagination of
e-mail: louise.emsell@med.kuleuven.be
neuroscientists and clinicians in equal measure.
W.V. Hecke, PhD However, diffusion tensor imaging (DTI) should
icometrix, Leuven, Belgium
not be held aloft simply for its artistic merit. DTI
Department of Radiology, Antwerp University has revolutionized the management of acute isch-
Hospital, Wilrijkstraat 10, Edegem, 2650 Antwerp,
emia and the assessment of brain lesions. Its abil-
Belgium
ity to visualize and segment white matter fiber
J.-D. Tournier, PhD
bundles has opened up new perspectives for neu-
Florey Institute of Neuroscience and Mental Health,
University of Melbourne, Parkville, VIC, Australia rosurgeons. Its capacity to quantify diffusion
within neural tissue has provided clinical research
Biomedical Engineering, Division of Imaging
Sciences and Biomedical Engineering, King’s with a remarkable tool with which to investigate
College London, London, UK the living brain in health and disease; and beyond

© Springer Science+Business Media New York 2016 7


W. Van Hecke et al. (eds.), Diffusion Tensor Imaging, DOI 10.1007/978-1-4939-3118-7_2
8 L. Emsell et al.

the brain, DTI continues to gain ground in the scale intensities that reflect the tissue property to
assessment of muscle and other organs such as the which the imaging sequence is sensitized. So for
kidney and prostate. Yet despite such promise, example, in T1-weighted images, fluid will
since its inception two decades ago [1], the impact appear dark, whilst in T2-weighted images, fluid
of DTI in general radiological practice has been will appear bright. The seasoned radiologist will
surprisingly limited. Possible reasons for this quickly be able to draw upon their knowledge of
include, amongst others, the relatively long time it how different types of tissue appear on different
takes to acquire and process the data compared to scans to distinguish between normal tissue and
conventional clinical MRI methods; the vast that affected by pathology, and be able to describe
number of different approaches to data acquisi- approximately where such pathology is located
tion and analysis; and the poor accuracy of the in anatomical terms. However, such conventional
technique and its sensitivity to a number of fac- scans are not intrinsically sensitive to the micro-
tors (described in this book), which means that structural architecture of the tissue and do not
maps and measures cannot be reliably reproduced explicitly capture or account for the influence of
under different conditions, such as across MRI tissue orientation on the MRI measurements.
scanners or between individual patients. These This is the added value of DTI, which allows it to
important caveats have been a stumbling block to be used to identify ischemic changes before they
the widespread adoption of DTI in the clinic, are visible on non-diffusion-weighted scans and
where being able to obtain reliable information on to map the orientation of (and thus differentiate
disease status in as short a time frame as possible between) white matter fiber bundles that cannot
is paramount. Recognizing and understanding be distinguished using any other imaging tech-
how such challenges could be overcome so that nique (Fig. 2.1).
DTI can be used optimally in a clinical setting,
either in research or potentially in clinical practice
in the future, forms the basis of this book. How Does DTI Work?
This opening chapter provides an introduction
to the DTI technique. The following pages pro- In an environment in which there are no obstacles
vide a brief overview of topics that will be cov- in their path, such as in a glass of water, mole-
ered in detail in the remainder of the book. In the cules jostling about due to thermal motion will
first part of the chapter, the biophysical basis of disperse in a uniform manner, traveling an equal
DTI is introduced, as well as the quantitative mea- distance in all directions. This is termed isotropic
sures that can be derived from it and some exam- diffusion. However, if the molecules encounter
ple applications. In the second part of the chapter, obstructions that are coherently oriented, they
the typical DTI pipeline is introduced, from the will no longer disperse equally in all directions,
initial hypothesis, through data acquisition, pro- and diffusion will be anisotropic. When consid-
cessing and analysis to interpretation. The final ered in the simplest terms, diffusion-weighted
section of the chapter highlights the pros and cons MRI measures the net displacement of water
of DTI in the context of different applications and molecules in a voxel over a few milliseconds. By
considers the potential role of DTI and related dif- measuring the degree and direction of diffusion,
fusion MRI techniques in the future. it aims to infer the structure of the local environ-
ment of the diffusing molecules. Consider that
during the short time frame of the diffusion mea-
Introducing the DTI Technique surement, the water molecules will travel a dis-
tance of several micrometers. If, during this time,
A Useful Contrast the water molecules are not obstructed in any
way, such as in cerebrospinal fluid, then the mea-
Classical clinical MRI techniques provide a sin- sured MRI signal will be approximately the same
gle scalar value for each image pixel (or, in 3D, in all directions. However, if the molecules inter-
each voxel), yielding an image made up of grey act with a boundary, such as a cell membrane,
2 Introduction to Diffusion Tensor Imaging 9

Fig. 2.1 Improved directional contrast with DTI: (a) On tinguishing between different fiber bundles. For example,
a T1-weighted image, white matter appears homoge- the cingulum bundle running anterior–posteriorly (green)
neous. (b) DTI can be used to generate diffusion anisot- is easily distinguished from the corpus callosum running
ropy (e.g., Fractional Anisotropy, FA) maps that can be ventromedially (red) on the color FA map (orange arrow),
color coded according to the principal direction of diffu- compared to the T1 image. Using the DEC convention,
sion, termed “directionally encoded color,” or DEC maps. diffusion in an anterior–posterior direction is green, ven-
The DEC-FA map is calculated by multiplying the FA tromedial is red, and superior–inferior is blue. [Images
map with the first eigenvector map. This is useful for dis- courtesy of T. Billiet and A. Leemans]

then their pathway will be obstructed, altering the the diffusion gradients are applied (typically
measured MRI signal. Moreover, if these bound- around 40 ms in a clinical scanner) and the dis-
aries are coherently oriented, as for example tance that separates the water molecules from a
within a white matter tract, then the measured physical obstacle. In brain tissue, for example,
signal will also be affected differently depending diffusing molecules will travel several microme-
on the orientation of the diffusion gradients rela- ters during a typical diffusion measurement. This
tive to the orientation of the tissue (Fig. 2.2). means that the diffusion signal is most sensitive
In brain tissue, there are many obstructions to to changes in microstructure within this range of
diffusion that molecules may encounter at the spatial scales [3].
length scale of the MRI measurement, such as It is also important to note that the typical spa-
macromolecules, organelles, and cell mem- tial resolution of a DTI scan is 2–3 mm, meaning
branes. These boundaries will give rise to both that diffusion across many heterogeneous
hindered and restricted diffusion, which is often microstructural environments, including differ-
attributed to water trapped within and between ent cell types, sizes, densities etc., contributes to
cells respectively. Therefore, by measuring the average signal in each image voxel.
changes in the MRI signal along different direc- In addition to the degree of potential molecu-
tions, it is possible to learn something about the lar displacement, the snapshot of diffusion also
underlying tissue architecture [2]. For example, depends on the traditional fundamental sources
in the case of white matter, the orientation of of contrast in the MRI signal, i.e., T1 and T2. In
axons strongly influences the direction of diffu- a typical DTI sequence, a long echo time is typi-
sion [3], which forms the basis for diffusion trac- cally unavoidable, which means that some impor-
tography (described later in this chapter and in tant microstructural elements with short T2
Chaps. 8, 11 and 12) (Fig. 2.3). relaxation times do not contribute to the mea-
It is worth noting, however, that the DTI tech- sured signal, notably myelin water. Concepts of
nique only provides a snapshot of diffusion diffusion in biological tissues and measuring it
within tissue over a given time frame and length with MRI are introduced in the following chapter
scale, which is determined by the duration that (Chap. 3).
10 L. Emsell et al.

Fig. 2.2 Compared to the


non-diffusion-weighted
image (center), the contrast
in the six diffusion-
weighted images (outer
images) changes as a
function of the orientation
of the tissue relative to the
direction of the applied
diffusion gradient

Diffusion tensor imaging extends these con- assessments, such quantitative measures can
cepts further by modeling diffusion as a mathe- be used to statistically compare patient groups
matical tensor, to describe the amount and with healthy subjects, or quantify changes over
direction of diffusion in each voxel by relating time, which may reflect normal developmental
the measured signal to the applied diffusion- and neurodegenerative processes or stages of
sensitizing gradient [1]. The tensor can be repre- disease.
sented as an ellipsoid whose main axis represents The most clinically useful DTI measures are
the principal direction of diffusion, and which presently the mean apparent diffusion coefficient
forms the basis of the most common quantitative (ADC) (alternatively, the “trace” or mean diffu-
DTI measures (Fig. 2.4) [4]. sivity (MD)), which describe the total amount of
diffusion in a given voxel. The most commonly
used metric in research studies is fractional
A Quantitative Technique anisotropy (FA), which characterizes the degree
of anisotropy in each voxel. The FA is consid-
One of the great features of the DTI technique is ered a summary metric describing the general
its ability to capture information about complex status of the underlying tissue architecture. For
microstructure and summarize it with a few example, a change in microstructural configura-
simple quantitative metrics derived from the dif- tion, such as cell loss, changes in cell density, or
fusion tensor. Unlike qualitative radiological increases in tissue water content, will result in a
2 Introduction to Diffusion Tensor Imaging 11

Fig. 2.3 Schematic representation illustrating how the direction of diffusion can then be followed in each voxel
diffusion tensor relates to axonal architecture. (a) to reconstruct a path through the image. This is known as
Coherently organized structures such as axons give rise to tractography or fiber tracking. (d) Many such tractogra-
anisotropic diffusion. (b) anisotropy can be characterized phy streamlines can be displayed together to form virtual
using the diffusion tensor, which can be represented geo- reconstructions, which may provide a reasonable approxi-
metrically as a three-dimensional ellipsoid. The longest mation of macroscopic anatomical white matter fiber
axis of the ellipsoid represents the principal direction of bundles. Note the difference in scale between (a, b) and
diffusion and is assumed to reflect the underlying axonal (c, d). [Adapted from Beaulieu C. The basis of anisotropic
orientation. (c) The contribution of many microscopic water diffusion in the nervous system—a technical review.
determinants of anisotropy is averaged and represented by NMR Biomed. 2002 Nov–Dec;15(7–8):435–55. With
a single tensor ellipsoid in each voxel. The principal permission from John Wiley & Sons, Inc.]

change in FA. It is important to understand, matter fiber bundles using tractography. In the
however, that FA is only a very indirect measure- clinic, this translates to the investigation of sus-
ment based on an oversimplified model of the pected acute ischemic stroke, to differentiate
behavior of diffusing water molecules averaged vasogenic versus cytotoxic oedema and to char-
over the scale of the voxel. This model makes acterize intracranial lesions such as pyogenic
many assumptions that are not satisfied in real abscess, infections, tumors, and trauma [7].
tissue. Moreover, the measurement of FA, and However, the predominant use of DTI is as a
other DTI-derived parameters are strongly influ- (pre)clinical research tool, where it has been
enced by many different factors related both to applied to study, most typically, white matter
the MRI technique itself, such as system-induced microstructure in a broad range of neurological
noise and artifacts, and physiological factors and psychiatric disorders [8].
such as the effects of perfusion and partial vol- The ability to estimate a principal direction of
ume. For these reasons, DTI parameters such as diffusion using the tensor has also yielded the
FA, MD, etc. should only ever be interpreted as tractography technique, which has been applied
a very indirect approximation of microstructural to study brain connectivity [9]. Essentially, trac-
status [5, 6]. tography aims to virtually dissect white matter
fiber bundles by propagating “streamlines” that
follow the main direction of diffusion, which is
Applications assumed to reflect the underlying orientation of
axons (Fig. 2.3). In one class of tractography
The main applications of DTI are in the assess- applications, the dissected bundles are simply
ment of tissue microstructure, predominantly in used as a visualization tool to locate the boundar-
brain tissue (although the number of non-brain ies of fiber bundles which otherwise cannot
applications is increasing) and to segment white be distinguished on conventional MRI images.
12 L. Emsell et al.

Fig. 2.4 The diffusion tensor. (a) geometric representa- map. The second and third eigenvalues are used to calcu-
tion of the diffusion tensor and derivative DTI parameter late the radial diffusivity (RD) (sometimes referred to as
maps. The diffusion tensor, (b(i)), can be mathematically λ⊥, transverse or perpendicular diffusivity). The mean
decomposed (b(ii)), into eigenvectors (b(iii)) represent- diffusivity (MD) is a measure of the overall diffusivity in
ing the direction of diffusion, and eigenvalues (b(iv)) rep- a particular voxel regardless of direction and is calculated
resenting the magnitude of diffusion. In the geometric as the average of the eigenvalues. The degree of anisot-
representation, the eccentricity of the ellipsoid character- ropy can be represented by the fractional anisotropy
izes the degree of diffusion anisotropy. The longest axis (FA), a scalar measure, without units, ranging between 0
is the first eigenvector and represents the direction of (isotropic—darkest grey on a standard FA map) and 1
maximal diffusion, along which the axial diffusivity (anisotropic—lightest grey on a standard FA map).
(AD) is calculated (sometimes referred to as λ||, longitudi- Further details about how to calculate and interpret FA
nal, or parallel diffusivity). The first eigenvector is also and other DTI measures are provided in Chap. 5. [Images
used to calculate the directionally encoded color (DEC) courtesy of A. Leemans]

This has been used in neurosurgical applications. the reconstructed streamlines do not reflect the
In another application, the segmented bundles are underlying anatomy. This major caveat of DTI is
used as regions of interest. One of the more con- now widely accepted in the scientific field and
troversial uses of tractography is to quantify rela- has resulted in a concerted effort to develop new
tionships between DTI tractography derived modeling techniques for assessing brain connec-
measures and functional grey matter regions to tivity [10].
infer something about the underlying brain con- It is important to understand that although
nectivity. This latter application is confounded by DTI is one of the most widely used implementa-
the fact that DTI cannot characterize more than tions, it is just one of a number of techniques
one dominant fiber direction in a voxel, and thus based on diffusion MRI (dMRI), all of which can
2 Introduction to Diffusion Tensor Imaging 13

be used to characterize microstructure in vivo. spending the extra time, cost, and potential
It is therefore incorrect to equate or synonymize inconvenience associated with extending an
DTI with other dMRI-based methods, or consider existing scan protocol by an extra 5–15 min.
it to be the only means to assess white matter The nature and extent of any hypothesized
“integrity” in vivo. The simplest form of dMRI is changes has an important impact on the choices
the basic diffusion-weighted scan, which is used that need to be made with regard to both the DTI
widely in clinical practice to assess early isch- data acquisition and analysis strategies. For
emic injury; DTI is a relatively simple extension example, if the goal of the DTI study is to iden-
of this. Beyond DTI, there are a host of “higher- tify acute ischemia, a simple <6 direction DWI
order” models, some of which can be applied to acquisition of one or two minutes will suffice to
“DTI” data (i.e., acquired using the same proto- calculate the ADC. However, if the goal is to
col) and some that require different dMRI acqui- visualize fiber tracts as part of a presurgical plan-
sition strategies, for example, acquiring data with ning work-up, a much longer, more advanced
several different b-values or many more gradient sequence is required including many more gradi-
directions [10]. Such higher-order models are ent directions (>30), greater diffusion weighting
sometimes encompassed under the general (i.e., b-value), and more advanced (non-DTI)
umbrella term “high-angular resolution diffusion reconstruction techniques. It is therefore impor-
imaging” or HARDI. Although strictly speaking, tant to consider what the motivation and ultimate
the term HARDI refers to the data acquisition aim of the DTI investigation is prior to data
strategy and higher-order models refers to the collection.
data reconstruction method, the two terms gener- In practice, particularly in a research context,
ally apply to more advanced, non-DTI-based a clear hypothesis may be lacking and DTI data is
dMRI methods (see Chap. 20). collected with a view to future exploratory analy-
sis. In such situations, it is often helpful to at least
anticipate what type of analysis might be con-
The DTI Pipeline ducted in the future by, for example, consulting
current scientific literature, and to collect data of
Designing and carrying out a DTI study in an sufficient quality and with a sufficient number of
optimal manner, either on an individual patient or gradient directions to be able to perform a range
in a research study, is not trivial. Extracting the of different analyses. There are few things more
rich, multivariate information captured in the dif- frustrating than spending time collecting data
fusion signal requires careful attention at many only to discover later that it is unsuitable due to
different stages in the study pipeline (Fig. 2.5) poor quality or an inadequate imaging sequence.
[11]. This next section introduces some of these The importance of optimizing the DTI acquisi-
considerations, which form the framework of the tion protocol before collecting data, and checking
remainder of this book. data quality at the time of scanning or as soon as
possible thereafter, cannot be stressed enough.

The Goal of the DTI Investigation


Data Acquisition
Although often taken for granted, the motivation
for undertaking a DTI study is most usually the As anyone working with MRI data will under-
expectation that there may be a measurable dif- stand, there are myriad parameter settings avail-
ference or change in microstructural properties able that all interact to give rise to the final image.
in a given individual or group of individuals that In conventional imaging, manipulating basic
can be correlated with a pathological or natural parameters will weight the sequence to a particu-
biological process such as neurodevelopment or lar tissue property, such as T1, T2, diffusion, per-
aging. This motivation must exist to justify fusion etc., alter the contrast and signal to noise
14 L. Emsell et al.

Fig. 2.5 Prototypal DTI study pipeline. Whole-brain differences in euthymic bipolar I disorder: a combined
tractogram and connectivity matrix. [Reprinted from magnetic resonance imaging and diffusion tensor imaging
Caeyenberghs K, Leemans A, Leunissen I, Gooijers J, voxel-based study. Bipolar Disord. 2013 Jun;15(4):
Michiels K, Sunaert S, et al. Altered structural networks 365–376. With permission from John Wiley & Sons.]
and executive deficits in traumatic brain injury patients. Axon micrograph. [Reprinted from Beaulieu C. The basis
Brain Struct Funct. 2014 Jan;219(1):193–209. With per- of anisotropic water diffusion in the nervous system—a
mission from Springer Verlag]. Voxel-based analysis technical review. NMR Biomed. 2002 Nov–Dec;15
figure. [Adapted from Emsell L, Langan C, Van Hecke W, (7–8):435–455. With permission from John Wiley &
Barker GJ, Leemans A, Sunaert S, et al. White matter Sons, Inc.] Corrected DTI maps. [Courtesy of A. Leemans]

ratio (SNR), and the spatial resolution of the ing more will improve the estimation of the diffu-
image. These same principles apply equally to sion tensor [13], and acquiring even more (e.g.,
DTI; however, the acquisition of both non- >45) will allow more advanced reconstruction
diffusion and multiple diffusion-weighted images methods to be used that may be able to resolve
adds an extra level of complexity. For example, complex fiber architecture [10]. However,
one must decide along how many noncollinear acquiring more directions will extend the scan
gradient directions diffusion weighting will be time and thus decrease tolerability and increase
applied. Mathematically, while only six are the likelihood of motion artifacts, which may
required for standard DTI [12] in practice acquir- degrade image quality. This is just one example
2 Introduction to Diffusion Tensor Imaging 15

out of many more that are described in Chap. 6. Data Analysis


The take-home message however is that it pays to
spend some time becoming familiar with the Having acquired and pre-processed the data, the
parameter choices that are especially relevant in next logical step is deciding how to extract useful
DTI, such as the degree of diffusion weighting information from it. Clearly, this will depend on
(b-value), the number and orientation of the the motivation for acquiring the data in the first
applied diffusion gradients, the echo time (TE), place, e.g., is it for visualization or quantification,
the need for adequate fat suppression and so on, for clinical use in an individual patient, or in a
and deciding what is the most optimal balance group study as part of clinical research?
between scan time and image quality for your Considering these types of questions prior to ana-
particular application [14]. lyzing the data (or preferably, before acquiring it)
will help in choosing which of the many available
analysis strategies to use to gain the most useful
Data Pre-processing and reliable insights. This topic is discussed and
a decision tree to aid such a thought process is
Having collected the raw DTI data (i.e., a series provided in Chap. 8 (Strategies and challenges in
of diffusion-weighted image volumes, each com- DTI analysis).
prising a number of 2D images with a particular Briefly, DTI analysis strategies can be divided
diffusion weighting), it is necessary to perform a into three main classes: (1) techniques that can be
number of steps in order to both correct the data used to assess the whole structure or organ (e.g.,
for typical artifacts associated with the EPI brain) as a single entity, (2) techniques that assess
sequence used to acquire the data, and to gener- a specific region (e.g., a specific white matter
ate DTI scalar maps (e.g., FA maps). For exam- fiber bundle or pathological lesion), and (3) tech-
ple, eddy currents induced by the rapid switching niques that assess individual voxels, and from
of the applied diffusion gradients often affect EPI this infer local alterations in DTI metrics.
sequences, which warp the DWIs along the direc- Examples include histogram analysis, manual or
tion of phase encoding. The relatively long scan automated tracing of lesions, tractography-based
time means that bulk motion effects are unavoid- segmentation of fiber bundles, voxel-based anal-
able in awake subjects. These geometric distor- ysis (VBA), and graph-based analysis of “con-
tions are often corrected by realignment to a nectivity” (Fig. 2.6).
non- or less deformed image such as a non- Each technique has its own strengths and limi-
diffusion-weighted image (sometimes known tations, and will be more or less suited to a given
informally as the “b = 0”) [10]. application than another. As the different
There are also different approaches for esti- techniques provide different information and are
mating the diffusion tensor from the raw DWI strongly influenced by their underlying assump-
data, which will have an impact on the accuracy tions and methodological implementation, it is
of any quantitative measures derived from it [15]. often useful to perform more than one type of
In fact, there are many such “under the hood” cal- analysis to gain insight into any observed
culations and image processing steps that con- changes. The greatest insights however may be
tribute to generating the final DTI parameters and achieved by combining DTI with other imaging
derivative maps, and which are easily over- modalities, and with functional and clinical
looked. While this may be due mainly to a lack of measurements.
awareness, it does highlight the need to work
with or consult specialists when collecting and
analyzing DTI data. Image pre-processing for Interpretation
DTI analysis is addressed in more detail in Chaps
8-11 of this book, and Chaps. 4 (from raw DWI Interpretation in the context of DTI imaging can
to tensor) and 7 (checking and correcting data) in be understood in terms of the qualitative visual
particular. interpretation of the DTI derived maps, or the
16 L. Emsell et al.

Fig. 2.6 DTI analysis methods: (a) Whole-brain tractog- matter differences in euthymic bipolar I disorder: a com-
raphy, (b), region-based tractography, (c) graph-based bined magnetic resonance imaging and diffusion tensor
connectivity models, (d) histogram analysis, (e) region- imaging voxel-based study. Bipolar Disord. 2013
of-interest analysis, and (f) voxel-based analysis. (a and 15(4):365–76. With permission from John Wiley & Sons,
c) [Reprinted from Caeyenberghs K, Leemans A, Inc.] (f) (right): [Reprinted from Holleran L, Ahmed M,
Leunissen I, Gooijers J, Michiels K, Sunaert S, et al. Anderson-Schmidt H, McFarland J, Emsell L, Leemans
Altered structural networks and executive deficits in trau- A, et al. Altered interhemispheric and temporal lobe
matic brain injury patients. Brain Struct Funct. 2014 white matter microstructural organization in severe
Jan;219(1):193–209. With permission Springer Verlag]. chronic schizophrenia. Neuropsychopharmacology. 2014
(f) (left): [Reprinted from Emsell L, Langan C, Van Mar;39(4):944–54. With permission from Nature
Hecke W, Barker GJ, Leemans A, Sunaert S, et al. White Publishing Group]

interpretation of changes in DTI parameters with lesion differentiation, the clinical interpreta-
detected by quantitative statistical analysis. With tion of the DTI parameter maps. Data quality
respect to the former, visual interpretation assessment is a vital and essential step in any DTI
involves both the assessment of the raw and pro- study and should not be neglected. The presence
cessed data quality, and, in some cases, such as of artifacts in the raw DWI data results in the
2 Introduction to Diffusion Tensor Imaging 17

misestimation of DTI parameters, and it is there- gation, and will also determine the quality and
fore important to be able to recognize them or nature of the final results. It pays to spend time
their consequences in the derived parameter becoming familiar with these considerations and
maps and tractograms [16]. Chapters 6 and 7 choices, not only to optimize a given investiga-
contain many pictorial examples of such arti- tion but also to aid the understanding and assess-
facts, as well as information about how they can ment of other DTI-based studies and findings
be avoided and corrected using different acquisi- reported in clinical and scientific literature.
tion and image processing strategies. There are many pros and cons of DTI (see
The interpretation of statistically significant information box: “pros and cons of DTI”) and the
changes in quantitative parameters is particu- art of using the technique optimally (which also
larly challenging. Although DTI is highly sensi- includes deciding whether it is appropriate for a
tive to changes in diffusion, it lacks specificity given application at all) necessarily involves a
and the derived parameters are strongly influ- careful cost-benefit analysis for a given applica-
enced by many factors. As it is extraordinarily tion. For example, scientists conducting explor-
difficult to disentangle the relative contribution atory research studies on large numbers of
of these biological and methodological factors to subjects may be less concerned with limitations
a given DTI parameter value in a given voxel, it in estimated fiber bundle trajectories than a neu-
is simply not possible to ascribe changes in DTI rosurgeon planning an operation on a patient. In
parameters to changes in specific microstruc- this context, it is clear that the limitations of DTI
tural elements such as the degree of myelination, assume greater significance, and in some applica-
or axon density [5]. In this context, although tions may outweigh the benefits, in a clinical
DTI may be influenced by changes in micro- setting (see Box 2.1).
structural integrity, it is by no means a direct
measure of it. Nevertheless, given its sensitivity
to changes in diffusion, DTI remains a useful The Future Role of DTI in Clinical
tool for noninvasively identifying potential dif- Practice
ferences attributable to disease status, and when
combined with other evidence can contribute In comparison to other radiological techniques, a
valuable complementary information about dichotomy exists between the perceived utility of
pathological mechanisms, treatment effects, and DTI in the clinic and in scientific research. Whilst
neurobiology. DTI is sometimes considered to be an advanced,
Chapter 5 (Quantitative DTI parameters) pro- somewhat novel technique on the brink of enter-
vides a detailed overview of this topic and guide- ing mainstream clinical practice, a large body of
lines for interpreting changes in quantitative the diffusion MRI scientific community is mov-
parameters. ing away from DTI-based imaging and develop-
ing new strategies to obtain information captured
from the diffusion signal that aim to overcome
The DTI Balancing Act some of the limitations of the technique. However,
despite this transition away from DTI in the
Of all the possible take-home messages, one of diffusion MRI field, and the fact that it is not
the most useful to remember is that with DTI, widely used in clinical practice, DTI is used
there is no “one-size fits all.” Carrying out a DTI extensively in research, having been applied in
investigation optimally is not to say there is ever thousands of (pre)clinical research studies to date.
a perfect solution or implementation of the tech- It is worth considering why this dichotomy
nique. As will become apparent throughout this exists. In order to be clinically useful, a given
book, the myriad choices that need to be made at technique or method of assessment should logi-
multiple steps throughout the DTI pipeline will cally satisfy a number of criteria. For example, it
be influenced by the ultimate goal of the investi- should be performed as quickly and reliably as
18 L. Emsell et al.

possible, and provide well-defined measures that


Box 2.1: Pros and Cons margins are different are reproducible across different clinical centers
Strengths and defined by standard, normative ranges. It
+ Can be used for in vivo assessment of should provide information that is complemen-
white matter tary to other clinical measures by providing addi-
+ Simple way of summarizing complex tional information that is not revealed by other
data techniques. It should have diagnostic or prognos-
+ Can visualize the location of major white tic utility; for example, by being able to detect
matter bundles pathology, distinguish between different diseases
+ Sensitive to changes in macro- and or for staging disease processes. Preferably, it
microstructure should also be inexpensive, accessible, and easy
+ Provides quantitative measures to implement on a large scale. When considering
+ Noninvasive such criteria, it is clear why DTI has not yet been
+ Useful complementary technique to con- adopted in mainstream clinical practice. At the
ventional MRI same time, one can also appreciate why after
+ Can be acquired in a relatively short time decades of failing to penetrate into clinical prac-
(5–10 min) tice it has not yet been abandoned. The potential
+ Has clinical utility in the assessment of of DTI to provide unique clinical solutions and
acute stroke complementary biomarkers in some areas of
+ Has potential complementary clinical medicine remains, and it is this potential that is
utility in neurosurgical planning presently fueling the substantial research efforts
+ Group analysis has provided useful in this domain.
insights in neurology and psychiatry Since the inception of DTI, there have been sig-
Limitations nificant technological and methodological
− Based on an oversimplified model advances in the field, as well as a greater aware-
− Not specific to a particular microstruc- ness of the limitations of the technique. These
tural feature (e.g., myelin content, axon advances in technology and understanding con-
density etc.) tinue to contribute to improvements in data qual-
− Unreliable in voxels with >1 dominant ity, to reducing scan time and to relating the
fiber direction: “crossing-fibers” (i.e., diffusion signal to the microarchitectural proper-
most of the brain white matter) ties that DTI aims to probe. Nowadays, it is per-
− Dependent on data quality haps more appropriate to think of DTI as just one
− Limited spatial resolution out of a range of diffusion MRI-based techniques
− Dependent on data acquisition (see Chaps 20 and 21). Its future utility in clinical
parameters practice and that of its relatives will ultimately be
− Quantitative measures influenced by determined by their ability to satisfy the aforemen-
many non-biological factors tioned criteria and improve patient healthcare.
− Changes in quantitative measures are
difficult to interpret Acknowledgements The authors would like to thank
− Results are user and parameter Thibo Billiet and Alexander Leemans for providing
images that were incorporated into the figures in this
dependent.
chapter.
− Different scanners have different acqui-
sition settings confounding reproduc-
ibility and standardization. References
− Cannot differentiate the directionality of
axons: afferent vs. efferent, anterograde 1. Basser PJ, Mattiello J, LeBihan D. Estimation of the
vs. retrograde pathways, inhibitory vs. effective self-diffusion tensor from the NMR spin
echo. J Magn Reson B. 1994;103(3):247–54.
excitatory connections, direct vs. indirect 2. Basser PJ, Jones DK. Diffusion-tensor MRI: theory,
routes experimental design and data analysis—a technical
review. NMR Biomed. 2002;15(7–8):456–67.
2 Introduction to Diffusion Tensor Imaging 19

3. Beaulieu C. The basis of anisotropic water diffusion 10. Tournier JD, Mori S, Leemans A. Diffusion tensor
in the nervous system: a technical review. NMR imaging and beyond. Magn Reson Med. 2011;65(6):
Biomed. 2002;15(7–8):435–55. 1532–56.
4. Basser PJ, Pierpaoli C. Microstructural and physio- 11. Soares JM, Marques P, Alves V, Sousa N. A hitch-
logical features of tissues elucidated by quantitative- hiker’s guide to diffusion tensor imaging. Front
diffusion-tensor MRI. J Magn Reson B. 1996;111(3): Neurosci. 2013;7:31.
209–19. 12. Basser PJ, Pierpaoli C. A simplified method to mea-
5. Jones DK, Knösche TR, Turner R. White matter sure the diffusion tensor from seven MR images.
integrity, fiber count, and other fallacies: the do’s and Magn Reson Med. 1998;39(6):928–34.
don’ts of diffusion MRI. Neuroimage. 2012;73: 13. Jones DK. The effect of gradient sampling schemes on
239–54. measures derived from diffusion tensor MRI: a Monte
6. Jones DK, Cercignani M. Twenty-five pitfalls in the Carlo study. Magn Reson Med. 2004;51(4):807–15.
analysis of diffusion MRI data. NMR Biomed. 14. Mukherjee P, Chung SW, Berman JI, Hess CP, Henry
2010;23(7):803–20. RG. Diffusion tensor MR imaging and fiber
7. Mukherjee P, Berman JI, Chung SW, Hess CP, Henry tractography: technical considerations. AJNR Am
RG. Diffusion tensor MR imaging and fiber tractogra- J Neuroradiol. 2008;29(5):843–52.
phy: theoretic underpinnings. AJNR Am J Neuroradiol. 15. Veraart J, Sijbers J, Sunaert S, Leemans A, Jeurissen
2008;29(4):632–41. B. Weighted linear least squares estimation of diffu-
8. Mori S, Zhang J. Principles of diffusion tensor imag- sion MRI parameters: strengths, limitations, and pit-
ing and its applications to basic neuroscience research. falls. Neuroimage. 2013;81:335–46.
Neuron. 2006;51(5):527–39. 16. Le Bihan D, Poupon C, Amadon A, Lethimonnier
9. Jbabdi S, Johansen-Berg H. Tractography: where do F. Artifacts and pitfalls in diffusion MRI. J Magn
we go from here? Brain Connect. 2011;1(3):169–83. Reson Imaging. 2006;24(3):478–88.
Concepts of Diffusion in MRI
3
Matthew Rowe, Bernard Siow,
Daniel C. Alexander, Uran Ferizi,
and Simon Richardson

• MRI scans can be made sensitive to the


Learning Points displacement of water molecules at millisec-
ond time scales via the use of modulated mag-
• Water molecules in biological tissue are in netic gradients using the gradient coils present
continuous motion. They collide with each in standard MR scanners.
other and with local molecules and structures • The pulsed gradient spin echo (PGSE)
such as cell membranes. Three separate types sequence combined with EPI readout is the
of diffusion are: free diffusion in free water most commonly used sequence for diffusion
and hindered and restricted diffusion in the MRI. It consists of an excitation pulse and a
presence of boundaries such as cell refocusing pulse, sandwiched between two
membranes. equal magnetic gradients which affect the
• Tissue structure hinders and restricts diffusion phase coherence of the spins and cause signal
of water; different tissue structures have dif- attenuation in the presence of diffusing
ferent effects on the diffusion profile of the molecules.
water.
• White matter has a highly coherent fibrous
structure, which encourages anisotropic diffu- What Is Diffusion?
sion of water; this effect can be exploited to
estimate the orientation of white matter fibers In 1826 a botanist named Robert Brown was
using DTI. Grey matter has a less coherent studying the seemingly random pattern of motion
structure, water diffuses more isotropically in that pollen grains exhibited when suspended in
grey matter, and the overall effect on diffusion water through his microscope [1]. Initially puz-
properties is much more complicated and zled, he attributed it to some biological phenom-
harder to interpret. enon of the pollen, but when he later observed the
same behavior with inanimate, inorganic sub-
stances, he rejected this hypothesis. It later
became clear that the motion that he had observed
M. Rowe, PhD (*) • B. Siow, PhD • D.C. Alexander, PhD was due to the buffeting of the pollen grains by
U. Ferizi, PhD • S. Richardson, PhD water molecules surrounding them. This led to
Microstructure Imaging Group, Centre for Medical
Image Computing, University College London
the revelation that liquids and gases were not
(UCL), Gower Street, London WC1E 6BT, UK static and lifeless as they might appear at first
e-mail: matthew.rowe.09@ucl.ac.uk glance. The atoms and molecules from which

© Springer Science+Business Media New York 2016 23


W. Van Hecke et al. (eds.), Diffusion Tensor Imaging, DOI 10.1007/978-1-4939-3118-7_3
24 M. Rowe et al.

they are constituent are in constant motion, molecule. The dynamics of this process are too
undergoing persistent collisions and energy complex to predict exactly, so from a practical
exchanges with other molecules and atoms. This perspective the direction of each molecule
led to an intuitive understanding of phenomena changes effectively at random. This process of
observed in everyday life, such as the spreading translation and randomly changing direction is
of an ink drop in a glass of seemingly motionless often referred to as a “random walk.” In the fol-
water or the propagation of a perfume scent lowing, we will refer only to the diffusion of
through still air. The physical and mathematical water molecules, as this is of importance in diffu-
theories of diffusion were developed and refined sion tensor imaging (DTI). However, the basic
over the next two centuries by prominent scien- principles discussed apply to all gases and
tists including Thomas Graham, Adolf Fick [2, 3], liquids.
and eventually Albert Einstein [4, 5], who devel- Consider the diagrams in Fig. 3.1. Let us imag-
oped a rigorous mathematical framework, which ine that the blue dots represent water molecules in
is still in use today. an open body of water such as in a glass of water.
The principles of diffusion and its importance In the first diagram, we see water molecules, all of
in biological tissue can be conceptualized simply, which are in motion and randomly colliding with
without a need to consider the mathematical basis each other, performing a random walk. The paths
of diffusion theory. Water molecules at room taken by three randomly selected molecules are
temperature, or at body temperature, are in con- outlined by the red trails in Fig. 3.1a. If we could
stant motion due to their inherent thermal energy. label a small cluster of the water molecules such
In the absence of any obstacles, a water molecule as those highlighted in red in Fig. 3.1b and watch
would continue to move in the same direction their progress over a short time, we would see the
forever in accordance with Newton’s first law of labeled molecules spread out evenly in each direc-
motion. However, in the presence of many other tion as in the latter diagrams 3.1c, d after times
water molecules, each and every molecule under- T and 2T respectively. In Fig. 3.1d it can also be
goes many collisions in a short space of time, seen that the labeled molecules are clustered
each of which change the direction of the water around the point of origin of the labeled set.

Fig. 3.1 Illustration of the


free diffusion of water
molecules in pure water
over time interval 2T. The
random walks of a
selection of molecules are
illustrated with red lines.
The progression of a small
group of molecules
highlighted in red is shown
in (b–d)
3  Concepts of Diffusion in MRI 25

This kind of diffusion is normally referred to as Hindered/Restricted/Free Diffusion


“free diffusion” as the water molecules are free to
move in any direction with only the other water We have already covered the concept of “free dif-
molecules obstructing them. Free diffusion leads fusion,” the diffusion of water molecules unim-
to a zero-mean Gaussian distribution of particle peded by the presence of any boundaries.
displacements. This means it is more likely for a However, in the presence of boundaries, the dif-
molecule to travel a shorter distance, than a longer fusion properties of water are different. There are
distance, and in fact the most likely distance to two more types of diffusion, which commonly
move is zero. It is important to note that a zero occur in biological tissue: restricted diffusion and
displacement does not imply that the molecule hindered diffusion.
has not moved at all during a particular time
period; it has simply back-tracked in its random
walk and is in a similar position at the start and Restricted Diffusion
end of the time period we are observing.
If we were to view this process in three Restricted diffusion occurs when water mole-
dimensions, we would observe that a labeled cules are constrained by impermeable boundar-
cluster of water molecules would spread out in a ies, which impedes the progress of water
roughly spherical profile. In free diffusion the molecules beyond a certain maximum displace-
progress of molecules is unobstructed by obsta- ment in a particular direction. In the context of
cles and is therefore independent of direction. biological tissue, this kind of diffusion occurs for
Diffusion that is independent of direction also water molecules trapped inside a cell boundary,
has a special term: “isotropic diffusion.” Things otherwise known as the “intracellular” region.
get more interesting when we consider the effect The motion of water molecules within this region
of the presence of boundaries on the propagation is restricted to within a certain range unless they
of water. If diffusing water molecules encounter can permeate the cell membrane. In this regime,
boundaries, such as cell membranes in nervous the diffusion properties of water are largely gov-
tissue, the presence of such boundaries will erned by the geometric properties of the con-
cause a departure from the isotropic, free diffu- straining membrane.
sion we observe in a glass of water. DTI gives us
a powerful tool to examine this departure from
free diffusion properties, which can give us Hindered Diffusion
insight into the geometric properties of the
boundaries and allow us to infer details about Hindered diffusion occurs when the progress of
tissue structure. water molecules as they diffuse is impeded by
In water at room temperature, the average obstacles, but not completely confined by a con-
distance moved of a water molecule in one sec- tinuous boundary as in the case of restricted dif-
ond is around 100 μm, or 1/10th of a millimeter. fusion. Hence the mean displacement of a set of
In 50 ms, which is a time of the order of magni- water molecules is reduced overall in any direc-
tude relevant to DTI sequences, the average dis- tion in which boundaries are encountered. The
placement of a water molecule is about 25 μm. net result is that the average displacement of
This length scale is of a similar magnitude to water molecules is reduced in directions, which
that of many cellular structures of central ner- impinge on boundaries. In biological tissue, this
vous system tissue. Hence if we probe the type of diffusion is exhibited by water present in
motion of water molecules at such timescales the interstitial space between cells, otherwise
with diffusion tensor imaging, we can probe the known as the “extracellular” region.
geometric structure of the tissue of the central In Fig. 3.2, we can see the difference between
nervous system at the cellular level from outside molecules exhibiting restricted diffusion and
the body. molecules exhibiting hindered diffusion. The red
26 M. Rowe et al.

Fig. 3.2 Illustration of


hindered and restricted
water inside and around
boundaries, such as cell
membranes. The red dots
represent molecules which
are restricted by the
presence of the boundaries
while the blue dots
represent molecules which
are hindered by the
presence of the boundaries

molecules inside the boundaries are restricted drical boundaries become highly dependent on
and cannot exceed a certain maximum displace- the orientation of the cylindrical boundaries.
ment as they are restricted from doing so by the Water diffuses a lot more readily along the axis of
cell boundary. The motion of a single water mol- a set of packed cylinders than perpendicular to it.
ecule is exemplified by the red path. The blue This is a phenomenon that becomes highly
molecules, however, can diffuse in any direction. important in DTI when studying neural tissue, as
Their progress is impeded, but not restricted, by we will see when we examine the cellular struc-
the presence of the boundaries. A typical path of ture of tissue in the next section. There is a spe-
a “hindered” water molecule is exemplified by cial term for this kind of directionally dependent
the dark blue path. Given enough time, these diffusion: “anisotropic diffusion.” The terms
“hindered” molecules could reach any distance; “anisotropic” and “isotropic” are regularly used
however, their progress is impeded by the pres- in DTI terminology, so it’s worth taking note of
ence of the boundaries. Therefore we see a depar- what they mean.
ture from free diffusion properties, which is
dependant on the geometry of the boundaries.
Of particular interest in DTI is the effect of Microstructural Tissue Properties
specifically shaped boundaries. It is easy to see at the Scale of Diffusion MR
from the previously mentioned examples and Measurements
illustrations how water molecules would behave
in and around a spherical boundary, but it is also In biological tissue, such as that of the central
interesting to consider how a diffusing water nervous system, the components of the cells that
molecule would behave inside a cylindrical make up the tissue create boundaries to diffu-
boundary as in Fig. 3.3. The cylindrical boundary sion. The various properties of cellular structures
presents an obstacle to diffusion in the direction such as the cell membrane, the viscosity of the
perpendicular to its axis, while presenting no material within the cell, and the presence of sub-
obstacle to diffusion along its axis. Therefore the cellular structures such as mitochondria, nuclei,
diffusion properties of water in and around cylin- and microfibrils have an effect on the diffusion
3  Concepts of Diffusion in MRI 27

Fig. 3.3 Illustration of


water molecules restricted
inside a cylindrical
boundary. The red lines
represent the paths taken
by a small selection of
molecules trapped inside
the cylinders

properties of water in and around the cell and this and there is only one axonal protrusion of each
has an impact on the measurements we take in neuron. The longest axons in the bodies of mam-
DTI. In the following, we will examine the cellu- mals, such as the sciatic nerve in humans, can be
lar composition of central nervous system tissue. many centimeters or even over a meter in length.
The fundamental building blocks of the brain’s They can vary in diameter between around 0.2
processing network are neurons. Neurons are and 20 μm in the brain [7].
electrically excitable cells and are typically Many axons throughout the central nervous
divided into two major parts, the soma or “body” system are covered by a fatty layer known as the
of the neuron and the neuritic protrusions: den- myelin sheath. The myelin sheath exhibits peri-
drites and an axon [6]. odic gaps called Nodes of Ranvier, which are
The main cellular structures are illustrated in extremely important for the transmittance of
Fig. 3.4. The body of the neuron, the soma, varies electrical excitation along the axon. Charge
in size and shape throughout the central nervous builds up at each node and hops to the next one in
system, typical sizes range from 5 to 100 μm. The rapid succession, facilitating fast, efficient com-
soma contains the cell nucleus and other cell munication of electrical excitation or “action
structures such as Nissl bodies, mitochondria, potentials” along the axon. The myelin sheath
and microfibrils. serves to both facilitate the propagation of action
The soma has numerous protrusions collec- potentials and insulate each axon from interfer-
tively termed “neurites.” Neurites fall into two ence from action potentials traveling in other
types: dendrites and axons. Dendrites are small, axons and electrically charged ions in the sur-
short, branched projections which taper towards roundings. The myelin sheath is composed of
the end. They facilitate short range exchange of several layers, which are formed by the winding
electrical and chemical signals between neigh- of the protrusion of an oligodendrocyte cell
boring neurons and axons. Most neurons typi- around the axon. Each oligodendrocyte can serve
cally also have a single axon. The axon is an many axons. The myelin, made of a multilayered
elongated protrusion of the nucleus which facili- wrapping of a protrusion of an oligodendrocyte
tates long range connection with other neurons cell, is illustrated in Fig. 3.5.
28 M. Rowe et al.

Fig. 3.4 Simple
illustration of cellular
components of neural
tissue including the soma,
axons, and glia

There are other types of cells that exist around


neurons. The aforementioned oligodendrocyte
cells are numerous and serve to provide the insu-
lating myelin sheath to axons. Microglia are small
cells which vary widely in size and shape and
serve as the immune defence for the central ner-
vous system. They act as macrophages do else-
where in the body, i.e., identifying and clearing
plaques, damaged neurons, and infectious agents.
Astrocytes are star-shaped cells that serve a num-
ber of maintenance and metabolic functions. They
are the most abundant cell type in the brain.
This brings us to an important distinction to
make between the two major tissue types in the
brain: grey matter and white matter. Figure 3.6 is
Fig. 3.5 Illustration of the multilayered myelin sheath a highly simplified illustration of the contrast
formed from a protrusion of an oligodendrocyte between grey matter and white matter structure.
3  Concepts of Diffusion in MRI 29

Fig. 3.6 Simple
illustration of the contrast
between grey matter and
white matter

Grey matter is the tissue which can be sands or millions, which are coherent and tightly
observed on the surface of the brain; this region is packed. Hence diffusion characteristics of water
often referred to as the “cerebral cortex.” The in white matter are often highly directionally
cerebral cortex is the outermost layer of neural dependent. The intracellular water tends to dif-
tissue and is usually 2–4 mm thick in humans. fuse much more readily along the axis of the
The cerebral cortex is divided up into different axons in a coherent bundle than in any other
areas which are active in cognition, motor, and direction and the extracellular water is also heav-
sensory responses. There are also some deep ily guided by the lattice structure formed by the
“subcortical” regions of grey matter, which are axons. Intracellular water tends to exhibit
located in and around the brainstem and serve restricted diffusion properties, while extracellu-
many basic purposes, such as cardiac and respira- lar water tends to exhibit hindered diffusion
tory regulation, sleep and appetite regulation, and properties, both of which were discussed in the
also as relays for connection between multiple previous section. Therefore probing the diffusion
cortex regions and the rest of the body. The grey characteristics of water in white matter can yield
matter mainly consists of the soma, the dendrites, information on the direction of bundles of axons
and other cells such as microglia and astrocytes. as they traverse the brain. This is of critical
The organization of grey matter tissue is importance to DTI as it provides us a very power-
highly complex, with many neuron bodies and a ful tool to probe the connectivity of the brain.
complex web of dendrites and glial cells. The dif- There are many other structures and sub-
fusion characteristics of water in grey matter are stances that affect the diffusion of water in and
consequently highly complicated, due to com- around cells in the central nervous system such as
plex structural trends in the tissue. As a result of cytoplasm, neurofilaments, mitochondria, the
this interpreting DTI measurements in grey mat- extracellular matrix, and blood vessels. However
ter is very challenging. the effect of these biological elements on DTI is
White matter is the tissue that lies underneath often negligible or too complicated to be effec-
the cortex and serves to connect neurons in tively modelled, and is hence beyond the scope of
different parts of the brain to form the neural this book.
network. It is composed almost entirely of Figure 3.7 shows an electron micrograph of
myelinated axons. The name “white matter” the optic nerve of a healthy rat sliced in a plane
comes from the fact that myelin is a white sub- perpendicular to the axis of the nerve fibers. The
stance; therefore white matter appears white optic nerve is a white matter structure comprised
when observed in dissection. The axons in white largely of myelinated axons. Many of the cellular
matter are organized into tight bundles of thou- structures discussed in this section can be
30 M. Rowe et al.

Fig. 3.7 Electron


micrograph of the optic
nerve of a healthy rat sliced
in a plane perpendicular to
the axis of the nerve fibers

observed in this picture. The filled red arrow indi-


cates the interior of an axon and the open arrow  asic Concepts: How to Diffusion
B
points out the compact myelin layer surrounding Weight the MRI Signal
axons. N indicates the nucleus of an oligodendro-
cyte; C indicates the cytoplasm of the same oligo-  he Pulsed Gradient Spin Echo
T
dendrocyte. M shows the nucleus of a microglial Sequence
cell. + indicates part of an astrocyte, * shows
hemochromatin, and the filled triangle shows the Currently, the Pulsed Gradient Spin Echo (PGSE)
Gogli apparatus of the oligodendrocyte. method is the most common way of sensitizing
the MRI signal to molecular diffusion. PGSE
sequences comprise a spin echo with magnetic
 ensitizing MR Measurements
S field gradients before and after the refocusing
to Diffusion pulse followed by an acquisition module
(Fig.  3.8). The effects of diffusion on spin echo
Magnetic resonance imaging relies on the inter- NMR experiments in a constant magnetic field
action of the applied magnetic fields with nuclei gradient were first formulated by Hahn [8] and
in the sample. “Spin” is a quantum mechanical extended by Carr and Purcell [9]. Stejskal and
property of a nucleus and over a large number of Tanner [10] described the PGSE sequence in
nuclei it gives rise to a net magnetization once which the magnetic field gradients are “pulsed”
placed in a magnetic field. The net magnetization instead of being constant over time. They also
of spins precesses with a frequency proportional derived the signal equation for the sequence in
to the local strength of the magnetic field. This is freely diffusing liquids (Eq. 3.1). Such is the
known as Larmor precession and it is by manipu- impact of their paper that the terms “Stejskal-
lating this relationship that we can acquire a mag- Tanner sequence” and “Stejskal-Tanner equation”
netic resonance image and also apply diffusion are still commonly used. The use of pulsed gradi-
weighting. For biological tissue, the majority of ents for diffusion weighting allows more control
the signal arises from the hydrogen nuclei in over how the signal is weighted by diffusion.
water molecules. Furthermore, it allows the diffusion weighting
3  Concepts of Diffusion in MRI 31

Fig. 3.8 Schematic of a PGSE pulse sequence

gradients to be separate from other pulsed gradi- (d) > (e) The second pulsed gradient rephases
ents to be used for spatial localization, thus allow- magnetization:
ing for the quantification of diffusion in MRI
experiments. • No diffusion: Magnetization is completely
rephased since the field experienced by the
molecules has not changed.
How the PGSE Signal Is Generated • Free diffusion: (i) Molecules have changed
position between the start of the first and sec-
Let us consider the evolution of net magnetiza- ond gradients (diffusion time) (ii) The field
tion during the PGSE sequence, ignoring T1 and that they experience has changed. (iii) The fre-
T2 relaxation, and assuming the pulsed gradients quency of precession has changed. (iv) The
are short. We will need to know one key concept: phase of the magnetization after the second
the frequency of precession of the net magnetiza- gradient will not be the same as before the first
tion is proportional to the field that the molecules gradient. (v) In free diffusion, the motion of
experience (i.e., the Larmor equation, f = γB, molecules is incoherent and the magnetization
where f is the frequency in Hertz, γ is the gyro- will have a distribution of phases. (vi) The net
magnetic ratio in Hertz per Tesla, and B is the magnetization is attenuated.
applied field in Tesla). Thus, when a magnetic (e) The signal is acquired: In the presence of
field gradient is applied across a sample, the net diffusion, the signal is attenuated.
magnetization will precess at different frequen-
cies across that gradient (Fig. 3.9).
Figure 3.10 shows the evolution of magnetiza- b
 -Value: A Handy Way to Quantify
tion during the PGSE sequence in the rotating Diffusion Weighting
frame. Also see Fig. 3.8 for corresponding points
along the pulse sequence. The “b-value” [11] quantifies the applied diffu-
(a) > (b) The excitation pulse rotates net mag- sion weighting of a pulse sequence in a single
netization onto the transverse plane. number. This single number has become the stan-
(b) > (c) The first pulsed gradient dephases the dard metric of quantifying diffusion weighting in
spin magnetization due to the variation of the fre- MRI sequences. A low b-value scan has more
quency of precession along the gradient. signal compared to a high b-value one (see
(c) > (d) The refocusing pulse rotates the mag- Fig. 3.11), or, in other words, a high b-value scan
netization about the y-axis. exhibits more signal attenuation in the presence
32 M. Rowe et al.

Fig. 3.9 Frequency and phase relationship of Larmor precession in the presence of a magnetic field gradient

of diffusion than a low b-value scan. So the normally include one or more zero b-value scans
b-value could be said to be a rough measure of for normalization. These scans are often referred
how much the signal in an image will be affected to as b-zero images.
by the diffusion of water in tissue. A b-value of Informally, the b-value is often given in units
zero will give no diffusion weighting at all in of s/mm2. For example, “b-value of 1000” is an
the image and diffusion weighted acquisitions often heard phrase in the clinic—roughly, this
3  Concepts of Diffusion in MRI 33

Fig. 3.10 The evolution of


magnetization during the
PGSE sequence

will normally give approximately 40–60 % sig- nal; hence this lowers the signal to noise ratio
nal attenuation in the brain, depending on the tis- (SNR). Therefore when choosing the b-value for
sue type in which the water is embedded. diffusion weighted scans, it is important to con-
Elsewhere in the body, b-values used in clinical sider the compromise between SNR and contrast.
investigations are often lower from 50 s/mm2 and High b-value scans can offer extra information
below to 500 s/mm2 [12, 13]. Selection of b-value about tissue structure, especially structure at very
is influenced by the intended application and a small length scales. However, we must keep in
balance between practical concerns and hardware mind that the SNR will be lower when planning
limitations [14]. It is worth noting that at higher an acquisition and processing image data. More
b-­values, as there is more signal attenuation in advanced techniques often use considerably
the presence of diffusion, there is less overall sig- higher b-values of 2000 or above [15–17].
34 M. Rowe et al.

The b-value summarizes the three parameters


G, δ, Δ used to control the amount of diffusion
weighting in a PGSE sequence. It should be noted
that the same b-value can produce very different
contrast depending upon the combination of G, δ,
Δ, the selection of which can be optimized for
particular applications and tissue types [19].

 ree, Hindered, and Restricted


F
Diffusion

Thus far we have discussed the diffusion MRI


Fig. 3.11 Diffusion weighted MRI signal as a function of signal in the case of free diffusion, i.e., molecules
b-value
are free to diffuse as in a large pool of liquid
(Fig. 3.1). Let us consider again a population of
diffusing molecules, if at time t = 0 they are all at
PGSE Signal the same point, then at t > 0 their spatial distribu-
tion is Gaussian (or in shorthand a “Gaussian dis-
The signal of a PGSE scan of a freely diffusing persion pattern” or just “Gaussian diffusion”).
sample is given by the Stejskal-Tanner equation: The average distance between the position of
d molecules at the start and end of the diffusion
S - ( g 2 G 2d 2 ( D - )) D
=e 3(3.1) time (or more technically, the root mean square
S0 (RMS) displacement) is given by:
which can be written in terms of the b-value as
RMS = 6Dt (3.3)
such:
For biomedical applications there are only a few
S
= e - bD specific locations in which free diffusion
S0
occurs—for example in cerebrospinal fluid in
where large cavities in the ventricular system. In other
d (3.2) tissues, diffusion is hindered and restricted by
b = g 2G 2d 2 (D - )
3 intra- and extra-cellular constituents and by the
where δ is the gradient duration, Δ is the time cellular membranes (Fig. 3.2). In these cases, the
between the start of each gradient, G is the gradi- diffusion constant quantified using the Stejskal-
ent strength, γ is the gyromagnetic ratio, S is the Tanner equation is not the innate diffusion con-
signal, S0 is the signal without diffusion weight- stant of the tissue water but rather an “apparent
ing gradients, and D is the diffusion constant. diffusion coefficient” (ADC)—equivalent to the
The PGSE signal decays exponentially as a self-diffusion constant of a more viscous, freely
function of the parameters of the applied gradi- diffusing liquid.
ents and the diffusion constant (see Fig. 3.11). In In tissue water that is not confined within the
imaging experiments, additional pulse magnetic microstructure of the tissue, for example in the
field gradients are applied for spatial localization. extracellular space, the RMS displacement is
These gradients, and other applied magnetic field approximated quite well by substituting D with
gradients, may contribute to diffusion weighting. ADC in Eq. (3.3). The relationship between ADC
These are often partially factored out by dividing and RMS displacement is less straightforward in
signal from scans with diffusion weighting gradi- the case of molecules that are confined within the
ents by that of scan with no or low diffusion tissue microstructure (or more accurately, where
weighting gradients, the latter being used to min- water molecules have a low probability of leaving
imize the effects from perfusion [18]. the bounding structure during the diffusion time
3  Concepts of Diffusion in MRI 35

such as water inside myelinated axons). For short 11. Le Bihan D, Breton E. Imagerie De Diffusion In Vivo
Par Résonance Magnétique Nucléaire. Comptes
diffusion times, RMS diffusion is well approxi-
Rendus De Académie Des Sciences De Paris.
mated by ADC. At long diffusion times the mole- 1985;301:1109–12.
cules will have had enough time to traverse across 12. Koh DM, Collins DJ. Diffusion-weighted MRI in the
the structure and “bounce off” the bounding wall. body: applications and challenges in oncology. AJR
Am J Roentgenol. 2007;188(6):1622–35.
In this case, RMS displacement is not well
13. Lansdown DA. Quantitative diffusion tensor MRI-
approximated by Eq. (3.3). On the scale of an based fibre tracking of human skeletal muscle. J Appl
MRI voxel, tissue contains a mixture of hindered Physiol. 2007;103(2):673–81.
and restricted “compartments”; thus quantifica- 14. Le Bihan D, Poupon C, Amadon A, Lethimonnier
F. Artefacts and pitfalls in diffusion MRI. J Magn
tion of indices of microstructure based solely on
Reson. 2006;24(3):478–88.
Eq. (3.3) should be used in an informed manner. 15. Jensen JH, Helpern JA, Ramani A, Lu H, Kaczynski
K. Diffusional kurtosis imaging: the quantification of
non-gaussian water diffusion by means of magnetic
resonance imaging. Magn Reson Med. 2005;53(6):
References 1432–40.
16. Tournier J, Calamante F, Connelly A. Determination
1. Brown R. A brief account of microscopical observa- of the appropriate b value and number of gradient
tions made in the months of June, July and August, directions for high-angular-resolution diffusion-
1827, on the particles contained in the pollen of weighted imaging. NMR Biomed. 2013;2013:
plants: and on the general existence of active mole- 1099–492.
cules in inorganic bodies. Phil Mag. 1827;4:161–73. 17. Alexander DC, Hubbard PL, Hall MG, Moore EA,
2. Fick A. Concerns diffusion and concentration gradi- Ptito M, Parker GJ, et al. Orientationally invariant
ent. Ann Phys Lpz. 1855;170:59. indices of axon diameter and density from diffusion
3. Fick A. Über diffusion. Ann Phys. 1855;94:59. MRI. Neuroimage. 2010;52(4):1374–89.
4. Einstein A. Über die von der molekularkinetischen 18. Le Bihan D, Breton E, Lallemand D, Aubin ML,
Theorie der wärme gefordete Bewegung von in Vignaud J, Laval-Jeantet M. Separation of diffusion
ruhenden Flüsigkeiten suspendierten Teilchen. Ann and perfusion in intravoxel incoherent motion MR
Physik. 1905;4:549–60. imaging. Radiology. 1988;168(2):497–505.
5. Einstein A. Investigations on the theory of Brownian 19. Alexander DC. A general framework for experiment
movement. New York, NY: Dover Publications, Inc; design in diffusion MRI and its application in measur-
1926. ing direct tissue-microstructure features. Magn Reson
6. Levitan IB, Kaczmarek LK. The neuron: cell and Med. 2008;60:439–48.
molecular biology. 3rd ed. New York, NY: Oxford
University Press; 2002.
7. Waxman SG, editor. The axon: structure, function and
pathophysiology. New York, NY: Oxford University Suggested Reading
Press; 1995.
8. Hahn EL. Spin echoes. Phys Rev. 1950;80(4):580–94. Crank J. Mathematics of diffusion. Oxford: Clarendon;
doi:10.1103/PhysRev.80.580. 1975.
9. Carr HY, Purcell EM. Effects of diffusion on free pre- Basser PJ, Mattiello J, Le Bihan D. Estimation of the
cession in nuclear magnetic resonance experiments. effective self-diffusion tensor from the NMR spin
Phys Rev. 1954;94(3):630–8. doi:10.1103/PhysRev. echo. J Magn Reson B. 1994;103(3):247–54.
94.630. doi:10.1006/jmrb.1994.1037.
10. Stejskal EO, Tanner JE. Spin diffusion measurements: Le Bihan D, Johansen-Berg H. Diffusion MRI at 25:
spin echoes in the presence of a time‐dependent exploring brain tissue structure and function.
field gradient. J Chem Phys. 1965;42(1):288–92. Neuroimage. 2012;61(2):324–41. doi:10.1016/j.
doi:10.1063/1.1695690. neuroimage.2011.11.006.
From Diffusion to the Diffusion
Tensor 4
Thijs Dhollander

• Eigendecomposition of the tensor; the mean-


Learning Points ing and interpretation of eigenvalues and
eigenvectors; and interpreting maps of eigen-
• The diffusion coefficient; the apparent diffu- values and directionally encoded color (DEC)
sion coefficient (ADC); their relation to the maps of eigenvectors.
pair of acquired diffusion-weighted images • Basic visualization of tensors by glyphs; the
(DWI) and nondiffusion-weighted images meaning of the tensor ellipsoid; and maps of
(B0); and a few general properties of the ADC common measures such as mean diffusivity
in relation to the amount of diffusion weight- (MD), fractional anisotropy (FA), DEC FA,
ing (the b-value). and a few other shape measures.
• The concept of directionality of the diffusion • The issue of tensor fitting; and specific tensor
sensitizing gradient; the concept of anisotropy fitting methods: linear least squares (LLS),
in the acquired data; and inferring useful weighted linear least squares (WLLS), nonlin-
information on the orientation of axon bundles ear least squares (NLS), and robust estimation
based on raw DWIs, normalized DWIs, ADC of tensors by outlier rejection (RESTORE).
maps, and spherical polar plots of the latter
two quantities.
• The apparent diffusion tensor; evaluating a The (Self-)Diffusion Coefficient
tensor model; the link between the diffusion
tensor and the acquired DWIs/B0; the mean- Measuring (Self-)Diffusion
ing and interpretation of the tensor elements; in the MR Scanner
and visualizing the tensors by spherical polar
plots (ADC peanuts). Sit down, relax, and grab yourself a glass of
water. Now put it in a nearby MR scanner (or
rather, imagine doing this). Acquire a (nondiffu-
sion-weighted) T2-weighted image as well as
T. Dhollander, PhD (*) one of those fancy new diffusion-weighted
The Florey Institute of Neuroscience and Mental
images. Now let’s see if we can recover the diffu-
Health, Melbourne Brain Centre, 245 Burgundy
Street, Heidelberg, VIC 3084, Australia sion coefficient of water from these two images.
More accurately, we’re talking about the self
Medical Imaging Research Center (MIRC),
KU Leuven, Leuven, Belgium ­diffusion coefficient here: it quantifies the freedom
e-mail: thijs.dhollander@florey.edu.au of movement of any single molecule of water, in

© Springer Science+Business Media New York 2016 37


W. Van Hecke et al. (eds.), Diffusion Tensor Imaging, DOI 10.1007/978-1-4939-3118-7_4
38 T. Dhollander

the glass of water. We’ll simply refer to it as D. The value of D is typically reported in mm2/s. For
Also note that we need (at least) two images: the our glass of water, D = 2.2 × 10−3 mm2/s should be
diffusion-­weighted image would appear exactly realistic at room temperature. If we were lying in
the same as the T2-weighted image, if it were not the scanner ourselves and performed the above
weighed down by the appearance of diffusion; i.e., calculation for a voxel of cerebrospinal fluid
we’re interested in the relative difference between (CSF) in the ventricles of our brain, a value of
both images. Since D should be the same in the about 3.1 × 10−3  mm2/s is to be expected. While
entire glass of water, we simply choose one voxel. CSF consists mostly (99 %) of water, the differ-
The intensity of the diffusion-­weighted image in ence can be explained by our body temperature,
this voxel will be referred to as S, while the (non- which is of course higher than the normal room
diffusion-weighted) T2-weighted image’s inten- temperature. One point is enough to fully fix the
sity equals S0. As explained in Chap. 3, the process slope of the line in (Fig. 4.1) and per consequence
of diffusion should have caused attenuation in S, also determine D. If we would have performed
so S should always be smaller than S0. The decay the measurement using a different b-value, we
of S relative to S0 is given by the so-called would obtain another point on this exact same
Stejskal-Tanner equation [1]: line. Using a higher b-value would result in more
decay, and thus a lower value for S (this can be
S = S0 •e - b•D (4.1)
most easily appreciated by looking at Eq. 4.1). A
The b-factor in this equation captures all the rel- lower value for S means a higher value for −
evant scanning parameters and was introduced ln(S/S0). Consequently, we are simply consider-
to take abstraction of them [2]. In general, it can ing a point further up the same line in (Fig. 4.1).
be seen as the amount of diffusion weighting
that is applied; i.e. how sensitive the acquisition
is to diffusion. Its value is typically set and Conclusions
reported in s/mm2. As a realistic value for our
simple experiment at hand, we could have cho- We are now able to calculate the self-diffusion
sen e.g. 800 s/mm2. We can rewrite this equation coefficient D of free water (be it in a glass or as
so it becomes CSF in the ventricles), using measurements from
S
- ln( ) = b•D (4.2)
S0

The left side of the equation only contains mea-
surements we obtained from the scanner (S and
S0), while the right side contains the scanning
parameters in function of which we did so (all
contained within the b-factor) and a constant (D
for our glass of water at room temperature). From
this we learn that a logarithmic transform (i.e., “−
ln(…)”) of our normalized measurement (i.e.,
“S/S0”) depends linearly on the applied diffusion
weighting (i.e., b). As they are both simply related
by a factor D, plotting − ln(S/S0) in function of b
yields a straight line through the origin, as shown Fig. 4.1  In case of free diffusion (e.g., in a glass of water,
in (Fig. 4.1). The slope of this line equals D: or CSF in the ventricles of the brain), the plot of − ln(S/S0)
in function of b-value is a straight line through the origin.
S
- ln( ) One point (grey short dashed lines) is enough to fully fix
S0 this line. It requires two images (S and S0) as well as
D= (4.3) knowledge of the b-value used to acquire S. The slope of
b the resulting line equals the self-diffusion coefficient D
4  From Diffusion to the Diffusion Tensor 39

a MR scanner and the Stejskal-Tanner equation.


The minimum requirements are a nondiffusion-­
weighted image (S0), a diffusion-weighted image
(S) and knowledge of the b-value that was used
for performing the acquisition of the diffusion-­
weighted image.

The Apparent Diffusion Coefficient

Apparent Complications

Feeling confident about the newly gained ability Fig. 4.2  In case of hindered/restricted diffusion (in tis-
to obtain D from the two images we acquired of sue, e.g. the grey matter of the brain), the plot of − ln(S/S0)
in function of b-value is a curve through the origin. Based
our brain, we also attempt to perform the same on one point (grey short dashed lines), we can calculate an
calculation in a voxel of gray matter. Suddenly, apparent diffusion coefficient (ADC). Just like D, it equals
however, we are confronted with a resulting the slope of the line that connects this point to the origin
value of about 0.9 × 10−3 mm2/s. Apparently, the (black long dashed line)
self-­diffusion coefficient of water has changed,
just because we measured it in the gray matter. couple of diffusion-sensitizing gradients. By
Maybe something went wrong with the scan? We ­taking abstraction of any complicated MR phys-
perform the acquisition again for a couple of dif- ics, we could say the MR scanner actually per-
ferent b-values. Carefully dotting out the obtained forms a simple experiment in each voxel: it takes
values of − ln(S/S0) in function of b and connect- a snapshot of all the water molecules, waits a bit,
ing everything loosely by hand, we obtain a curve and then takes another snapshot. During the short
such as the one depicted in (Fig. 4.2). Apparently, waiting time, however, the molecules have the
the self-diffusion coefficient of water now even opportunity to diffuse a bit. Per consequence, a
changes in function of our chosen acquisition relative displacement of each molecule can take
parameters. Using Eq. (4.3) to calculate D place in between both snapshots. The expected
equates to connecting a certain measured point signal of the original (e.g., T2-weighted) sequence
on this curve with the origin by a straight line (as is attenuated in function of the amount of dis-
shown in Fig.  4.2) and assuming its slope still placement of all water molecules in the voxel (as
equals D. Since the obtained values are clearly well as the amount of applied diffusion weight-
lower than expected and they also seem to vary in ing). From the measurements of such an experi-
function of b, such a value is referred to as an ment (relative to a nondiffusion-weighted image),
apparent diffusion coefficient (ADC) [3]. It’s cal- the Stejskal-Tanner equation is able to reliably
culated from the measurements in exactly the calculate D… if and only if nothing disturbs the
same way as D: experiment. However, in tissue, such as gray mat-
ter, there are cell membranes all over the place.
S
- ln( ) Because the water molecules happen to bump into
S0
ADC = (4.4) the cells—i.e., they are hindered—they have a
b harder time to diffuse further away during the
To understand the behavior of the obtained ADC experiment. Water inside the cells may even be
in regions containing tissue (e.g., gray matter), we restricted to a confined space. And thus, our cal-
need to look into how the acquisition of a diffu- culation of D will apparently yield a lower out-
sion-weighted image works. An existing (e.g., come, which is why we call it the ADC instead.
T2-weighted) sequence is modified by adding a The time the experiment allows the molecules to
40 T. Dhollander

diffuse is one of the parameters that makes up the deceiving ADC instead. However, you have to
b-value. It’s easy to imagine that a larger diffusion look at it from the bright side: we now effectively
time will allow more molecules to hit some of have access to a probe that tells us something
these cell membranes. Hence, the effect of the about these cells that hinder/restrict diffusion.
hindered/restricted diffusion on our measurement That’s right: even though our voxel size might be
will increase with b-value; yet another reason to quite crude (2 × 2 × 2  mm3 or larger is not
refer to the outcome of our calculations using the unusual), the measured values are sensitive to
term “ADC”. This is also illustrated in (Fig. 4.3): differences in structure at a micrometer scale! We
using a larger b-value renders the measurement of are not interested in the ADC for the purpose of
S less sensitive to (truly) free diffusion, in favor of quantifying diffusion itself, but rather to investi-
hindered/restricted diffusion. As such, S will be gate properties of the tissue that apparently
less attenuated and the value of − ln(S/S0) will be caused the diffusion process to behave in the way
smaller than expected, yielding a downward cur- that we measure.
vature when plotting − ln(S/S0) in function of b. Before moving on, let’s investigate one more
This finally leads to an important property of the property of the ADC that teaches us something
ADC in tissue, as indicated in (Fig. 4.3): using a else about its capacity in distinguishing different
higher b-value results in a lower ADC. tissues. Consider the setting in (Fig. 4.4): it pres-
ents again − ln(S/S0) in function of b, but this time
for measurements at two different locations (e.g.,
Apparent Advantages in the brain). Looking at the plots and applying
what we just learned, we can safely say that the
At this point, you might start to wonder what the voxel at position pg contains more hindering/
point is of trying to find out D in voxels contain- restricting tissue than the voxel at position pf. The
ing tissue, only to end up having to deal with a former voxel’s plot shows greater curvature,

Fig. 4.4  The contrast of the ADC, e.g., between two dif-
Fig. 4.3  The ADC is dependent on the b-value used to ferent tissues at voxel positions pf and pg, is dependent on
acquire S. Due to the downwards curvature of the plot the b-value used to acquire S. Due to different tissue prop-
of − ln(S/S0) in function of b-value, a larger b-value results erties, both plots of − ln(S/S0) in function of b-value show
in a lower ADC. An explanation lies, e.g., in the fact that a different curvature. The tissue at pf imposes less hin-
increasing the diffusion time allows more molecules to drance/restriction on the diffusion as compared to the tis-
bump into cell membranes. This will on average decrease sue at pg and thus the accompanying curve is closer to a
their final displacement, resulting in a reduced amount of straight line. Hence, ADCf is larger than ADCg for a given
attenuation of S and finally leading to a lower value b-value. Increasing the b-value also results in an increase
for − ln(S/S0) than expected in a free (non-hindered/ of the relative difference between both ADC values, i.e.,
restricted) environment an increase of contrast
4  From Diffusion to the Diffusion Tensor 41

while the latter better approximates the straight reduced SNR. Due to these dependencies, inter-
line we would expect in case of free diffusion. preting/reporting the ADC only makes sense
Due to this difference in curvature of both plots, when the b-value is also specified. Finally, com-
the relative difference in magnitude of − ln(S/S0), paring ADC values or maps originating from
and thus also ADC, increases for larger b-values. acquisitions with different b-values does not
In other words, using a larger b-value results in a make a lot of sense.
better contrast when calculating an ADC map.
This fact of course begs the question why we
should still limit ourselves to a certain b-value. Gradient Directions and Anisotropy
That is, why not use an absurdly high b-value for
maximal contrast? The two most important fac- Anisotropic Complications
tors that generally contribute to the b-value are
the strength of the applied diffusion-sensitizing Up to now, we’ve been silently ignoring yet
gradients and the time that we allow the water to another important fact that will complicate every-
diffuse during the experiment. The former is lim- thing even more. It concerns that diffusion-sensi-
ited by what we can achieve with available hard- tizing gradient: it’s about time we started taking
ware. The latter is fully under our control. into account that it’s applied along a certain
Allowing a too long diffusion time, however, direction. Nothing to worry about, if it were not
might result in other more macroscopic motion to for the fact that our measurements are only sensi-
be captured and thus confounding our measure- tive to diffusion along this direction. Actually,
ments. Even if this would not be the case, we also that is not fully correct; it’s better to say that they
have to recall that S only decays further in func- are only sensitive to diffusion with a component
tion of b-value (remember Eq.  4.1 again?). The along this direction. Before we start talking fur-
noise level of our measurements, on the other ther about directions, let’s settle on some refer-
hand, does not decrease; that is, using a higher ence frame. We define three (perpendicular) axes
b-value yields a lower signal-to-noise ratio through the brain as follows: x runs from left to
(SNR)! right, y from back to front, and z from bottom to
top. So suppose we would apply the diffusion
gradient along the direction of x, what are the
Conclusions implications then? It basically means that the
measurements are fully sensitive to diffusion
We have learned why the MR measurements in along x, but gradually less sensitive to diffusion
combination with the Stejskal-Tanner equation along directions that increasingly deviate from x,
are not suited to calculate the true self-diffusion up to the point where they are completely insensi-
coefficient of water in voxels containing tissue, tive to diffusion along directions perpendicular
e.g., where diffusion is hindered or even to x (i.e., directions in the yz-plane).
restricted. The obtained apparent diffusion coef- But why should we worry about directionality
ficient (ADC), on the other hand, can provide of diffusion anyway? In our earliest experiments
interesting information about the microstructure with a glass of water or CSF in the ventricles, we
of the tissue under investigation. The minimum shouldn’t: diffusion takes place equally in all
requirements for obtaining it are again a directions. In tissue randomly containing cells—
nondiffusion-­weighted image (S0), a diffusion-­ imagine a bunch of spherical cells packed
weighted image (S) and knowledge of the b-value together—diffusion is hindered and restricted,
that was used for performing the acquisition of yet probably more or less equally in all direc-
the diffusion-weighted image. The ADC is, how- tions. So again, there’s nothing to worry about: as
ever, dependent on the b-value: a higher b-value we are in both cases studying isotropic measure-
results in a lower ADC. It also improves the con- ments, it is sufficient to only measure along a
trast (e.g., of the ADC map), but at the cost of a single direction. Our findings (e.g. calculating the
42 T. Dhollander

ADC) should have been the same for measure- (Fig.  4.5), we start by presenting a classic
ments along any other direction. But let’s con- T1-weighted and T2-weighted image for refer-
sider the more interesting case of the white matter ence. Next is the nondiffusion-weighted image:
in the brain: it consists of long coherent bundles it’s again a T2-weighted image, but it already
of axons, almost resembling a bunch of cylindri- shows the lower spatial resolution at which DWI
cal tubes packed closely together (see Chap. 3). datasets are typically acquired. In this case, the
One can imagine that water molecules in between voxel size equals 2.2 × 2 × 2  mm3. Because the
and inside these tubes have an easier time diffus- image is not diffusion-weighted, but it is acquired
ing along them rather than perpendicular to as part of a DWI dataset, we also sometimes
them. We thus say that diffusion in the white mat- (informally) refer to it as the “B0” (it equals a
ter is anisotropic. diffusion-weighted image with a b-value of 0).
But how relevant is this? Is this anisotropy For convenience, we already applied a whole
large enough to be measured; i.e. can we see it in brain mask to it. On the second row of (Fig. 4.5),
our diffusion-weighted images? To answer this three diffusion-weighted images (DWIs) are
question, we’ll introduce some real data. In shown (also masked). They were all acquired

Fig. 4.5  Top row: T1-weighted image, T2-weighted gradients along the direction of x, y and z. The arrows
image, B0 image (“diffusion-weighted image” with a indicate a region in the genu of the corpus callosum
b-value of 0, i.e., non diffusion-weighted). Bottom row: (GCC), where the anisotropy can be easily seen and
diffusion-weighted images (DWIs) acquired by applying understood
4  From Diffusion to the Diffusion Tensor 43

using exactly the same amount of diffusion  etting a Grip on the Information


G
weighting: b = 800  s/mm2. The diffusion-­ Overload
sensitizing gradients are, however, applied along
different directions: respectively along the direc- In practice, however, we will typically perform
tion of x, y and z. Differences in contrast can the acquisition using more than three different
clearly be seen, which consequently confirms gradient directions. In the previous example, we
that we will have to account for anisotropy in our were just lucky that the structure under investiga-
measurements. tion accidently happened to run along one of the
three mutually perpendicular directions that we
sampled. If it would instead be running at any
Anisotropic Advantages other oblique angle, these three measurements
would clearly be inadequate to determine its
Just as when we introduced the ADC, we’ll also direction. In our experiment at hand, however,
try to use this fact to our advantage: we now we actually acquired DWIs for a total of 45 dif-
have access to a probe that might even provide ferent gradient directions! The specifics of such
us with information on the anisotropy of the an acquisition are presented in the gradient table,
microstructure that hinders and restricts the pro- that contains one row for each acquired image,
cess of diffusion. Applying what we have learned representing its gradient direction and b-value.
from this chapter up to this point, let’s see if we The gradient table for our current experiment is
can already figure out something useful from provided in (Fig.  4.6). As it was already quite
these three diffusion-­weighted images. Consider tedious to infer information by mentally combin-
the indicated region in the genu of the corpus ing three images, considering 45 DWIs all at
callosum (GCC): it has a low DWI-intensity once is nearly impossible. To begin with, we will
along x, but a (relatively) higher DWI-intensity no longer visualize the original DWIs, as they are
along y and z. Because we know that more diffu- still only representing a (partially) decayed
sion causes increased decay of S (the DWI- T2-weighted signal. Because of this, these DWIs
intensity), we can conclude from these images suffer so-called T2 shine-through: a higher inten-
that there is more free diffusion along x, while sity might not (only) result due to hindered/
there is more hindrance and restriction along y restricted diffusion; it might also be caused by an
and z. Translating this to “reality”, we might originally high T2 intensity (e.g., in areas con-
infer that a bundle of tubelike axons runs along taining CSF). Therefore, it is more evident to
the left-right axis in this region, connecting both consider DWIs after normalization by the B0
hemispheres of the brain. Note that we are apply- (i.e., the normalized measurements “S/S0”). Such
ing inductive reasoning here: we know that such normalized versions of our original DWIs for the
a left-right oriented structure would result in three x, y, and z gradient directions are shown in
such a pattern of diffusion and thus also such (Fig. 4.7). Next up is the actual challenge of visu-
DWI measurements along these three directions, alizing the information of all 45 normalized
yet we reason that the latter measurements were DWIs in a conveniently organized way. Rather
effectively caused by the former structure. than showing 45 separate images, we could try to
Considering we only measured along three combine all information of each single voxel and
directions, that’s a pretty strong conclusion. Of visualize it within that particular voxel. Since the
course, inherently we might have also applied different values of S/S0 are a function of the gra-
possible anatomical knowledge and the fact that dient direction, a spherical polar plot is the per-
a structure along this direction makes sense con- fect candidate for the job. In such a plot, the
sidering the spatial/anatomical neighborhood of radius of a sphere is locally manipulated to equal
the region (i.e., the region is in between both the function value at that three-dimensional
hemispheres). angle. We also smoothly interpolated the values
44 T. Dhollander

Fig. 4.6  The gradient


table contains one row for
each acquired image. The
x, y and z components of
the gradient direction are
provided in the first three
columns, while the b-value
is given in the last one. A
b-value of 0 indicates a B0
image; the gradient
direction is irrelevant in
such a case. The red
encircled rows refer to the
DWIs presented in Fig. 4.5

between the 45 directions in order to achieve the Associating larger values with less diffusion
final visualization in (Fig. 4.7). Note that, due to still feels a bit awkward, to say the least. So why
the multitude of information on display, we have don’t we simply employ the ADC values? Easy
to zoom in up to a reasonable level to show enough: just calculate the 45 ADC maps from the
everything with the required amount of detail. normalized DWIs using Eq. (4.4). We present
We choose to further focus on the region of the these maps—again for the three x, y, and z gradi-
GCC that was the subject of our earlier thought ent directions—in (Fig.  4.8). This time, larger
experiment. Furthermore, a little extra color was values equal more free diffusion. We can just as
added to the plot: each point on the surface of the well create a spherical polar plot of the 45 ADC
spherical polar plots is colored according to its values in each voxel, which is again provided for
direction: red is assigned to x, green to y and blue the GCC region in (Fig.  4.8). Larger values are
to z. In (the middle of) the GCC, we spot larger now conveniently oriented along the direction of
values for green (y) and blue (z), and smaller val- the greatest amount of free diffusion, and colored
ues for red (x). By linking larger values to accordingly. Finally, remember that property of
hindrance/restriction, we can thus confirm our the contrast increasing with b-value? Of course, it
hypothesis of an axonal bundle connecting left also applies for measurements (and ADC values)
and right. acquired using different gradient directions: using
4  From Diffusion to the Diffusion Tensor 45

Fig. 4.7  Top row: DWIs for the x, y, and z gradient direc- of the normalized DWI values in a region of the GCC,
tions, after normalization by the B0 (i.e., the normalized overlaid on a map of the average normalized DWI value
measurements “S/S0”). Bottom row: Spherical polar plots

a higher b-value will increase the contrast in white matter up to a measurable extent. Again
these spherical polar plots. However, as we rea- using this to our advantage, we now have a probe
soned before, the SNR will also drop. for the anisotropy of microstructure in each voxel.
There are different ways to visualize data result-
ing from acquisitions using many different gradi-
Conclusions ent directions, yet the most convenient option was
a spherical polar plot of the ADC values in each
We started taking into account the fact that the voxel: such a visualization shows larger values
diffusion-sensitizing gradient is applied along a along the direction exhibiting the greatest amount
certain direction. The resulting DWI measure- of free diffusion. Optional color coding is typi-
ment is only sensitive to diffusion with a compo- cally done according to a directional scheme: red
nent along this direction. From DWIs acquired for x (left-right), green for y (back-front), and blue
using different gradient directions, we could con- for z (bottom-top). The requirements for investi-
clude that anisotropic diffusion takes place in the gating the anisotropic nature of diffusion are a B0
46 T. Dhollander

Fig. 4.8  Top row: ADC maps for the x, y, and z gradient directions. Bottom row: Spherical polar plots of the ADC
values in a region of the GCC, overlaid on a map of the average ADC value

(nondiffusion-­weighted image), a number of yet require more scanning time. And finally, a
DWIs and knowledge of the b-value and gradient higher b-value yields better contrast—also in the
directions that were used for performing the spherical polar plots of, e.g., the ADC values—
acquisition of the DWIs. This latter point is very, but will reduce SNR.
very important! Did we just stress that enough?
Because it is (very, very important): without the
accompanying b-value and gradient directions, The (Apparent) Diffusion Tensor
the full set of carefully acquired DWIs is nigh
useless; we wouldn’t be able to associate the (nor-  otivation and Implications
M
malized) measurements nor the ADC values with of Modeling
any directions. This vital piece of information
should thus be stored with the data; it is often Looking back at the spherical polar plots of the
summarized in a gradient table, as shown in ADC values in (Fig.  4.8), we notice that they
(Fig. 4.6). On the number of gradient directions: appear quite noisy. That’s not surprising, since
more measurements are of course always better, they simply present a logarithmic transformation
4  From Diffusion to the Diffusion Tensor 47

of the original (normalized) data: nothing is mod- tensor elements later. Given such a tensor D, we
eled, all the measurement noise is still showing can “evaluate” it for a given direction g by using
(albeit logarithmically transformed … remember the following expression:
this, as it will happen to bug us later on). And
thus models were invented. Without going into g T Dg = g x2 Dxx + g y2 Dyy + g z2 Dzz
(4.6)
the how and why of some historical choices that + 2 g x g y Dxy + 2 g x g z Dxz + 2 g y g z Dyz
have been made in model development, we’ll just
introduce the (legendary) diffusion tensor model where gT is the transpose of g. The right side of
[4] that is central to the theory and practice of the equation simply shows what you would
DTI. In this context, to be exact, we should refer obtain if you did the symbolic math by hand
to it as the apparent diffusion tensor. This name using the vector and tensor element symbols
refers to the fact that we will employ a tensor to from Eq. (4.5). The outcome of this expression—
represent/model the values of the ADC in func- if we were to fill in some specific numbers repre-
tion of (gradient) direction, in each voxel. This senting the vector and tensor elements—is thus a
means that, once we have somehow determined single scalar number: the value of our tensor
the correct parameters of this model in each model, along a given direction. As we will now
voxel, we can evaluate it for as many directions employ such a tensor to symbolize the ADC val-
as we like in order to visualize it again as a spher- ues, we can simply plug it into the good old
ical polar plot of (modeled) ADC values. As the Stejskal-Tanner Eq. (4.1) to obtain the following
diffusion tensor model has only six parameters expression:
(compare this to the 45 ADC values we just - b•gT Dg
obtained from our dataset in each voxel!), it will S = S0 •e (4.7)
greatly simplify the features of our directional Don’t take this one lightly: this is the essence of
profile of the ADC. DTI. It provides the direct relationship between
the chosen experimental parameters (b and g),
the measurements (S and S0), and the parameters
Understanding DTI, in Theory: of the diffusion tensor model (D). It now effec-
The Maths! tively includes the gradient direction g that we
took abstraction of before, while the vehicle to
Mathematics … it’s not as hard as it sounds, so describe the ADC is no longer a single number,
let’s just get to it then! From this point on, we but a tensor that can describe values that vary in
will represent a (gradient) direction by a three-­ function of (gradient) direction. Just like we did
element column vector g, and the apparent diffu- before with the Stejskal-Tanner equation, we can
sion tensor D by a 3 × 3 symmetric matrix: rewrite Eq. (4.7) to single out the parts that equate
to the ADC:
gx Dxx Dxy Dxz
S
g = [ g y ] D = [ Dxy Dyy Dyz ] (4.5) - ln( )
S
gz Dxz Dyz Dzz g T Dg = 0
(4.8)
b
For the mathematics (and software that employs The right-hand side equals the expression of the
it) to work out well, g should be a unit vector. As ADC that we introduced before (i.e. Eq.  4.4),
stated before, the tensor D has only six free while the left-hand side simply says that we
parameters (the tensor elements Dxx, Dyy, Dzz, Dxy, would like to see this ADC value arising from our
Dxz, Dyz) because its matrix is symmetric: the ele- model D when evaluated for the gradient direc-
ments above and below the main diagonal are the tion g that this particular ADC value relates to.
same. We’ll get into the meaning of these separate Completely writing out the left-hand side
48 T. Dhollander

expression using Eq. (4.6) finally yields the with it? For starters, let’s take a look at some
­following result: maps of these six tensor elements. There are two
distinct categories amongst them: the diagonal
g x2 Dxx + g y2 Dyy + g z2 Dzz elements (Dxx, Dyy, Dzz) and the off-diagonal ele-
+2 g x g y Dxy + 2 g x g z Dxz + 2 g y g z Dyz (4.9) ments (Dxy, Dxz, Dyz). The former are presented
= - ln( S / S0 ) / b on the first row of (Fig. 4.9), while the latter are
shown on the second row. The interpretation of
If we now perform a DWI experiment as before the diagonal elements is straightforward: they
(acquiring S and S0, carefully noting down b and represent ADC values along the respective
g), we can fill in everything but the six unknown directions of x, y, and z. Because we’ve shown
parameters of the diffusion tensor model. As maps of the original (unfitted) ADC values
solving a single equation for six unknowns is along these directions in (Fig. 4.8), we can com-
quite an impossible task, we’ll clearly need more pare them directly to the maps of Dxx, Dyy, and
of these equations, and by consequence more Dzz in (Fig. 4.9). Indeed, they look more or less
acquisitions. Mathematically, we know that at alike. The more careful observer may note that
least six equations will be necessary to be able to the latter look less noisy. This is not surprising:
determine the full apparent diffusion tensor. In they represent fitted values, i.e., all 45 measure-
practice, we’ll be needing at least six DWIs for ments contributed to them. From what we
different gradient directions as well as a single already know, we can even figure out mathemat-
B0 to normalize our measurements to. As stated ically why e.g. Dyy corresponds to the value of
before, it is essential that every DWI is tied to its the tensor model (i.e. the ADC) along y: just fill
respective gradient direction. Acquiring more in [0 1 0]T (i.e., the direction of y) as direction g
DWIs (for different gradient directions) will in Eq. (4.6) and evaluate using the right-hand
lead to more than six equations. In such a set- side expression; the outcome trivially equals
ting, no exact solution for the six unknown ten- Dyy. We can thus conclude that the diagonal ele-
sor elements generally exists, because the full ments are in practice meaningful and quite easy
system of equations is overdetermined. This to understand. The off-­diagonal elements (sec-
actually is a good thing! Even though we can ond row of Fig. 4.9), on the other hand, offer a
find a single exact solution in case of six DWI less intuitive source of information. They repre-
measurements, this solution will also exactly sent the covariance between each pair of axes
represent all the noise in the data. If we perform (i.e., xy, xz, and yz). That’s because the full dif-
the acquisition using a larger amount of differ- fusion tensor is actually a covariance matrix.
ent gradient directions, a solution will have to be Apart from being a great conversation starter at
found that fits the data as good as possible. We an engineering party, those last two sentences
then hope that the part that doesn’t fit the model won’t get you anywhere in daily practice: the
(i.e., the residuals) is the noise, which we (opti- off-diagonal elements just don’t really have a
mally) don’t want to model anyway. We’ll focus direct practical use or meaning. The only reason
on the issue of tensor fitting later. For the time we do discuss them here, is to emphasize what
being, let’s take it for granted. they don’t mean: they do not represent the val-
ues of the ADC along some diagonal direction
(so don’t mistake them for that!). This should
 nderstanding the Tensor Elements,
U also be clear from the fact that they equally
in Practice cover a range of positive as well as negative val-
ues (while ADC values should not be negative).
So, effectively applying such a tensor fitting One final property worth mentioning though: if
method to our 45 gradient direction dataset at all off-diagonal elements are zero, the tensor is
hand, we end up with six numbers in each voxel, perfectly aligned to the x-, y-, and z-axes. What
i.e., the components which describe an apparent that means will become more clear if we visual-
diffusion tensor. Now what can we actually do ize the tensor in 3D.
4  From Diffusion to the Diffusion Tensor 49

Fig. 4.9  Top row: Maps of


the diagonal diffusion tensor
elements (Dxx, Dyy, Dzz).
Middle row: Maps of the
off-diagonal diffusion tensor
elements (Dxy, Dxz, Dyz). The
background grey level
equals zero; darker/brighter
levels represent negative/
positive values. Bottom row:
Spherical polar plots of the
ADC values provided by the
diffusion tensor model in a
region of the GCC, overlaid
on a map of the average
ADC value. Note the
characteristic peanut shapes
that appear in the GCC

 nderstanding the Tensor, in Practice:


U within regions. In regions where a single bundle
Peanuts! of axons is present, e.g. the GCC, these plots typi-
cally take on the shape of peanuts. The advantage
Talking about visualization, let’s take a look at a of modeling is that some features of interest, such
spherical polar plot of the ADC values actually as the main direction of the tensor, are recovered
represented by the fitted diffusion tensors. These more prominently. From the region shown in
are shown for our trustworthy GCC region on the (Fig.  4.9), it is now also more evident that the
bottom row of (Fig. 4.9), and can again be directly nearby CSF in the ventricles exhibits an isotropic
compared to the original (unfitted) values in pattern of diffusion. And finally, for those who’d
(Fig. 4.8). From this comparison, it is obvious that like to go just that extra mile in interpretation:
the noisy appearance has been greatly reduced: consider again one of those curious maps of the
while the original plots showed a unique and dif- off-diagonal elements, Dxy, and note that it indeed
ferent pattern in each voxel (due to the varying shows a value of zero for the voxels right in the
noise), the directional profiles that represent the middle of the GCC, where the main directions of
tensor fitted values are much more consistent the tensors are nicely aligned to the x-axis.
50 T. Dhollander

Conclusions represent ADC values along the x-, y-, and z-axes,
while the off-diagonal elements represent the
We have introduced the (apparent) diffusion ten- covariance between pairs of those axes. Maps of
sor model [4], which is used in DTI to represent the former thus provide a meaningful interpreta-
the ADC values of our measurements along dif- tion, while maps of the latter are neither intuitive
ferent (gradient) directions. The diffusion tensor nor useful in daily practice. Just don’t mistake the
D is represented by a 3 × 3 symmetric matrix, off-diagonal elements for ADC values along
containing six unique tensor elements, and can be some oblique angle. A final overview of the most
evaluated along any direction g by the expression important steps taken up to this point is shown for
gTDg. Casting this expression in the role of the a single voxel in the middle of the GCC in
ADC value in the Stejskal-Tanner equation yields (Fig. 4.10): from raw DWI measurements, to cal-
the one and only equation at the core of DTI: it culated ADC values, and finally the fitted tensor!
directly relates the experimental parameters, the
measurements and the parameters of the diffu-
sion tensor model to each other. This equation
can again be rewritten to show clearly that the Eigenvalues and Eigenvectors
diffusion tensor model is meant to fit the ADC
values. As there are now six unknowns in this  he Tensor Elements: Not Very
T
equation, the minimum requirements for obtain- Practical
ing the diffusion tensor are a B0 (non diffusion-
weighted image), at least six DWIs and Looking back at the ADC peanuts that represent
knowledge of the b-value and gradient direc- the diffusion tensor values in (Fig.  4.9) and
tions that were used for performing the acquisi- (Fig.  4.10), we notice that the tensor model
tion of the DWIs. We simply cannot stress enough indeed did a good job in capturing the most
that knowledge of the b-value and gradient important features of the angular ADC profile:
directions, i.e., the full gradient table as shown in we can clearly observe its main direction, the
(Fig.  4.6), is absolutely essential to fill in the maximal ADC value (along this main direction),
equations and obtain the diffusion tensors! Fitting etc. and how these features relate to each other
the tensor model to data with a larger (than six) over larger regions (e.g., qualitatively observe the
number of DWIs reduces the noisy appearance of curving global path of the axon bundle in the
the ADC values when visualized as a spherical GCC). Although these very practical features are
polar plot. In regions of white matter containing a in each voxel captured and described by those six
single consistent bundle of axons, the plot has a unique tensor elements, it’s not immediately
characteristic peanut shape that clearly shows clear how. We do have the diagonal tensor ele-
features such as the main direction of the tensor. ments (Dxx, Dyy, Dzz) that come with a clear
The diagonal elements of the diffusion tensor ­interpretation (i.e., the ADC values along x, y,

Fig. 4.10  From DWI data to the tensor, for a single voxel in the middle of the GCC. Spherical polar plots of the DWI
values (left), the ADC values (middle) and the ADC values evaluated from the fitted tensor (right)
4  From Diffusion to the Diffusion Tensor 51

and z). From the ADC peanuts in (Fig. 4.9), we benefits are twofold. These “new diagonal tensor
can tell for instance that the value of Dxx will elements” should provide us with everything we
coincide with the maximal ADC value of the pea- need to know about the shape and size of the ten-
nuts right in the middle of the GCC, because sor, independently of its orientation. On top of
those peanuts are nicely aligned along x. that, the customized set of axes by itself also
However, as we move away from that middle describes the full 3D orientation of the tensor. To
region, the orientation of the peanuts changes, conclude, such a representation thus effectively
causing Dxx to gradually take on lower values. splits up information about the orientation and
The information on the maximal ADC value of the shape/size of the tensor, while the classical
the peanuts is now “spread out” somewhere six tensor elements mix it all up.
between Dxx and Dyy. And to make matters even
worse, the information on how this “spread” is
balanced between those components, is in turn  aths to the Rescue:
M
encoded somehow by Dxy, one of those elusive The Eigendecomposition
off-diagonal tensor elements. That’s also why we
didn’t run into all that trouble right in the middle Now that we know what we want, the question
region: Dxy equals zero in that part of the GCC. remains how to obtain it. In this case, we are
lucky: the mathemagician can help us out with
something called eigendecomposition. Applied
Reasoned Wishful Thinking to the diffusion tensor, it basically boils down to
of Alternatives rewriting the 3 × 3 symmetric tensor in the fol-
lowing format:
So, what is at the core of all this confusion and
why do we need to be so tedious about trying to D=
infer useful information from the tensor compo-
(4.10)
nents? The answer is simple: our definition of
   l1 0 0  ε1 
axes (i.e., x, y, and z) is in fact quite artificial
and—more importantly—very rigid. To formu- [ε1 ε 2 ε3 ] × [ 0 l2 0 ] × [ ε 2 ]
late it in another, maybe more clear, way: the    0 0 l3  ε3 
axon bundles simply couldn’t care less about how
we happened to define our globally fixed axes; where λ1 ≥ λ2 ≥ λ3, and ϵ1, ϵ2, and ϵ3 are three-­
they just happily twist and curve through the full element unit vectors that are mutually perpen-
3D space. On those rare occasions where the ten- dicular to each other. The right-hand side
sor perfectly aligns to our predefined axes, we get intuitively reads: start with a tensor (with diago-
lucky: the off-diagonal elements become zero nal elements λ1, λ2, and λ3) aligned to the axes (x,
and the three diagonal components describe the y, and z), and then reorient it to a new set of axes
shape and size of the tensor in a more direct, intu- (ϵ1, ϵ2, and ϵ3). The process of eigendecomposi-
itive manner. But how do we solve our problem tion aims to reverse this set of actions: it starts
in all those other voxels then? As we just stated, with the diffusion tensor D, and subsequently
the axon bundles are not going to adjust them- tries to figure out which axis aligned tensor could
selves to our axes; and thus the only solution will have been reoriented to which new set of axes in
be to adjust our axes to them in each voxel order to obtain D. The result is referred to as the
instead. So, what we are looking for is a new eigenvalues (λ1, λ2, and λ3) and eigenvectors (ϵ1,
description of the diffusion tensor that provides a ϵ2, and ϵ3) of D. They come in so-called eigen-
set of axes aligned to the tensor as well as three pairs (e.g., λ2 is paired to ϵ2): an eigenvalue (e.g.,
“new diagonal tensor elements” to describe the λ2) represents the ADC value of the tensor along
tensor within this new local set of axes (the “new the direction of the corresponding eigenvector
off-diagonal tensor elements” become zero). The (e.g., ϵ2). The eigenvector ϵ1 that is associated
52 T. Dhollander

with the largest eigenvalue λ1 is also referred to as before, the combination of all three eigenvalues
the principal eigenvector. It plays quite an impor- fully encodes the exact total shape and size of the
tant role in DTI: due to its orientation along the tensors (and by consequence, the ADC peanuts)
peak direction of the ADC peanut, it’s indicative by providing the ADC value along three perpen-
of the local direction of the axon bundle. While dicular axes aligned to the tensors (the eigenvec-
the largest eigenvalue λ1 equals the maximal tors). In some regions (e.g., the GCC, or the white
value of the ADC peanut, the smallest eigenvalue matter in general) a larger mutual difference
λ3 represents its minimal value. between the eigenvalues can be seen as compared
to other regions (e.g., the CSF). This clearly
relates to the differing amounts of anisotropy that
 nderstanding the Eigenvalues,
U we could also see in the ADC peanuts. Because
in Practice all information on the shape and size of the ten-
sors is stored in the eigenvalues, they will also be
Maps of the eigenvalues (λ1, λ2, and λ3) are pre- the basis for other tensor measures that are inde-
sented in the top row of (Fig. 4.11). A strict order- pendent of the tensor’s orientation; but we’ll get
ing (λ1 ≥ λ2 ≥ λ3) is always enforced. As stated to that later.

Fig. 4.11  Top row: Maps of the eigenvalues (λ1, λ2, λ3). Bottom row: Directionally encoded color (DEC) maps of the
eigenvectors (ϵ1, ϵ2, ϵ3)
4  From Diffusion to the Diffusion Tensor 53

 nderstanding the Eigenvectors,
U mation on the size/shape of the tensor from infor-
in Practice mation on its orientation. This is achieved by
recovering a set of axes (the eigenvectors) that is
Directionally encoded color (DEC) maps [5] of locally aligned to the tensor as well as three ADC
the eigenvectors (ϵ1, ϵ2, and ϵ3) are provided in values (the eigenvalues) of the tensor along these
the bottom row of (Fig. 4.11). Since each eigen- new axes. The solution thus comes as a set of
vector has unit length, no magnitude information eigenpairs: a certain eigenvalue encodes the ADC
is represented in these maps; only orientation is along a specific eigenvector. Whereas the eigen-
encoded. This is achieved by assigning the three values encode the size/shape independently of the
elements of an eigenvector to the red, green and orientation, the eigenvectors describe the orienta-
blue channels of a color image. As the eigenvec- tion independently of the size/shape. A final sche-
tor itself is specified relative to the original (x, y matic (2D) example, illustrating these properties
and z) axes, the meaning of the colors is similar and providing an overview of the relation between
to the scheme we used before for displaying the most important tensor-related numbers we’ve
spherical polar plots: red is linked to x, green to y, come across up to this point, is shown in
and blue to z. As mentioned before, one of the (Fig.  4.12). The eigenvector associated to the
most important outcomes of DTI is the orienta- largest eigenvalue is also referred to as the princi-
tion of the principal eigenvector ϵ1. Within pal eigenvector. Mapping eigenvectors is typi-
regions of the white matter (e.g., the GCC), the cally done by use of directionally encoded color
DEC map of ϵ1 shows a consistent and smoothly (DEC) maps. One of the key practices in DTI con-
evolving pattern that can intuitively be related to sists of mapping the principal eigenvector, since
the local orientation of the axon bundles. In it is indicative of the local orientation of the axon
regions such as the CSF, the orientation of ϵ1 bundles. In regions of white matter such as the
proves to be more or less random, resulting in a GCC, this map shows a consistent pattern. In
noisy appearance of its DEC map in those par- regions of (nearly) isotropic diffusion such as the
ticular regions. Associated with the isotropic pat- CSF, however, the principal eigenvector becomes
tern of diffusion in these regions, we should ill defined, leading to a noisy appearance of the
ideally observe a spherical ADC plot (instead of associated DEC map. Since there are no axon
a peanut), satisfying λ1 =  λ2 =  λ3. However, due to bundles hindering/restricting the diffusion in such
random noise in the data, there might be a slight a region, the principal eigenvector therein is pretty
deviation from this pattern. The orientation of the meaningless anyway.
principal (and any other) eigenvector is entirely
determined by the random noise in such a case.
Whenever two (or all three) of the eigenvalues of Visualizations, Measures, and Maps
a given tensor are (nearly) equal, we say that the
corresponding eigenvectors become ill defined. Aiming for Usability: Tensor Glyphs

While all maps and visualizations (using, e.g.,


Conclusions spherical polar plots) presented up to this point
have provided us with great insight into the
We introduced the eigendecomposition of the dif- underlying information that eventually leads to
fusion tensor in its eigenvalues and eigenvectors. the diffusion tensors and describes their main
The six diffusion tensor components mix up infor- features, we are yet to encounter the visualiza-
mation on the shape, size and orientation of the tions and maps that we’re most likely to run into
tensor and it becomes hard to untangle the infor- when processing DTI data in practice. Let’s first
mation we’re typically interested in by purely have a look at the most common 3D visualization
intuitive reasoning on these components. Our new of the diffusion tensor, which is not the ADC
representation, however, nicely separates infor- ­peanut we’ve already become acquainted with
54 T. Dhollander

Fig. 4.12  Example of 2D tensors and the convenience of diagonal tensor elements encode the value of the ADC
eigenvalue decomposition. Left: Perfectly axis aligned peanut along the global axes, yet do not fully define the
tensor. The diagonal tensor elements directly define the tensor shape. The eigenvectors provide a new set of axes
tensor shape. The eigenvectors coincide with the global along which the eigenvalues directly provide the informa-
axes and the eigenvalues are equal to the diagonal tensor tion on the shape
elements. Right: General (not axis aligned) tensor. The

(even though it most directly shows all the values tion of their full 3D shapes: e.g. in (Fig. 4.13), we
that the tensor represents). Instead, we will only freely rotated the slice of glyphs to a certain
visualize the most prominent features that define angle. Especially when the axon bundles are not
its size/shape and orientation, as provided by its running “in plane”, the ability to freely rotate the
eigenvalues and eigenvectors: meet the tensor tensor field is a helpful addition. A reason to pre-
glyphs! Two variations are shown in (Fig. 4.13) fer ellipsoids (instead of e.g. cuboids) might be
for our familiar GCC region: cuboids and ellip- that they do not overexaggerate some features of
soids. In general, a mostly primitive 3D shape is the tensor in cases where those features are not
chosen (e.g., a rectangular cuboid or a scalene very meaningful or appropriate anyway. A good
ellipsoid) and its three main dimensions are example is the isotropic diffusion in the CSF, as
scaled by the eigenvalues (or a transformation seen in (Fig.  4.13): the cuboids become cubes,
thereof) and aligned along the eigenvectors. but still clearly indicate the orientation of the
Optionally, the glyph is colored according to the eigenvectors, even though they are ill defined in
DEC map of the principal eigenvector (i.e. using this region. The ellipsoids, however, take on the
the map of ϵ1 in (Fig. 4.11)). Comparing our pre- shape of spheres, and thus any visual cues of the
vious ADC peanuts in (Fig. 4.9) with the newly eigenvectors inherently fade away (apart from
obtained glyphs in (Fig. 4.13), we can clearly tell the coloration, which is of course also not very
the latter score higher on the usability scale: they informative in this region). Another reason why
show a more contrasting description of the fea- ellipsoids are a meaningful choice is that they
tures that really matter (and still fully define the actually come with a true meaning when scaled
tensor and thus its ADC peanut). The most com- using the square roots of the eigenvalues [6]:
monly visualized glyph shape is the ellipsoid [6], under the model of diffusion that DTI assumes, if
but since a cuboid requires far fewer polygons to we would investigate a single water molecule
be drawn onscreen, it lends itself for faster inter- that starts at the center of the ellipsoid and is
action with larger tensor fields. Interaction with a allowed to diffuse randomly during a fixed time
field of glyphs is useful for a better characteriza- interval, then there is an equal chance for it to
4  From Diffusion to the Diffusion Tensor 55

Fig. 4.13  Tensor glyphs


(top: cuboids; bottom:
ellipsoids) in a region of
the GCC. The glyphs are
colored according to the
DEC map of the principal
eigenvector ϵ1

displace to any specific point on the surface of ant measures: they tell us something about the
this ellipsoid. It might take a few reads of that size or shape of the tensors, independently of their
sentence before one may grasp its meaning, and orientation. Therefore, they are typically defined
we won’t even go into why it is true; the fact just in function of the eigenvalues of the tensors. Let’s
is that there exists a pretty good reason to prefer start with a straightforward one: the mean diffu-
these ellipsoids over any other specific glyph! In sivity (MD) [7]. It is defined as follows:
practice, however, any glyph will do for explor-
l1 + l2 + l3 Dxx + Dyy + Dzz
ing the data (even though some software may MD = = (4.11)
offer many different options), as long as it’s easy 3 3
on the eyes and the processing power of the As simply being the average of the eigenvalues, it
machine one is working on. describes the overall size of the tensor and as
such represents a rotationally invariant ADC
measure. A map of it is provided in (Fig. 4.14).
 apping Size: Mean Diffusivity (MD)
M The same contrast is sometimes also referred to
and Friends as the trace [7], which equals the sum of the
eigenvalues. Other related variants exist, such as
Now let’s consider some of the more common dif- the pair of axial diffusivity (equating to the first
fusion tensor measures. All the measures we’re eigenvalue) and radial diffusivity (equating to the
about to present are so-called rotationally invari- average of the second and third eigenvalues). As
56 T. Dhollander

seen in Eq. (4.11), the MD can (surprisingly) also tion becomes ill defined. Hence, the FA map is
be obtained by averaging the diagonal tensor ele- the perfect candidate to weight the DEC map of
ments: even though these individual elements are the principal eigenvector from (Fig. 4.11): doing
dependent on the orientation of the tensor, their so will hide the colors in regions where they are
average is not. An important warning at this ill defined (i.e., where they are noisy, confusing,
point: this does not mean that we can simply and meaningless). The result is known as the
acquire three DWIs using perpendicular gradient DEC FA map [5], and is also presented in
directions, and subsequently average the three (Fig. 4.14). This map isn’t the most iconic DTI
ADCs in order to obtain the same rotationally map for no reason: it’s a very handy one and
invariant MD [7]! It only applies for three per- becoming acquainted with the color encoding is
pendicular ADC values as evaluated from a ten- key to quickly interpreting a lot of the valuable
sor model, so six gradient directions are still the and unique information in the dataset at once.
bare mathematical minimum in order to account Mentally processing DEC should become second
for the anisotropy in the measurements! nature; red for x (left-right), green for y (back-­
front), and blue for z (bottom-top). Take a look
again at the original (grayscale) FA map. Notice
 apping Fractional Anisotropy (FA)
M how easily we could be tempted to believe that
and Orientation every bundle-like feature of this map represents
an in-plane axonal bundle. Now shift your atten-
Next up is the fractional anisotropy (FA) [7], tion back to the DEC FA map, and realize that
probably the most unique selling point of DTI. It blue stands for an orientation perpendicular to
is calculated by the following hefty formula: the visualized slice. There you have it; that’s
some indispensable DEC information for you!
FA =
3 (l1 - l ) + (l2 - l ) + (l3 - l )
2 2 2

× (4.12)  xploring Shape Space and Reaching


E
2 l12 + l22 + l32 Beyond…

where l is the average of the three eigenvalues. Finally, let’s briefly touch upon a triplet of
In words, this amounts to the standard deviation slightly more exotic measures: a linear measure
of the eigenvalues divided by their root mean (cl), a planar measure (cp), and a spherical mea-
square. Or, more simply: a measure for how sure (cs) [8]. This is what their formulas look like:
much the eigenvalues differ, but normalized, so it
becomes independent of their absolute magni- l1 - l2 2 × (l2 - l3 )
cl = cp =
tude. As such, it describes an aspect of the shape l1 + l2 + l3 l1 + l2 + l3
of the tensor, independently of its size (and of 3 × l3 (4.13)
course, orientation). Because of the way the for- cs =
l1 + l2 + l3
mula is carefully normalized, the FA takes on
values in an interval between zero and one, the All of them are again automatically restricted to
former representing perfect isotropy (i.e., all an interval of values between zero and one. The
eigenvalues are equal) and the latter correspond- sum of these three measures exactly equals one.
ing to perfect anisotropy (e.g., the extreme case The full triplet of measures provides the coordi-
where only λ1 would have a nonzero value). An nates of our tensor in some “shape space”: a
FA map is provided in (Fig. 4.14). From this, we higher cl means a more linear, prolate, cigar-­
learn that the white matter clearly has higher shaped tensor ellipsoid; a higher cp means a more
anisotropy than any other (healthy) tissue in the planar, oblate, pancake-shaped tensor ellipsoid; a
brain. As we already know, in regions of low higher cs means a more spherical, isotropic, ball-­
anisotropy, the principal eigenvector’s orienta- shaped tensor ellipsoid. Just like the FA, these
4  From Diffusion to the Diffusion Tensor 57

Fig. 4.14  Top row: Maps of the mean diffusivity (MD), sure (cs), combination of cl and cp using red and green
fractional anisotropy (FA), DEC FA. Bottom row: Maps of color channels
a linear measure (cl), planar measure (cp), spherical mea-

measures each describe an aspect of the shape of cannot simply cause such a pattern if all axons in
the tensor, independently of its size (and of the voxel are coherently running along the same
course, orientation). One could even use them to direction: they must be curving or dispersing
come up with new anisotropy measures, such as (within certain planes), or maybe more than one
1 − cs =  cl + cp [8]. Maps of the shape measures are population of axons is present in such voxels.
provided in (Fig.  4.14). We also present a map Whatever the underlying situation might be in
where we employ the red and green color chan- those voxels, the planarity hints at certain limita-
nels to encode cl and cp. This final map’s absolute tions of the DTI model ….
intensity thus equals the custom anisotropy mea-
sure we just mentioned, while the color somehow
shows what “kind of anisotropy” is present: lin- Conclusions
ear or planar. Interestingly, we notice (in the pre-
sented slice) that mostly the central part of the We introduced some of the most mainstream
corpus callosum (including the GCC region we visualizations and maps that one is bound to run
have been considering in all our examples) shows across in daily DTI practice. These include the
highly linear behavior, while many other regions visualization of the tensors by glyphs (most nota-
of white matter contain a decent portion of pla- bly the diffusion tensor ellipsoid) and maps of the
nar diffusion. Reasoning about the axon bundles mean diffusivity (MD), fractional anisotropy
as a bunch of cylindrical tubes, as we did before, (FA), DEC FA as well as a slightly more exotic
58 T. Dhollander

triplet of linear, planar and spherical measures material to write a decent book on the subject
(cl, cp, cs). Other measures exist, but these are alone, consider the following as your average
typically variations of—or at least heavily quick and dirty hitchhiker’s guide to making the
inspired by—the ones we presented: axial diffu- right choice. In the foreign restaurant of tensor
sivity, radial diffusivity, the trace, several variants fitting, it should enable you to more or less trans-
of anisotropy measures, etc. The most standard late the menu, pick something that you’re not
measures of them all, however, are the typical allergic to, all the while giving you the confi-
couple of MD and FA: the former representing dence that your choice will leave you satisfied up
the average size of the tensor (independent of to a certain level, but also allow you to leave the
shape and orientation) and the latter encoding its restaurant in time, so you can still catch your bus.
anisotropy, an aspect of the shape of the tensor How they actually arrange stuff in the kitchen
(independent of size and orientation). Information though, is the least of our concerns. Have a seat;
on the orientation can also be included by com- the daily menu consists of: linear least squares
bining the FA map and a DEC map of the princi- (LLS), weighted linear least squares (WLLS), and
pal eigenvector in order to obtain the DEC FA nonlinear least squares (NLS). On top of that,
map. This map is not only iconic for DTI, but it’s there’s also today’s special: robust estimation of
also a very handy tool to quickly gain insight into tensors by outlier rejection (RESTORE). Now
any DTI dataset. let’s have a look at our advice!

Tensor Fitting Methods LLS: Quick and Dirty

Facing the Issue Linear least squares (LLS) is the most basic


choice. It will solve your system of equations—
Up to now, we’ve been taking an essential step in of which each single one takes on the form of Eq.
the whole process for granted: the actual tensor (4.9)—by minimizing the sum of squared residu-
estimation. As a matter of fact, this is the least als of those equations. In the case where we have
trivial step along the pipeline; everything else more than six equations (as opposed to only six
we’ve discussed up to this point simply consists unknowns), it is typically impossible to perfectly
of applying some well-defined and quite straight- satisfy all equations. The error or difference that
forward formulas in the right order. Even the still exists between the left- and right-hand sides
eigendecomposition—or at least what we desire of one such equation is referred to as its residual.
it to yield for an outcome—is exactly defined In practice, minimizing the sum of the squared
(i.e., Eq. 4.10; no more, no less), and we can rely residuals of all the equations amounts to “spread-
on computer science to provide us with an algo- ing out” those unavoidable residuals as much as
rithm that does the job. In those cases where the possible over all of them. This is equivalent to
outcome proved to be ill defined (e.g., isotropic stating that each equation has an “equal say” in
diffusion), the ill defined parts of the outcome the process of the fit. A great advantage of LLS is
(i.e., the eigenvectors) were not informative any- that it can be implemented as a single-step pro-
way. But the tensor estimation … that’s an cess. That’s right: it just takes a single specific
entirely different beast! Different, because this operation on the whole system of equations to
time even the definition of “what we want” is not automatically obtain the solution that optimally
all that clear. Or is it? We simply want the tensor minimizes that sum of squared residuals.
to fit the data (or the ADC values …?) as good as Depending on your hardware and the size and
possible, right? But what is “as good as possi- number of DWIs in the dataset, you can have
ble”? It’s vague, that’s what it is; and hence, a your tensors rolling out in mere seconds! In fact,
plethora of definitions and associated fitting the tensors we generated in this chapter—and
methods exist. While it easily provides enough thus all the resulting visualizations and maps
4  From Diffusion to the Diffusion Tensor 59

we’ve been looking at—are the result of a single has to be made to LLS to take them into account
quick application of LLS.  For that specific pur- and obtain a WLLS fit, which now truly provides
pose, LLS is certainly well suited: qualitatively us with the optimal correct fit! It still only takes a
speaking, our tensors and the subsequently calcu- single (slightly bigger) operation on the whole
lated maps of tensor measures look perfectly fine. system of equations to get this solution: it might
So, why don’t we just stick with LLS for all take a few extra seconds, but it still just remains a
intents and purposes then? The problem is subtle matter of mere seconds to have your tensors
and quite well hidden: it actually concerns the again rolling out; yet much more accurately. If
fact that we allowed each equation to have an we would have generated all the maps in this
equal say in the fit. This assumes that each value chapter based on a WLLS fit of the tensors, you
that we are trying to fit, was provided to us with wouldn’t have noticed the difference: it doesn’t
an error (i.e., the measurement noise) of a “simi- suddenly change the visually informative con-
lar magnitude”. In more professional words and trast of those maps. For quantitative purposes
adapted to how we specifically formulated Eq. (such as group studies) though, it certainly mat-
(4.9): LLS assumes that the noise on the ADC ters, a lot: WLLS already removes a great deal of
values (i.e., the right-hand side of each equation, inherent biases on final measures (such as MD
which we are trying to fit with the left-hand side) and FA) that are typically caused by careless use
results from a distribution that has the exact same of LLS. So, why don’t we just stick with WLLS
variance for all our different ADC values. While for all intents and purposes then? Again, a sneaky
this is the case for our DWI values (that originate problem manifests itself: to determine the
directly from the scanner, where the noise is weights, we need the magnitudes of the original
“officially” added to the measurements), it does data … without the noise. Of course, once we
not apply to the ADC values: the distribution of have obtained a fitted tensor, we could reason that
the noise on the original data is logarithmically we got rid of the noise (because, optimally, only
transformed along with those data to obtain the the noise is left in the residuals). We could then
ADC values! You might remember that we men- evaluate that tensor for all gradient directions and
tioned this before, additionally stating that it calculate from the ADCs back to the DWIs, i.e.
would happen to “bug us later on”. So now, here the noiseless magnitudes that we needed to deter-
it is: officially bugging us. The problem at hand is mine the weights. So, if we could obtain a fitted
that each “measured” (i.e., calculated) ADC tensor, then we would also have our weights; but
comes with noise of a different variance: i.e., we in order to obtain a fitted tensor, we need those
can trust some ADCs more (or less) than others. weights in the first place. Yes, that’s a chicken-
It seems sensible to weigh the amount of say of and-­egg problem we’re facing here. No perfect
each equation in the fit with this information. solution exists (and thus, unfortunately, also
That’s where weighted linear least squares WLLS can never be perfect). A first approach
(WLLS) kicks in. could be to just use the magnitudes of the original
noisy data to determine the weights. This may,
however, result in a worse outcome as compared
WLLS: Still Quick, Less Dirty to using plain old LLS! A second trick is to start
by performing a LLS fit, and get DWI magnitudes
Weighted linear least squares (WLLS) assigns to from this fit (where the noise should then already
each equation (still of the form of Eq.  4.9) a be accounted for up to a great extent) in order to
weight according to how much the original noise determine the weights for a subsequent WLLS fit.
variation is affected by the logarithmic transform One could then even repeat this process in the
of the data. These weights directly depend on the hope of getting gradually better fits and subse-
magnitudes of the original data (i.e., the intensity quent weights for the next fit (but this typically
of the different DWIs). In practice, once these does not add much: most of the “magic” is in
weights are known, only a limited modification using that first LLS just for a robust set of
60 T. Dhollander

weights). In practice, it’s all still fast: a LLS could come up with a nifty trick of finding that
­followed by a WLLS. The danger lies in the fact lowest point in a single step. NLS, on the other
that some software packages might perform hand, is just dropped somewhere on the land-
WLLS using the first approach, yielding worse scape and has to start a walk in the unprepared
results as compared to LLS. On the other hand, a hitchhikers fashion: without a map (because the
responsible implementation of WLLS using the landscape is too big and complex) and just rely-
second approach should definitely lead to better ing on its eyes and feeling to gradually move to
results, without any significant increase in com- lower regions. In theory, truly solving the NLS
putation time as compared to LLS: it’s typically problem will yield the optimal result. However,
still done in mere seconds! However, we can the problem is not easy to solve. Due to the lim-
never truly know the correct weights due to the ited range of sight in the mountains, an NLS
chicken-and-egg format of the problem: so the algorithm might get stuck in a suboptimal valley
approach doesn’t fix everything. The core of the (not knowing there exists another lower valley
original problem was that the noise got logarith- somewhere). Some algorithms are more robust
mically transformed in the ADC values, and that against this than others, but it’s nearly impossible
we formulated Eq. (4.9) based on an ADC value to come up with an algorithm that never makes
in the left and right hand side of the equation. these mistakes. In general, many NLS algorithms
Knowing now that we actually want to compare exist that will in most cases further outperform
the values of the original signals, can’t we mod- WLLS. In some specifically challenging voxels
ify that equation so it compares stuff that isn’t though, such an algorithm might fail to converge
logarithmically transformed? Easy enough: just or get stuck in the previously mentioned subopti-
remove the logarithm by taking the exponential mal valleys. If and when the algorithm might
of both sides! Now we are facing the correct form detect this, it could for instance perform a WLLS
of the equation, but sadly, it also lost its linearity fit instead (still better than nothing or something
in the unknowns: the linear sum of these really wrong, right?). Because NLS algorithms
unknowns on the left hand side now appears are forced to take a walk in the mountains any-
under that exponential function. Long story short: way, they may also come with extra bells and
it’s a nonlinear equation. That’s where nonlinear whistles allowing them to generate a solution that
least squares (NLS) kicks in. specifically satisfies some constraints. Due to
noise in the data, LLS and even WLLS can some-
times come up with tensors that have one or more
 LS: A Long and Brave Quest
N negative eigenvalues. Of course, this doesn’t
in the Mountains make sense: negative eigenvalues, and thus nega-
tive ADCs, have no physically sensible meaning.
Nonlinear least squares (NLS) will try to solve an An NLS algorithm can be guided to not encoun-
overdetermined system of nonlinear equations, ter such unwanted cases in the first place: barriers
again aiming to minimize the sum of squared can be put up on the landscape in order to simply
residuals of those equations. Going into details deny the NLS hitchhiker access to these forbid-
about this one is nigh impossible: many methods den areas. This all typically does come at an extra
exist. They all share a common thing, though: computational cost, and thus your valuable time.
they take a much, much longer time to reach a Depending on your hardware, the size of the
solution as compared to LLS and WLLS. They dataset and the kind of NLS algorithm (and the
are basically facing the fiendishly difficult bells and whistles it might come with), some of
­problem of finding the lowest point in the lowest these strategies may take anywhere from a few
valley of a mountainous landscape in a six- minutes to several hours to finish calculating
dimensional world. Actually, LLS and WLLS your tensors. Using a brain mask (so no unneces-
also did, but due to the specific simple shape of sary calculations are performed for voxels out-
the landscape when the equations are linear, they side of the brain) is typically strongly advised to
4  From Diffusion to the Diffusion Tensor 61

reduce running time. And guess what? Even if of outliers, enough measurements should of
the hitchhiker would be so extremely experienced course still be left to reliably obtain the final fit.
that he would always find the lowest point in the Those measurements are even needed to actually
landscape, his optimal NLS solution could still reliably classify the other ones as outliers in the
be unsatisfying. That’s because the data aren’t first place. Hence, data redundancy is an impor-
only messed up by noise, but possibly also by tant requirement. Even very recently, further
outliers! Motion, distortions, cardiac pulsation, improvements have still been made to relax that
signal dropout, ghosting … artifacts are abun- redundancy requirement up to a certain extent
dant in MRI. Some can be avoided during acqui- [10]. Given that the DTI model is about 20 years
sition, others can be partially dealt with by old now, this certainly proves that the fitting
preprocessing, but in the end some still leave problem still remains far from trivial.
their mark on the data when we offer it to our
favorite tensor fitting method. They cause outli-
ers: data points that have lost all of their informa- Conclusions
tive value by taking on truly silly values that
don’t fit the picture, at all. That’s where robust We took a bite out some of the most common ten-
estimation of tensors by outlier rejection sor fitting methods: linear least squares (LLS),
(RESTORE) kicks in. weighted linear least squares (WLLS), nonlinear
least squares (NLS), and robust estimation of ten-
sors by outlier rejection (RESTORE). It is typi-
 ESTORE and Beyond: Expecting
R cally said that this specific ordering is one of
the Unexpected increasing complexity, implying increasingly bet-
ter results at the cost of an even steeper increase of
Robust estimation of tensors by outlier rejection computation time (especially for the nonlinear
(RESTORE) [9], as its name suggests, will handle methods). This is generally true; provided that
outliers by rejecting them. To reject them, they each variant is implemented as good as possible
first have to be detected though. To do this, it will (we rely on the responsibility of the software
start with a NLS fit. It will subsequently assign developers here). If your dataset has enough data
each measurement a weight, depending on how redundancy (let’s say, DWIs for more than 30–40
well it fits the picture. Another NLS is performed, unique gradient directions [10]), we could easily
where each equation is weighted according to always advise you to use RESTORE. However,
how well its measurement fit the picture before. depending on the specific implementation of
This process is repeated until convergence. The RESTORE, the hardware, the size or even number
final weights should now be a reliable measure of datasets you have to process, etc. it might take
for how well each measurement does (not) fit in, quite a while (possibly up to several hours) before
i.e., for its “outlier-ness”. Those measurements you have access to your tensors for further pro-
that meet a certain threshold are officially cessing. All the bells and whistles in these
regarded as outliers, and simply kicked out of the advanced nonlinear algorithms may not be neces-
game. The final fit is then performed by employ- sary, if you’re just concerned about having a quick
ing only the surviving “non-outlier” data. While qualitative look at the data. For quantitative pur-
this is an ingenious and very robust strategy, it poses though, we certainly advice to go “beyond
does have a few implications. A first one is the LLS.” A very big gain is already achieved by
fact that it might have to perform several subse- WLLS (if implemented responsibly), at a minimal
quent NLS fits: that will surely have an impact on extra computational cost. Certainly be on the look-
the total computation time. It could on average out for the method your favorite piece of DTI soft-
take more than three times as long as compared ware is packing, or even what different choices it
to a single NLS fit [9]. A second one is the fact might be offering; as you now speak and under-
that, after kicking out a possibly decent amount stand some basic tensor fitting language!
62 T. Dhollander

Final Conclusions make sure that it is effectively packed with your


data. Your next concern then is to get it imported
In this chapter, we provided an overview that correctly into your DTI software package. Unless
took us all the way from the raw DWI data to the that package supports a whole list of different
diffusion tensor and even further to some of the (more and less) standards, you might be up for
more common visualizations and measures. This yet another daunting task. We’re lucky up to
fact by itself makes for the most important con- some extent, however, as the “diffusion commu-
clusion: while the more “classical” imaging nity” and the specific supporting communities
modalities (e.g., T1, T2) are obtained straight revolving around some software packages are
from the scanner, the maps that are typically often very active and responsive: your specific
employed in the practice of DTI (e.g., MD, FA, question could be answered quickly after a sim-
DEC FA) result from a postprocessing pipeline: ple e-mail to a support mailing list. Once you get
i.e. these maps are calculated, not directly your workflow up and running, the use of DTI in
acquired. Most of this pipeline is clearly defined; your daily practice should provide you with new
but for the actual tensor fitting, there are quite a and exciting insights!
few options. The more advanced methods may
also take a reasonable time to be computed. Some
scanner software offers the option to directly References
show and export MD, FA, DEC FA, and even
other maps; however, don’t let that fool you: this 1. Stejskal EO, Tanner JE. Spin diffusion measurements:
software still has to go through all the steps we’ve spin echoes in the presence of a time-dependent field
come across in this chapter. Also, if the software gradient. J Chem Phys. 1965;42:288–92.
2. Le Bihan D, Breton E. Imagerie de diffusion in vivo
almost instantly provides you with e.g. a DEC FA par résonance magnétique nucléaire. C R Acad Sci
map, you should now be aware that it may prob- Paris. 1985;301(Série II):1109–12.
ably not have performed much more than a sim- 3. Le Bihan D, Breton E, Lallemand D, Grenier P,
ple LLS fit (which might of course be sufficient, Cabanis E, Laval-Jeantet M.  MR imaging of intra-
voxel incoherent motions: application to diffusion and
if you’re just qualitatively inspecting the data). perfusion in neurologic disorders. Radiology.
The scanner software also has to rely on the same 1986;161:401–7.
DWI dataset for this, and thus is not any more or 4. Basser PJ, Mattiello J, Le Bihan D. Estimation of the
less reliable in general than any other piece of effective self-diffusion tensor from the NMR spin
echo. J Magn Reson Ser B. 1994;103:247–54.
software if it comes to providing you with accu- 5. Pajevic S, Pierpaoli C. Color schemes to represent the
rate maps: if you do use its features, certainly orientation of anisotropic tissues from diffusion ten-
also try to find out what (tensor fitting) algo- sor data: application to white matter fiber tract map-
rithms it employs under the hood! If you want to ping in the human brain. Magn Reson Med. 1999;42:
526–40.
take advantage of the plethora of different avail- 6. Basser PJ, Mattiello J, Le Bihan D. MR diffusion ten-
able (freeware) software packages that imple- sor spectroscopy and imaging. Biophys J.  1994;66:
ment several advanced tensor fitting methods 259–67.
(and further postprocessing steps, such as fiber 7. Basser PJ. Inferring microstructural features and the
physiological state of tissues from diffusion-weighted
tractography), you’ll need to export the raw DWI images. NMR Biomed. 1995;8:333–44.
data from your scanner. We’ve also stressed at 8. Westin CF, Peled S, Gudbjartsson H, Kikinis R, Jolesz
several occasions that these images are quite FA.  Geometrical diffusion measures for MRI from
worthless if they don’t come with the accompa- tensor basis analysis. Proc Intl Soc Mag Reson Med.
1997;5:1742.
nying gradient directions and b-values. More and 9. Chang LC, Jones DK, Pierpaoli C. RESTORE: robust
more manufacturers are starting to take this into estimation of tensors by outlier rejection. Magn Reson
account and tuck that information safely away in, Med. 2005;53:1088–95.
e.g., the DICOM headers, the headers of their 10. Chang LC, Walker L, Pierpaoli C.  Informed
RESTORE: a method for robust estimation of diffu-
own proprietary formats, or even in separate files sion tensor from low redundancy datasets in the pres-
(containing a gradient table in one way or ence of physiological noise artifacts. Magn Reson
another). However they do it, just try to somehow Med. 2012;68:1654–63.
4  From Diffusion to the Diffusion Tensor 63

Suggested Reading Kingsley PB.  Introduction to diffusion tensor imaging


mathematics: Part II. Anisotropy, diffusion-weighting
factors, and gradient encoding schemes. Concept
Basser PJ, Özarslan E.  Anisotropic diffusion: from the
Magn Reson A. 2006;28:123–54.
apparent diffusion coefficient to the apparent diffusion
Kingsley PB.  Introduction to diffusion tensor imaging
tensor. In: Jones DK, editor. Diffusion MRI: theory,
mathematics: Part III. Tensor calculation, noise, simu-
methods, and applications. New  York, NY: Oxford
lations, and optimization. Concept Magn Reson A.
University Press; 2010.
2006;28:155–79.
Kingsley PB.  Introduction to diffusion tensor imag-
Jones DK, Basser PJ. “Squashing peanuts and smashing
ing  mathematics: Part I.  Tensors, rotations, and
pumpkins”: how noise distorts diffusion-weighted MR
­eigenvectors. Concept Magn Reson A. 2006;28:
101–22. data. Magn Reson Med. 2004;52:979–93.
Quantitative DTI Measures
5
Kathleen M. Curran, Louise Emsell,
and Alexander Leemans

structure should not be performed unless


Learning Points accompanied by a thorough investigation of
their mathematical and geometrical properties.
• Visual assessment of diffusion MR images • Normal brain development and ageing, and
can be supported by nonspecific quantitative the timing and severity of injury/pathology
measures that can be derived from DTI data, should be considered when interpreting DTI
including fractional anisotropy and mean metrics.
diffusivity.
• Variation in image acquisition parameters in
both individual cases and group studies affects  hy Is Quantification Important
W
both qualitative and quantitative analysis. in Medical Imaging?
• Fractional anisotropy can be modulated by
numerous biological and methodological fac- Radiological diagnosis is based almost entirely
tors and should not be blindly interpreted as a on subjective visual evaluation and quantitative
quantitative marker of white matter integrity. image analysis is rarely employed. While routine
• Interpreting changes of the axial and radial dif- clinical diagnosis can be determined qualita-
fusivities on the basis of the underlying tissue tively, image quantification is essential for under-
standing the basic disease mechanisms which
underlie neurological or psychiatric disorders of
K.M. Curran, PhD (*)
the human brain, and for the development of bio-
Complex & Adaptive Systems Laboratory, markers of brain disease which can be used to
School of Medicine & Medical Sciences, evaluate pathology status and treatment efficacy.
University College Dublin, Dublin, Ireland Advanced and automated tools play an essen-
e-mail: kathleen.curran@ucd.ie
tial role in extracting such information about spe-
L. Emsell, PhD cific brain regions or structures within the patient
Translational MRI, Department of Imaging
and Pathology, KU Leuven and Radiology,
population (e.g., hippocampal atrophy). These
University Hospitals Leuven, Leuven, Belgium tools provide objective criteria, quantification,
Universitair Psychiatrisch Centrum (UPC), KU
and a high level of precision (reproducibility),
Leuven, Leuven, Belgium which can inform statistically driven conclusions.
A. Leemans, PhD
Without quantification, group differences cannot
PROVIDI Lab, Image Sciences Institute, University be statistically analyzed, and image-based find-
Medical Center Utrecht, Utrecht, The Netherlands ings cannot be correlated with clinical outcomes.

© Springer Science+Business Media New York 2016 65


W. Van Hecke et al. (eds.), Diffusion Tensor Imaging, DOI 10.1007/978-1-4939-3118-7_5
66 K.M. Curran et al.

Whilst clinicians are well trained in reading which several quantitative measures describing
scalar images, i.e., the grayscale images charac- the amount of diffusion and its orientational pref-
teristic of computed tomography and conven- erence can be derived [4]. While direct visualisa-
tional MRI sequences, they are less familiar with tion of the diffusion tensor components is not
deriving quantitative information from complex readily interpretable, the ellipsoid model is per-
imaging data such as captured using dMRI. As haps the most widely used visual representation
the preceding chapters of the book demonstrate, [4]. An ellipsoid is a three-dimensional represen-
dMRI data provides additional information that tation of the diffusion distance in the X, Y, and Z
requires nontrivial processing before the data can planes by molecules in a given diffusion time. In
be analyzed and interpreted. Not only does it the ellipsoidal representation (Fig. 5.1), the ori-
offer insight into tissue microstructure, but also entation of the axes is provided by the three
the orientational information captured in the eigenvectors e1, e2, and e3 while the radius of the
diffusion-­weighted MR signal can be used to ellipsoid along its axes is proportional to l1 ,
generate impressive and useful qualitative results l2 , and l3 (see Box 5.1).
on the shape of fibre bundles and the connection
patterns of brain regions. However, whilst the ori-
entation information contains important informa-
tion on brain connectivity, it is important to Box 5.1  What Is the Difference Between
remember that it is insufficient for its complete Eigenvalues and Eigenvectors?
characterisation and there are many challenges in Eigenvalues: The ADC values of the ten-
disentangling the true meaning of quantitatively sor along the directions of the eigenvectors.
derived metrics (see Chap. 11 for further details They describe the shape and size of a ten-
about using DTI to study brain connectivity). sor, independently of its orientation.
The reduction of the measured diffusion infor- Eigenvectors: A new set of customized
mation to a diffusion tensor and then to a scalar axes for a tensor, aligned along its specific
value means that when changes or differences are orientation. They describe the orientation
found in one of the scalar metrics, it is difficult to of a tensor, independently of its size and
draw conclusions about the exact cause at a shape.
microstructural level. While this can be consid- Eigenvalues are rotationally invariant,
ered a drawback of dMRI, the systematic infor- whilst eigenvectors are rotationally
mation reduction can also be advantageous. For variant.
example, the human brain is a complex system,
the complete characterization of which is cur-
rently not possible. If we want to characterise its
anatomical status, and compare it with different
populations, we need to find a way to summarise Having obtained the diffusion tensor for each
the complexity in a more simple form. dMRI voxel and having computed the eigenvectors and
offers such a solution as it provides a quantitative the corresponding eigenvalues, several metrics
means of systematically reducing the anatomical can be derived (Fig. 5.2), including standard met-
information into manageable scalar indices. rics such as fractional anisotropy (FA), a measure
of the diffusion anisotropy, and trace or mean dif-
fusivity [6] which reflect the average amount of
 hich Quantitative Measures Can
W water diffusion in a voxel.
Be Calculated from a DTI Dataset? It is important to note that some measures
depend on the orientation of the tissue relative to
Earlier chapters in this section (e.g., 4) describe the applied gradient (rotationally variant), whilst
how the dMRI signal can be used to calculate the others do not (rotationally invariant) as this has
diffusion tensor, the basic building block from implications for interpretation and for image
5  Quantitative DTI Measures 67

Fig. 5.1 Schematic illustration of the relationship eigenvalues and vectors form the basis of quantitative
between the mathematical diffusion tensor and its ellip- DTI parameters. Top left image: Adapted from Beaulieu
soid representation. Decomposition of the tensor into C. The basis of anisotropic water diffusion in the nervous
eigenvectors and eigenvalues provides information on system – a ­technical review. NMR Biomed. 2002 Nov-­
the orientation and amount of diffusion, respectively. Dec;15(7–8):435–55. With permission from John Wiley
Various mathematical formulas as a function of the & Sons, Inc.

r­egistration (see Chap. 10). For example, eigen- Diffusivity Measures


vector orientations are rotationally variant but FA
and trace are rotationally invariant. Apparent  race
T
­diffusion coefficient (ADC) values, on the other The trace (Tr) of the diffusion tensor (D) reflects
hand, are rotationally variant. Recall from Chap. the overall water content. Trace, Tr (D), is a com-
4 that ADC quantifies the magnitude of diffusion monly used clinical measure, which gives an indi-
along a given gradient direction and depends on cation of the overall diffusivity in a given voxel
the strength of the diffusion weighting, i.e., and is computed as the sum of the three eigenval-
b-value. In the case of anisotropic tissue, for ues λ1, λ2, and λ3 or the sum of the diagonal ele-
example, ADC is highest (diffusion is fastest) ments of D (Dxx, Dyy, Dzz). Trace is completely
along the length of the pathway and lower in rotationally independent, and therefore unlike
other directions [7]. with ADC, changes in Tr(D) can be attributed
68 K.M. Curran et al.

solely to changes in tissue structure. Trace has  adial Diffusivity (RD)


R
become an important metric in the assessment and Radial (or transverse or perpendicular) diffusiv-
diagnosis of stroke [1, 8]: ity, l^ , is a measure used to express the diffusiv-
ity perpendicular to the principal direction of
Tr( D) = l1 + l2 + l3 = Dxx + Dyy + Dzz
diffusion:
l2 + l3
 ean Diffusivity (MD)
M l^ =
2
Mean diffusivity characterizes the overall mean
squared displacement of molecules (average
ellipsoid size) and is simply a scaled version of Westin Measures
the trace Tr(D) [9, 10]: In addition to the basic ellipsoidal representa-
tion, Westin et al. [11] proposed a set of geo-
Tr ( D) l1 + l2 + l3 Dxx + Dyy + Dzz
D= = = metrical diffusion measures to quantify the
3 3 3 diffusion ellipsoid’s shape in terms of its linear
MD, sometimes denoted mathematically as D, is a (cl), planar (cp) and spherical (cs) anisotropy
measure of the overall diffusivity in a particular components:
voxel regardless of direction. Just like Tr(D), MD
l1 - l2
is low within white matter but high, for example, in cl =
the ventricles, where the movement of water mol- l1
ecules is unrestricted. This measure of overall dif- l - l3
fusion rate can be used to delineate the area affected cp = 2
l1
by stroke, as demonstrated by van Gelderen [8].
l
 xial Diffusivity (AD)
A cs = 3
l1
Axial (or longitudinal or parallel) diffusivity, l||
is simply the diffusivity along the principal axis cl + cp + cs = 1

of the diffusion ellipsoid and is given by λ1 (see
Box 5.2). The linear component describes how prolate or
cigar shaped the ellipsoid is, the planar compo-
nent describes how oblate or disc shaped it is, and
the spherical component describes how sphere or
Box 5.2: What Is the Difference Between ball-like the ellipsoid is. See Fig. 5.3 for an illus-
ADC, Trace, and MD? trative example.
ADC: amount of diffusion in a single The linear and planar diffusion tensor geome-
direction try indices have been used as criteria to distin-
Trace: sum of the eigenvalues, or “mean guish single-fibre voxels from crossing-fibre
ADC” voxels [12]. To some extent, voxels where the
MD: Trace/3 planar diffusion coefficient (cp) is largest, i.e.,
The ADC depends on diffusion anisotropy larger than linear (cl) and spherical coefficients
and is rotationally variant, whereas trace (cs) can be classified as crossing-fibre configura-
and MD are measures of the average tion voxels; all voxels where linear diffusion is
amount of diffusion in a voxel irrespective largest can be classified as single-fibre configura-
of the gradient direction or underlying tion voxels. A high cs may not only arise from
microstructure. multiple-fibre populations but also from partial
volume effects with CSF [12].
5  Quantitative DTI Measures 69

Fig. 5.2  Example parameter maps derived from diffusion fibre orientation (color) and the degree of anisotropy
imaging. Note the changing contrast in the diffusion-­ (intensity). Abbreviations: B0, non-diffusion-weighted
weighted images (DWIs), reflecting tissue orientation image, DWI, diffusion-weighted image, MD, mean diffu-
changes relative to the applied diffusion gradients. The sivity, AD, axial diffusivity, RD, radial diffusivity, A-P,
color FA map, bottom center, is generated by multiplying anterior-posterior, L-R, left-right, I-S, inferior-superior,
the direction-encoded color map, generated from the prin- DEC, direction-encoding color, FE, first eigenvector, FA,
cipal eigenvector of the diffusion tensor, with the FA map. fractional anisotropy
The colour FA map therefore contains information about

Anisotropy Measures expression based on the eigenvalues of the


­diffusion tensor:
The degree of anisotropy describes how molecu-
3 (l1 - l ) 2 + (l2 - l ) 2 + (l3 - l ) 2
lar displacements vary as a function of orientation FA =
(ellipsoid eccentricity) and is related to the pres- 2 l12 + l22 + l32

ence and coherence of oriented structures [9].
This rotationally invariant, dimensionless mea-
 ractional Anisotropy (FA)
F sure, expresses the anisotropy of the tensor rang-
FA is a metric used to quantify the ratio between ing from 0, when the tensor is completely
the magnitude of the anisotropic component of isotropic to 1 when diffusion is bound to a single
D and the entire magnitude of D, the diffusion axis.
tensor. FA values lie in the range [0, 1] and can In addition to the FA, many other measures of
be calculated in each voxel using the following anisotropy have been proposed, including, but
70 K.M. Curran et al.

Fig. 5.3  Westin measures: Panel (a) illustrates the geo- illustrates how useful additional information can be
metric decomposition of the diffusion tensor, D, (yellow), obtained by combining the measures in a single map. By
into triangular barycentric space, characterised by cigar omitting the spherical component, it is possible to more
(red), disc (green), and ball-shaped (blue) ellipsoids. easily distinguish between linear and planar diffusion
Axial maps of the corresponding Westin measure, cl, cp, (bottom image). Compare to the fractional anisotropy
and cs can be found beneath each ellipsoid. Panel (b) map (top)

not limited to, the relative anisotropy [6], disper- data (i.e., data acquired with one non-zero b-value,
sion of the principal diffusion direction [13] or e.g., b = 1000 mm/s2), reconstructed with the dif-
mode of anisotropy [14]. fusion tensor, which cannot model more than one
Further discussion of how FA relates to tissue orientation per voxel (see Chap. 20). However,
microstructure and other technical features of new multi-shell approaches can be used to gener-
dMRI data are discussed in a later section of this ate alternative metrics, which provide additional
chapter. information about the underlying tissue micro-
structure. These multi-shell approaches are
described in detail in Section VI of this book
 uantitative Parameters Derived
Q “Beyond DTI”; however, a brief summary of rep-
from Multi-Shell dMRI resentative examples is provided below.

Anisotropy present at a microscopic level (for


instance, in the presence of oriented structures, Diffusion Kurtosis Imaging
such as dendrites in the brain cortex) may not
exist at a voxel level due to the averaging effect Diffusion kurtosis imaging (DKI) is a recent MR
over the many different directions present in the technique that employs diffusion-sensitising gra-
voxel. This presents a problem for single-shell dients similar to that used in DTI, but acquires
5  Quantitative DTI Measures 71

three or more diffusion weighting b-values owing to the complexity of analyzing such data
instead of two [15]. While this technique is gen- and because of clinically prohibitive scan times.
erally not used in a clinical environment due to Neurite orientation dispersion and density imag-
time constraints (typical scan times are in the ing (NODDI) has recently been proposed as a
order of 20 min), DKI has been applied to study more clinically feasible alternative and can be
microstructural changes in a number of preclini- used to estimate the density and angular variation
cal and clinical research populations [16–18]. of neurites (dendrites and axons) in-vivo [26].
Whilst DTI is concerned with modeling hindered NODDI is based on a three-compartment tissue
Gaussian diffusion (see Chap. 3), DKI captures model and data is acquired using at least two
information about redistricted diffusion, which shells differing from one another only in choice
can be approximated to water bound by cell of b-values and optimized for clinical (research)
membranes and which is inaccessible to feasibility (scan time <30 min).
DTI. This means that DKI may provide more It should be remembered however that all the
sensitive and specific markers for tissue injury techniques based on tissue models are still lim-
than DTI data alone [19, 20]. Since the acquisi- ited by the simplicity of the model and provide
tion protocol used to obtain a DKI dataset only indirect measures that may relate to tissue
includes all the information necessary to derive a features, but do not actually directly quantify, for
standard DTI dataset, it can be used to calculate example, neurite density, in the same way as a
both types of indices. DKI measures include the histological examination.
kurtosis anisotropy (KA), mean, axial and radial
kurtosis (MK, AK, RK, respectively). These
measures quantify the degree of non-Gaussianity Section Summary
and can be regarded as indices of tissue compart-
mentalization or complexity [15]. For example, a • The diffusion tensor provides a means to
high mean kurtosis may reflect an increase in tis- quantify the amount and orientational prefer-
sue complexity. This is in contrast to high mean ence of diffusion at a voxel level.
diffusivity, which would reflect an increase in • A number of rotationally invariant scalar
freely diffusing water. Further information about parameters can be derived from the diffusion
DKI can be found in Chap. 21. tensor, including the FA, MD, AD, and RD.
• The mean ADC, trace(D), and MD are all
measures of the average diffusion in a voxel.
 issue (Compartment) Model-Based
T They relate to the absence of barriers to water
Approaches diffusion.
• FA is the most widely used DTI measure and
Whilst DTI and DKI do not assume a specific describes the degree of diffusion anisotropy in
biophysical tissue model, other MRI based a voxel. It is related to the presence of barriers
frameworks aim to incorporate additional fea- to diffusion, such as axonal membranes.
tures into their models that reflect some proper-
ties of tissue microstructure such as the behavior
of water in intracellular and extracellular com-  he Influence of Image Acquisition
T
partments [21]. For example, approaches such as on DTI Parameters
CHARMED [22], AxCaliber [23], and ActiveAx
[24] enable the extraction of a multitude of Deriving scalar values from dMRI data and even-
microstructural parameters (axon diameter distri- tually comparing them between groups of sub-
bution, mean axonal diameter, and axonal den- jects and/or correlating them with other
sity) [25]. To date however, these approaches parameters begins with the raw data acquisition,
have been applied primarily in a research context followed by a pipeline of image processing steps.
72 K.M. Curran et al.

Each one of these steps is susceptible to sources reproducibility of measures at different field
of bias, which may not only limit the accuracy strengths is largely dependent on signal-to-noise
and precision of DTI parameter estimation, but ratio and the effect of artifacts [27]. It is commonly
can lead to substantial errors. A more detailed accepted that scanning at higher field strengths
coverage of this topic is performed in the follow- increases signal-to-noise (SNR); therefore one
ing chapters of this section (Chaps. 6 and 7); would expect higher fields to equate with higher
however, a brief summary of selected influential quality. Although this is the case for conventional
factors is presented below. imaging, the competing decreases in T2 time and
increased b0 inhomogeneity associated with
increasing field strength, coupled with increased
Effect of Field Strength distortions due to eddy currents, magnetic suscep-
tibility, and chemical shift artifacts, off-set the gain
Clinical MR systems typically used for routine in image quality in DTI. Nevertheless, it has been
DTI scanning of humans are 1.5 or 3 T, although a shown that the uncertainty of fitted DTI parame-
small number of specialist research centers offer ters decreases with increasing field strength, which
the possibility of scanning (predominantly healthy) may impact positively on fibre-tracking results
subjects at 7 T. As DTI parameters should not be [27]. Figure 5.4 illustrates the effect of field
dependent on the static magnetic field strength, the strength in a single subject.

Fig. 5.4  Comparison of FA measured in two ROIs in the T: FA mean = 0.67 (SD = 0.18) and at 7 T: FA mean = 0.66
same subject at 1.5 and 7 T. Coronal DTI data of the (SD = 0.14). Note the difference in image quality at differ-
human brain in the same subject acquired at 1.5 and 7 ent field strengths, which in this subject has a relatively
T. FA was calculated in the centrum semiovale at 1.5 T: minor effect on the FA when averaged across each
FA mean = 0.48 (SD = 0.11) and 7 T: FA mean = 0.47 ROI. SD = standard deviation [Legend data provided cour-
(SD = 0.08); and in the genu of the corpus callosum at 1.5 tesy of FMRIB Centre, University of Oxford]
5  Quantitative DTI Measures 73

Effect of Number of DWIs be strongly dependent on structural orientation


[30]. For example, the lowest variance in a
Reconstruction of the diffusion tensor requires a parameter such as FA is found when the fibre is
minimum of seven MR images [28], one without aligned with one of the sampling orientations,
any diffusion sensitizing gradients applied and and is largest when the fibre is at the greatest
at least six diffusion-weighted images with angle to the sampling vectors. These variations
gradients applied in non-collinear directions. will effectively increase the standard deviation of
­
Mathematically, only six non-collinear diffusion-­ measurements obtained from a region of interest
weighted directions are necessary to reconstruct (ROI) encompassing voxels containing tissue
the diffusion tensor; however, in practice more with different fibre orientations. Such increased
images should be acquired to improve the accu- variance will reduce the statistical power for
racy of tensor estimation [29]. quantitative analyses of mean diffusivity in dif-
When using the tensor model, the more direc- ferent ROIs and will, in general, spuriously
tions that are acquired the better the angular reso- increase the heterogeneity of the apparent trace
lution that can be achieved; however, research within ROIs (Fig. 5.5).
has shown that the “cost” (in this case scan time) Combined with the number of diffusion
versus benefit curve starts to flatten out at around weighting directions used, there is a requirement
30–32 unique directions. Jones et al. [29] found to acquire multiple non-diffusion-weighted
that robust determination of mean diffusivity, FA images if large numbers of DWIs are acquired
and tensor orientation requires a dMRI sampling using different gradient orientations. The optimal
scheme in which at least 30 unique and evenly ratio of diffusion weighted to non-diffusion-­
distributed sampling orientations are employed. weighted images was calculated to be roughly
However, for anisotropy measurement only, the 9:1 [31]. Typically one b = 0 image is acquired for
measurements will be robust when at least 20 every 8–9 diffusion-weighted images. HARDI
unique sampling orientations are used. When methods typically use a higher number of gradi-
comparing indices derived from DT-MRI, if the ent directions (>45) depending on the reconstruc-
number of sampling orientations is low (<30) and tion method used, and similar to DTI, increasing
not uniformly distributed over the surface of a the number of directions improves the angular
sphere, then the variance in derived indices can precision achievable [32].

Fig. 5.5  Illustrates synthetic color FA maps for (a) 16 (blue), (b) 30 (red), and (c) 60 (green) directions. Increasing the
number of gradient directions increases SNR and therefore the accuracy of estimated the DTI measurements
74 K.M. Curran et al.

Fig. 5.6  Illustration of change in contrast and SNR with increasing b-value [33]

case in an estimated 60–90 % of brain voxels


Effect of  b-Value [34], which again emphasizes the need for cau-
tion when interpreting DTI measures. Several
The number and strength of b-values influence groups have proposed methods for elucidating
the derived measures of diffusion and anisotropy complex tissue microstructure by examining the
[30] (Fig. 5.6). Attention to the choice of diffu- non-Gaussian diffusion behavior that only
sion sensitisation parameters is important when becomes apparent at higher b-values (see [32] for
making decisions regarding clinical feasibility a review).
(acquisition time) and obtaining normative
measures.
Within an ROI, the mean and variance of the Effect of Image Quality
trace will be dependent on the b-value [30]. The
trace of the tensor is dependent on the amount of Noise
diffusion weighting used to characterise it [30].
As the diffusion weighting is increased there is The effect of noise on anisotropy measurements
an increasing dissociation between grey and derived from DT MRI was first described by
white matter, with the trace in white matter being Pierpaoli and Basser [35]. Noise in the diffusion-­
lower than in grey matter. This not only intro- weighted signals will mean that, even in a per-
duces more heterogeneity within ROIs, but also fectly isotropic medium such as a glass of water,
means that comparison with normative databases it is not possible to obtain three identical eigen-
or data from other centres and, by extension, values. Low signal-to-noise ratios correspond to
multicenter studies is problematic unless the high eigenvalue discrepancies. Thus, there is a
same degree of diffusion weighting is employed noise-induced bias in measurements of anisot-
(which is seldom the case). ropy. The variance in anisotropy increases as the
The diffusion characteristics of a voxel con- added noise increases. However, the mean value
taining a single-fibre population can be charac- remains approximately constant in the white mat-
terized by a tensor whose associated ellipsoid is ter, but increases rapidly in the grey matter [30].
prolate. In voxels containing multiple fibre popu- Therefore unless the acquisitions are matched so
lations (e.g., crossing-fibre regions), the diffusion that the SNR in the non-diffusion-weighted
characteristics observed at low b-values can still images is the same and, equally importantly, so
be described by a single tensor but the diffusion that the same number of b = 0 images and
ellipsoid may be less prolate or may become diffusion-­weighted images are acquired, then
spherical or even oblate. In such cases, the tensor comparing anisotropy values across different
model does not adequately reflect the underlying subjects, time points, and centers is particularly
tissue microstructure. Unfortunately, this is the problematic (see Box 5.3).
5  Quantitative DTI Measures 75

that results will be different when using different


Box 5.3: Comparing DTI Measures estimation methods, and therefore, it is important
in Different Studies when comparing measures of diffusion anisot-
It is not trivial to directly compare DTI ropy to establish not only what acquisition was
measures (e.g., FA) derived from data used, but also what tensor estimation routine was
acquired on different scanners with differ- used (see previous Chap. 4 for further discussion
ent acquisition parameters (e.g., b-value). on the topic of tensor estimation strategies).
Example: For a given patient, the FA in SNR of the diffusion-weighted images is also
the corpus callosum is found to be 0.8. In influenced by the diffusion weighting factor or
another study, the FA in the corpus callo- b-value used. Low b-values provide higher SNR
sum in a healthy individual is reported to but at a cost of reduced angular resolution.
be 0.9. Intuitively, one may conclude that Correspondingly, high b-values are better for
the patient has a lower FA than normal. HARDI acquisitions but SNR is significantly
This is incorrect because the FA was calcu- reduced.
lated based on data with different acquisi-
tion parameters, and possibly also using
different tensor estimation and analysis Artifacts
techniques. The correct way to assess if a
patient has a different FA value in the cor- Although general guidelines exist for optimizing
pus callosum compared to a healthy indi- a DTI acquisition protocol in terms of SNR,
vidual would be to do a case–control group b-value, voxel size, diffusion gradient directions,
study with patients and healthy control and cardiac gating [40], large variations in data
subjects scanned with identical scan proto- quality remain as a result of differences in scan-
cols and processed/analyzed in an identical ner hardware, pulse sequences and available scan
manner. times. Tournier et al. [32] and Jones et al. [36]
present excellent reviews on pre-processing
dMRI data and the recommended quality assess-
ment that should be performed. There are a num-
The SNR depends linearly on the voxel vol- ber of artifacts that can be identified in DTI data,
ume. Clinical protocols typically limit voxel including geometric distortions, ghosting, and
sizes to a minimum of about 2 mm3 at 3 Tesla (T) signal dropouts. Detailed information regarding
or 2.5 mm3 at 1.5 T. Reducing the edge length the prevention, recognition, and correction of
from 2 to 1.5 mm would reduce the SNR by more such artifacts is provided in Chaps. 6 and 7 of this
than half [36]. This reduction in SNR can only be book.
compensated for by repeating and averaging the Such artifacts can affect the accuracy of the
measurements, if averaging is performed in the tensor estimation, and by extension, the derived
complex domain and appropriate models of noise DTI parameters. For example, FA values greater
are invoked [36]. The SNR should never be below than one can result from negative eigenvalues in
about 3:1 in any of the DW images in order to the diffusion tensor, which typically occur at the
avoid the problems associated with the rectified interface between the CSF and the surrounding
noise floor [30]. Results reported in [37] and [38] white matter, as artificially low-intensity rims
suggest 10:1 as a safe minimum (Box 5.3). [36]. Correcting for CSF-contamination partial
It has also been found that that even within the volume effects in the structures of interest on a
three popular regression methods (linear least voxel-by-voxel basis prior to drawing inferences
squares, LLS, weighted-linear least squares, about underlying changes in white matter struc-
WLLS, nonlinear least squares, NLLS) that do tures is therefore recommended [41].
not explicitly account for the noise-floor, there Not only the artifacts themselves, but also the
are differential responses [37, 39]. This means strategies employed to correct them may also
76 K.M. Curran et al.

introduce errors. For example, when correcting nal is an open and complicated question and
for motion and eddy-current induced geometric relies on a number of modelling assumptions,
distortions by performing affine registration of which may or may not be correct [36]. The gen-
the diffusion-weighted images to one of the non-­ eral mobility of water molecules depends on bar-
diffusion weighted images, it is important to also riers and obstacles imposed by microstructure,
reorient the encoding vectors with the same rota- e.g., cell membranes, myelin sheaths, and micro-
tion matrix [42]. Neglecting to perform this tubules. Such barriers slow down the diffusing
important step may have minimal impact on sca- particles (“hindered diffusion”) or even impose
lar indices such as FA but it can introduce biases an upper limit on their overall mean-square dis-
of the order of a couple of degrees to estimates of placement (“restricted diffusion”). The distinc-
the principal eigenvector, or peaks in the fibre tion between restriction and hindrance is
orientation distribution (fODF) or diffusion ori- important when interpreting diffusion MR data.
entation distribution (dODF). While the tensor parameters are influenced by
It is also important to correct for any residual both restricted and hindered diffusion, the tensor
eddy currents along the phase-encode direction model assumes Gaussianity and therefore trans-
by modulating the signal intensity back to its cor- lates restricted into hindered diffusion.
rect value by scaling the intensity in proportion to It is important to understand that when there is
the change in the volume of the voxel. Neglecting any component of displacement along the applied
to do this can introduce biases in quantitative gradient axis that this will lead to signal attenua-
metrics and estimates of orientation [30]. tion. In other words, if the gradient is applied
along a given axis, water molecules do not have
to be moving parallel to this axis to cause signal
Section Summary loss. It is only when the displacement is perfectly
perpendicular to the encoding axis that there will
• DTI metrics are influenced by a number of be no contribution to signal loss since it is only at
factors related to data acquisition, including this orientation that there is no component of dis-
the magnetic field and gradient strength, the placement along the encoding axis.
b-value, the number of gradient directions,
and image quality.
• Pre- and post-processing strategies used to  elating DTI Parameters
R
estimate the tensor and correct for artifacts to Neurobiology
will affect the calculation of DTI measures.
Fractional Anisotropy

I nterpreting Quantitative Diffusion Several DTI indices can be derived from the
Measures eigenvalues to quantify the properties of white
matter noninvasively, but the most widely used is
… in most in vivo DTI cases, all that is proven is FA, which is an index of the amount of anisot-
that there is a change in the diffusion parameters of ropy. FA describes the directional coherence of
water in a specific neural region, the interpretation water diffusion in tissue and is generally inter-
of which is merely a plausible hypothesis.
(C. Beaulieu, [43]). preted as a quantitative biomarker of white mat-
ter “integrity.” This is because pathological
Diffusion-weighted MRI measurements studies tend to show a reduction of FA associated
reflect the amount of hindrance and restriction with neurodegenerative processes [44–48] and
experienced by water molecules moving with a developmental studies tend to show an increase
component of displacement along the axis of the of FA through infancy, childhood, and adoles-
applied gradient, averaged over a voxel. Exactly cence [49–53], whilst IQ or improved perfor-
how restriction and hindrance influence the sig- mance in particular cognitive domains often
5  Quantitative DTI Measures 77

fibres. Furthermore, as axon count and myelin


Box 5.4: The Effect of Timing on DTI Metrics: are strongly correlated, it is impossible to dif-
Stroke ferentiate between them when interpreting FA
The acute assessment and monitoring of changes. For this reason, FA should not be
stroke evolution remain the most useful equated with an index of myelination or myelin
and widely adopted clinical applications of damage.
DTI. Within several minutes of stroke onset Large regional differences have been observed
there is a substantial decrease in mean in WM FA measurements. These differences fol-
ADC/MD in ischaemic brain tissue (by low a typical pattern of high FA in the core of
30–50 %). The basic mechanism underly- fibre bundles and low FA in the periphery [58],
ing this decrease remains unclear [1], but although there are exceptions. For example, in
may be due to reduction in extra- and intra- regions of where fibre bundles cross, FA is low.
cellular water mobility, a shift of water Regional anisotropy may arise from differential
from the extracellular to intracellular space, rates in developmental and degenerative trajecto-
an increase in the intracellular diffusion ries for different fibre pathways [51]. For exam-
restriction due to changes in membrane ple the superior longitudinal fasciculus matures
permeability, an increased tortuosity in the at a relatively later stage of development than
extracellular space due to cell swelling [2] other white matter fibre bundles, and an anterior-­
and the consequences of cytotoxic oedema. posterior gradient of FA decline in later adult-
The initial drop in MD pseudo-normalizes hood has been observed [59, 60]. This regional
around a week later, as a result of blood– heterogeneity introduces its own challenges with
brain barrier breakdown, damage to cell regard to both study design and the interpretation
membranes, and vasogenic oedema, and of results, as discussed later in this chapter and in
gradually continues to rise over the follow- Chaps. 8 and 13.
ing months. In contrast, anisotropy briefly
increases in the hyper-acute phase (<7 h)
before decreasing to below pre-lesion lev- Mean Diffusivity
els within 2 or 3 days [3].
Recall that MD is a measure of the overall diffu-
sivity in a particular voxel regardless of direction.
It is highest in areas where water diffuses most
correlates with increased FA [54–56]. However, freely, such as in the ventricles, and is lowest in
equating FA with an index of white matter “integ- areas of high tissue complexity and hence, more
rity” is an oversimplified interpretation because barriers to diffusion, such as in grey matter. MD
FA cannot disentangle the individual microscopic or “mean” ADC is an important measurement
contributions (partly due to the relatively large when assessing the evolution of stroke as
voxel size) [12, 30, 36]. ischemia-­induced changes in tissue water content
FA is influenced by large, oriented macro- can be visualized on DWI and ADC maps before
molecules, organelles and membranes. The they appear on T2-weighted images (see Box
degree of myelination, axon packing, the rela- 5.4). In the adult brain, white matter water con-
tive membrane permeability to water, the inter- tent is lower than that of the grey matter (65 %
nal axonal structure, and the tissue water content versus 85 %); however, the MD values for the
all contribute to tissue anistropy [57]. The two regions are virtually identical [61, 62]. This
degree of anisotropy is often most strongly cor- indicates that white matter is less restrictive to
related with axon count and density [43], whilst water diffusion than grey matter and may be
the degree of myelination correlates with FA, related to the fact that water diffusion parallel to
but does not determine tissue anisotropy, which axons is relatively unrestricted, compared to dif-
has also been demonstrated in non-myelinated fusion perpendicular to axons or in grey matter.
78 K.M. Curran et al.

Brain water content decreases with maturation. eigenvalues of the diffusion tensor without
In the immature brain, the MD of white matter is checking the alignment of the corresponding
almost twice that of the fully myelinated brain eigenvectors with the underlying tissue struc-
due to the large extracellular spaces present in tures, especially when comparing patients with
unmyelinated white matter [63]. During brain healthy controls [68]. The comparison of eigen-
maturation, structures such as cell and axonal values between different subjects or compari-
membranes become more densely packed, and sons of the contralateral side of a tract affected
water molecule mobility becomes increasingly by pathology in the same subject may be mean-
restricted. As white matter develops, changes in ingless because they could represent completely
water diffusion perpendicular to white matter different physical information.
fibres may indicate changes due to premyelin-
ation (change in axonal width) and myelination
[23]. Differences in water content could also Fibre Count
affect the contrast between white and grey matter
in the paediatric brain [64, 65]. Therefore, as with Although not strictly a DTI parameter, the “fibre
FA, it is important to consider the age of the study count” (number of streamlines that pass through
population when interpreting changes in MD. or between given regions of interest) can be
derived from DTI-based tractography analysis
(see Chap. 11). It is sometimes (incorrectly) used
Axial and Radial Diffusivities as a direct measure of connectivity or fibre
­density [36]. For this reason, the use of the term
Recall that axial diffusivity, l|| is simply the dif- “fibre count” has been discouraged and it is pro-
fusivity along the principal axis of the diffusion posed that reporting the number of streamlines is
ellipsoid (λ1), whilst radial diffusivity, l^ is an a safer and unambiguous way of reporting results
average of diffusion along its two minor axes, [36]. Similarly, the number of streamlines pass-
which expresses the amount of diffusivity per- ing through a voxel will be modulated by fea-
pendicular to the principal direction of diffusion, tures of the pathway (curvature, length, width,
or, in single-fibre populations, perpendicular to myelination) and local variations in SNR and
the direction of fibre orientation. Some studies therefore interpreting this measure as fibre den-
have related AD and RD to specific microstruc- sity is problematic. In this context, it may also be
tural features. For example, axial diffusivity has inappropriate to compare the streamline count
been associated with axonal damage, and frag- between white matter structures that have differ-
mentation in particular, whilst radial diffusivity ent shapes [36].
has been associated with axonal density, myelin
integrity, axonal diameter, and fibre coherence
[3, 66].  TI Parameters as Complementary
D
However, it is important to emphasize that the Measures
diffusion direction associated with the axial dif-
fusivity is not always preserved in pathological Measures derived from the diffusion tensor, such
tissue and is not always aligned with the under- as FA, essentially combine the contributions from
lying expected tissue architecture [67]. the different sub-compartments of white matter
Therefore, interpreting changes in axial and into a single metric. Improving the biological
radial diffusivities in terms of underlying bio- specificity of diffusion MRI demands improve-
physical properties, such as myelin and axonal ments in both acquisition and modeling schemes.
density is discouraged, unless accompanied by a Advanced MR methods may provide putative
thorough investigation of their mathematical and cellular markers, such as “axon/neurite density,”
geometrical properties [68]. In this context, it mean axon diameter, axon diameter distributions,
also inappropriate to statistically compare the and neurite dispersion (e.g., CHARMED [22],
5  Quantitative DTI Measures 79

AxCaliber [23], ActiveAx [24], and NODDI tion of abnormalities, are all important factors
[26]). As quantification of myelin via diffusion is when relating DTI metrics to clinical outcome
extremely problematic, other MR contrast mech- measures. This next section summarizes some of
anisms may provide complementary information, these issues.
for example, quantitative magnetization transfer
imaging [69, 70] and multicomponent relaxome-
try (e.g., [71, 72]). Indeed, recent work has dem- Model Limitations
onstrated the utility of this approach in the
assessment of the microstructural basis of T2 As described earlier in this chapter, and revisited
hyperintensities in neurofibromatosis (NF1) [18]. throughout this book, DTI measures are derived
from an oversimplified mathematical representa-
tion of the average diffusion in a given voxel
Section Summary given a number of assumptions, most of which
cannot be satisfied in the “real-world” situation.
• DTI measures can be correlated with micro- Although it is beyond the scope of this
structural features such as axon count and ­introductory text to go into detail on this topic, it
density, myelination, and membrane is useful to consider some of the limitations of
permeability. the tensor model (further discussion can be found
• It is inappropriate to attempt to interpret DTI in Chap. 20).
measures in terms of specific microstructural For example, the tensor model assumes that
features or as a measure of white matter diffusion follows a Gaussian distribution, when
integrity in fact, typical dMRI sequences primarily capture
• Fiber count cannot be equated with fibre or signal from intracellular water, which is restricted
axon density. Reporting the number of stream- and hence follows a non-Gaussian distribution. It
lines is more appropriate. also assumes a single fibre direction in each
• Complementary information from other imag- voxel, but we know that this condition is rarely
ing modalities, advanced dMRI techniques, satisfied in (complex mammalian) neural tissue.
and histology can support the biological inter- Other, perhaps less intuitive assumptions are that
pretation of DTI metrics. the temperature of the diffusing molecules
remains constant and they remain in the same
environment, e.g. there is no exchange between
Challenges of Interpretation intra and extracellular compartments. In reality,
variations in temperature will occur as a function
One of the great accomplishments of DTI is of the thermal conductivity of the examined tis-
reducing complex information into a handful of sue and the proximity of blood vessels, and water
simple, sensitive, useful measures. However, this molecules may move between compartments
oversimplification comes at a price, and that price during the application of the diffusion-sensitizing
is the lack of specificity of DTI metrics and their gradients.
dependence on many methodological and bio- Another important limitation concerns the
logical factors. By extension, this means that ellipsoid representation of the diffusion tensor.
changes in DTI metrics cannot be ascribed to a Recall that the degree of eccentricity of the ellip-
single factor (or biological/pathophysiological soid reflects the degree of anisotropy, i.e., a long,
feature), and this makes the interpretation of thin ellipsoid reflects highly anisotropic diffusion
changes in DTI metrics extremely challenging. (i.e., high FA), and a more spherical ellipsoid
For example, the demographics of subjects and reflects more isotropic tissue and (i.e., low FA).
controls, the timing and severity of injury or However, FA reflects the relative contributions of
pathology, technical factors related to image the axial and radial diffusivities, such that differ-
acquisition and analysis, and the nature and loca- ent combinations of axial or radial diffusivity can
80 K.M. Curran et al.

give rise to the same FA. In this context, by only pathology, as detected with DTI, may change
examining FA, it is not possible to identify the over time and it is therefore important to system-
origin of the observed values. For instance, the atically assess the timing of DTI after injury, par-
prolate ellipsoid with λ1 = 3, λ2 = 1, λ3 = 1 and ticularly in the acute and sub-acute periods
oblate ellipsoid: λ1 = 7, λ2 = 7, λ3 = 1 (arbitrary (between 2 weeks and 1 year) [76] (see Box 5.4).
units) have the same FA value of 2/√11, but have
different AD and RD values [73].
Complex Tissue Architecture
and Crossing Fibres
Biological Confounds
One of the most important confounds in DTI
Demographics analysis is the inability of the tensor model to
correctly characterise diffusion in regions of
Abnormalities in DTI pararmeters are typically complex fibre architecture (i.e., when an image
defined on the basis of comparison with a healthy voxel contains fibre populations with more than
control group because universal thresholds for one dominant orientation, such as in bending or
abnormality have not yet been established. In this interdigitating fibre configurations at the voxel
context, a number of studies have examined DTI level) [32] (Chap. 20). The impact of crossing
changes in healthy controls across the lifespan. fibres on the main DTI metrics is summarized
As described earlier, FA typically increases and below.
MD decreases through childhood and adoles-
cence [74] until the fourth decade when white I mpact of Crossing Fibres on FA
matter volume begins to decrease [51, 75]. The FA, in particular, is strongly affected in areas
However, the lack of standardized DTI acquisi- of complex fibre architecture [35]. FA values are
tion and analysis protocols, means that standard, lower in such areas because there is no single
normative data describing thresholds of normal- dominant diffusion direction and the diffusion
ity across the general population are presently profiles of the different fibre configurations aver-
unavailable. This situation is likely to change in age out. Consider how the shape of the diffusion
the future as large-scale, harmonized multicenter ellipsoid would change in a voxel with more than
studies and data-sharing gain ground. one fibre bundle. This effect is clearly visible on
In addition to age-related effects, other factors a standard mid-coronal FA in the semiovale
may confound DTI findings, including brain vol- region, at the intersection of multiple fibre path-
ume, gender, ethnicity, level of education, hand- ways (Fig. 5.7).
edness, medical comorbidity, alcohol and In neurodegenerative conditions, the deterio-
smoking use, and medication status, to name but ration of one fibre bundle could result in an adja-
a few (see Chap. 13 for further information). cent or functionally related fibre bundle becoming
more dominant, resulting in a paradoxical
increase in FA. In some neuropathological stud-
Timing ies, for example, investigating Wallerian degen-
eration and mild cognitive impairment, higher FA
In addition to age, sex and anthropometrics, values have been observed in patients than in
injury mechanism and the chronicity of injury healthy controls [77, 78].
can greatly influence DTI metrics and it is there-
fore important that these issues are considered I mpact of Crossing Fibres on the Trace
when designing studies and interpreting results. The b-value and the number, orientation, and
Primary injury and secondary injury play differ- trace of individual fibre populations within a
ent roles in the evolution of pathology as a func- voxel affect the trace [48]. In a voxel with two
tion of time post-injury. Microstructural fibre populations, the trace in that voxel is not
5  Quantitative DTI Measures 81

Fig. 5.7  The dark, low-anisotropy region (silver box) given voxel. Compare the more spherical tensor ellipsoid
that is typically visible in the centrum semiovale on coro- glyphs in this region (b), with those obtained from higher
nal FA maps (a) reflects the inability of the tensor to char- order models (in this case, constrained spherical decon-
acterize more than one dominant fibre direction in a volution) (c)

only dependent on the trace values of the under- purely SF-configurations, the MD will be rela-
lying fibre populations but also depends on the tively uniform. Likewise, for areas of tissue
angle of intersection between these two fibre where there is uniformity in the complexity of the
populations. As the angle between the two popu- tissue, the MD may be lower but it will be uni-
lations increases, the trace in a crossing fibre formly lower. However, in regions that contain a
region gradually decreases with respect to trace mixture of SF and crossing fibre configurations
in a region with only a single population, reach- that take different geometrical forms, there will
ing its minimum when the populations are be a larger variation in MD. Consequently, there
orthogonal. The trace therefore depends on the will be a higher variance in such regions, and
configuration of the crossing, i.e., the angle of therefore less statistical power to detect differ-
intersection between populations and the vol- ences in MD.
ume fraction of each of the fibre populations in a
voxel [12]. I mpact of Crossing Fibre
Configurations on AD and RD
I mpact of Crossing Fibres on Mean Wheeler-Kingshott and Cercignani [68] have
Diffusivity demonstrated the challenges of interpreting
There are a number of factors that may influence changes in axial diffusivity (AD) and radial dif-
the estimate of MD, for example the choice of fusivity (RD) in crossing fibre regions. Their
tensor estimation routine or the set of gradient experiments revealed that AD increased when
sampling vectors [79]. Vos et al. [12] demon- the RD of one of the underlying fibre popula-
strate that the MD is lower in complex white mat- tions was increased. Similarly, RD decreased
ter configurations, compared with tissue where when there was a reduction in AD in one of the
there is a single dominant fibre (SF) orientation underlying populations. They propose a frame-
within the voxel. They also show that the magni- work to address some of these issues [80]. Vos
tude of this reduction depends on various factors, et al. [12] have also shown an associated reduc-
including the relative contributions of different tion in one or more of the tensor’s eigenvalues
fibre bundles, microstructural properties, and with lower MD values in regions of complex
acquisition settings such as the b-value. fibre architecture. With two fibre populations in
The dependence of the MD on the tissue a voxel, the diffusivity becomes more planar,
geometry has implications for statistical testing. leading to an underestimation of λ1 and an over-
In regions that are comprised of voxels with estimation of λ2.
82 K.M. Curran et al.

Technical Issues v­ entricles. Vos et al. demonstrated that FA and


MD are modulated by fibre bundle thickness, ori-
Partial Volume Effects entation and curvature as a result of PVE [36]
and recommend accounting for these features in
Spatial resolution is an important consideration DTI analysis.
when assessing anisotropy and the aggregate
range of diffusivities across the tissues compos-
ing the voxel, as it will determine to a large I mpact of CSF Contamination
extent, the influence of partial volume effects on Diffusion Metrics
(PVEs) (Fig. 5.8).
PVE is defined as the intra-voxel heterogene- Given that diffusion in CSF is isotropic and has a
ity of different tissue organizations. For example, mean diffusivity that is approximately four times
within one voxel, a variety of tissue types (grey larger than water in tissue, it is clear that partial
matter, white matter, CSF, vascular tissue) may volume contamination any voxel by CSF will
be present, each with a different type and degree influence DTI parameter measurements [82]. This
of cellular architecture and fluid content. When is particularly problematic at the interfaces of tis-
averaged out over the voxel as part of the tensor sue with CSF-filled spaces [83] and is an impor-
reconstruction, the contribution of all these dif- tant confound in studies of development and
ferent tissue structures will give rise to a single ageing, and in pathological volume change [82].
dominant diffusion direction. The relative contri- CSF-suppression techniques such as FLAIR
bution of each tissue type will determine the have been used to ameliorate CSF contamination
dominant diffusion direction, so for example, a at the point of acquisition [84] but this can pro-
voxel located in the core of a white matter bundle long acquisition time. Jones et al. [36] recom-
will be less influenced by PVE than for example, mend the use of a multicomponent modeling
a voxel at the edge of a bundle, or near the solution [85, 86].

Fig. 5.8  The thickness of


a fibre bundle modulates
the DTI metrics along its
length, with smaller,
thinner bundles being more
susceptible to PVE than
larger, thicker bundles. In
this example, FA is highest
in the middle of a
cross-sectional ROI of the
cingulum compared to at
the bundle periphery. The
scatter plot (left bottom) is
based on data from
Szczepankiewicz et al.
2013 [81]
5  Quantitative DTI Measures 83

In a recent study, Metzler-Baddeley et al. [41] similar across approaches, whilst subtle, more
highlight the importance of correcting for CSF-­ local effects or spurious findings tend to occur
contamination partial volume effects in structures less frequently.
of interest on a voxel-by-voxel basis prior to
drawing inferences about underlying changes in
white matter structures. They found that diffusiv- Section Summary
ity metrics (mean diffusivity, axial and radial dif-
fusivity) were more prone to partial volume CSF • Disentangling the relative contributions of
contamination errors than fractional anisotropy. biophysical, pathological and methodological
After free water elimination (FWE) based voxel- factors to DTI measures is challenging and
by-­voxel partial volume corrections [85], the sig- confounds their interpretation.
nificant positive correlations between age and • The tensor model is an over-simplification,
diffusivity metrics, in particular with axial diffu- which provides useful summary measures in
sivity, disappeared whereas the correlation with single-fibre regions such as the corpus callo-
anisotropy remained. Free water elimination may sum and fornix, but has limited applicability
be a more useful strategy than correcting for in regions of complex microstructure or
whole brain volume, which had little effect in “crossing-fibres.”
removing these spurious correlations [41]. • Investigators should consider biological con-
founds such as subject demographics, the
 he Role of the Analysis Technique
T ­timing and severity of pathology and the loca-
DTI can be used to study brain structure at a tion of DTI changes when interpreting results
voxel, regional or whole-brain level (See Section • Cross-validation of results using different
III). Regional analyses include those in which an analysis techniques can yield useful informa-
a priori region of interest is chosen for study, and tion about the reliability and location of DTI
tractography-based analysis, in which an a priori metric changes.
fibre bundle (or bundles) of interest is selected
for investigation. In both approaches, typically,
average diffusion values such as FA are extracted Chapter Summary
from voxels within the ROIs or tracts for subse-
quent analysis. Whole-brain analyses include Qualitative assessment of diffusion MR images
voxel-based analysis (VBA) (Chap. 10) and his- can be supported by quantitative measures
togram analyses of all the voxels in the brain derived from DTI data. However, such metrics
image or in a white matter mask. are influenced by many biological and method-
Because the different analysis approaches ological factors, and should therefore be inter-
utilize different assumptions and image process- preted with due caution. Fractional anisotropy is
ing strategies, it is possible that different results a summary measure derived from DTI, which
can be obtained from each type of analysis. For describes the directional coherence (anisotropy)
example, in a recent investigation of bipolar dis- of water diffusion within tissue, while mean axial
order endophenotypes, Chaddock et al. [87] and and radial diffusivity may more specifically
Emsell et al. [88] report differences in the extent describe the direction and magnitude of tissue
of regional FA changes and association with water diffusion.
genetic risk in the same dataset. This does not Equating FA with an index of white matter
necessarily mean the results from one type of integrity is an oversimplified interpretation
analysis are correct and the other incorrect. In because FA cannot disentangle individual micro-
fact using multiple analytic strategies is impor- scopic contributions at the voxel level. Similarly,
tant for cross-validation of results. The most the interpretation of measures that are sensitive to
commonly implicated regions are generally the sorting of the eigenvectors or to the effect of
84 K.M. Curran et al.

noise and partial volume, such as the axial and MR imaging of anisotropic water diffusion in cat cen-
tral nervous system. Radiology. 1990;176(2):439–45
radial diffusivities, should be discouraged unless
[Research Support, Non-U.S. Gov’t Research
accompanied by a thorough investigation of their Support, U.S. Gov’t, Non-P.H.S. Research Support,
geometrical properties. U.S. Gov’t, P.H.S.].
It is important to emphasise that comparing 8. van Gelderen P, de Vleeschouwer MHM, DesPres D,
Pekar J, van Zijl P, Moonen CTW. Water diffusion and
data that have been acquired using different
acute stroke. Magn Reson Med. 1994;31(2):154–63.
acquisition parameters may be meaningless. This 9. Basser PJ. New histological and physiological stains
is particularly important in ROIs that contain derived from diffusion-tensor MR images. Ann N Y
complex tissue geometry, which will result in Acad Sci. 1997;820:123–38.
10. Le Bihan D. Diffusion, perfusion and functional mag-
greater variation in diffusion measures leading to
netic resonance imaging. J Mal Vasc. 1995;20(3):
higher variance in ROIs across subjects and 203–14.
therefore less statistical power to detect differ- 11. Westin CF, Maier SE, Mamata H, Nabavi A, Jolesz FA,
ences in diffusion measures. Kikinis R. Processing and visualization for diffusion
tensor MRI. Med Image Anal. 2002;6(2):93–108.
The accuracy and reliability of DTI based
12. Vos SB, Jones DK, Jeurissen B, Viergever MA,

results can be improved by undertaking quality Leemans A. The influence of complex white matter
assurance, appropriate pre- and post-processing architecture on the mean diffusivity in diffusion ten-
to correct for artifacts, and by incorporating sor MRI of the human brain. Neuroimage. 2012;59(3):
2208–16.
PVE-related covariates into statistical analysis.
13. Douaud G, Behrens TE, Poupon C, Cointepas Y,

Jbabdi S, Gaura V, et al. In vivo evidence for the
Acknowledgements  The authors would like to thank Dr. selective subcortical degeneration in Huntington’s
Niall Colgan, University College London for producing disease. Neuroimage. 2009;46(4):958–66.
and analysing the synthetic data using the Camino: Open-­ 14. Ennis DB, Kindlmann G. Orthogonal tensor invariants
source diffusion-MRI reconstruction and processing tool and the analysis of diffusion tensor magnetic reso-
[89] and Dr. Karla Miller and FMRIB Centre, University nance images. Magn Reson Med. 2006;55(1):136–46.
of Oxford for supplying the 1.5 and 7 T data. 15. Jensen JH, Helpern JA. MRI quantification of non-­
Gaussian water diffusion by kurtosis analysis. NMR
Biomed. 2010;23(7):698–710 [Research Support,
N.I.H., Extramural Research Support, Non-U.S. Gov’t
References Review].
16. Van Cauter S, De Keyzer F, Sima DM, Croitor Sava
1. Sotak CH. The role of diffusion tensor imaging in the A, D’Arco F, Veraart J, et al. Integrating diffusion kur-
evaluation of ischemic brain injury - a review. NMR tosis imaging, dynamic susceptibility-weighted
Biomed. 2002;15(7-8):561–9 [Review]. contrast-­enhanced MRI, and short echo time chemical
2. Norris DG, Niendorf T, Leibfritz D. Health and shift imaging for grading gliomas. Neuro Oncol.
infarcted brain tissues studied at short diffusion times: 2014;16:1010.
the origins of apparent restriction and the reduction in 17. Gooijers J, Leemans A, Van Cauter S, Sunaert S,
apparent diffusion coefficient. NMR Biomed. Swinnen SP, Caeyenberghs K. White matter organiza-
1994;7(7):304–10. tion in relation to upper limb motor control in healthy
3. Concha L. A macroscopic view of microstructure: subjects: exploring the added value of diffusion kurto-
using diffusion-weighted images to infer damage, sis imaging. Brain Struct Funct. 2014;219:1627.
repair, and plasticity of white matter. Neuroscience. 18. Billiet T, Maedler B, D’Arco F, Peeters R, Plasschaert
2014;276:14–28. E, Leemans A, et al. Characterizing the microstruc-
4. Basser PJ, Mattiello J, LeBihan D. Estimation of the ture of “unidentified bright objects” in neurofibroma-
effective self-diffusion tensor from the NMR spin tosis type 1: a combined in vivo multi-exponential T2
echo. J Magn Reson B. 1994;103(3):247–54. relaxation and multi-shell diffusion MRI analysis.
5. Beaulieu C, Allen PS. Determinants of anisotropic Neuroimage Clin. 2014;4:649–58.
water diffusion in nerves. Magn Reson Med. 19. Van Cauter S, Veraart J, Sijbers J, Peeters RR,

1994;31(4):394–400 [Research Support, Non-U.S. Himmelreich U, De Keyzer F, et al. Gliomas: diffu-
Gov’t]. sion kurtosis MR imaging in grading. Radiology.
6. Basser PJ, Pierpaoli C. Microstructural and physio- 2012;263(2):492–501.
logical features of tissues elucidated by quantitative- 20. Veraart J, Poot DH, Van Hecke W, Blockx I, Van der
diffusion-­
tensor MRI. J Magn Reson B. 1996; Linden A, Verhoye M, et al. More accurate estimation
111(3):209–19. of diffusion tensor parameters using diffusion
7. Moseley ME, Cohen Y, Kucharczyk J, Mintorovitch J, Kurtosis imaging. Magn Reson Med. 2011;65(1):
Asgari HS, Wendland MF, et al. Diffusion-weighted 138–45 [Research Support, Non-U.S. Gov’t].
5  Quantitative DTI Measures 85

21. Panagiotaki E, Schneider T, Siow B, Hall MG,


35. Pierpaoli C, Basser PJ. Toward a quantitative assess-
Lythgoe MF, Alexander DC. Compartment models of ment of diffusion anisotropy. Magn Reson Med.
the diffusion MR signal in brain white matter: a tax- 1996;36(6):893–906.
onomy and comparison. Neuroimage. 36. Jones DK, Knösche TR, Turner R. White matter

2012;59(3):2241–54 [Comparative Study Research integrity, fiber count, and other fallacies: the do’s and
Support, Non-U.S. Gov’t]. don’ts of diffusion MRI. Neuroimage. 2013;73:
22. Assaf Y, Basser PJ. Composite hindered and restricted 239–54.
model of diffusion (CHARMED) MR imaging of the 37. Jones DK, Basser PJ. “Squashing peanuts and smash-
human brain. Neuroimage. 2005;27(1):48–58. ing pumpkins”: how noise distorts diffusion-weighted
23. Assaf Y, Blumenfeld‐Katzir T, Yovel Y, Basser MR data. Magn Reson Med. 2004;52(5):979–93.
PJ. AxCaliber: a method for measuring axon diameter 38. Descoteaux M, Deriche R, Le Bihan D, Mangin JF,
distribution from diffusion MRI. Magn Reson Med. Poupon C. Diffusion propagator imaging: using
2008;59(6):1347–54. Laplace’s equation and multiple shell acquisitions to
24. Alexander DC, Hubbard PL, Hall MG, Moore EA, reconstruct the diffusion propagator. Inf Process Med
Ptito M, Parker GJM, et al. Orientationally invariant Imaging. 2009;21:1–13.
indices of axon diameter and density from diffusion 39. Veraart J, Sijbers J, Sunaert S, Leemans A, Jeurissen
MRI. Neuroimage. 2010;52(4):1374–89. B. Weighted linear least squares estimation of diffu-
25. Assaf Y, Alexander DC, Jones DK, Bizzi A, Behrens sion MRI parameters: strengths, limitations, and
TEJ, Clark CA, et al. The CONNECT project: com- pitfalls. Neuroimage. 2013;81:335–46 [Research
­
bining macro-and micro-structure. Neuroimage. Support, Non-U.S. Gov’t].
2013;80:273. 40. Jones DK, Leemans A. Diffusion tensor imaging.

26. Zhang H, Schneider T, Wheeler-Kingshott CA,
Methods Mol Biol. 2011;711:127–44.
Alexander DC. NODDI: practical in vivo neurite ori- 41.
Metzler-Baddeley C, O’Sullivan MJ, Bells S,
entation dispersion and density imaging of the human Pasternak O, Jones DK. How and how not to correct
brain. Neuroimage. 2012;61(4):1000–16 [Research for CSF-contamination in diffusion MRI. Neuroimage.
Support, Non-U.S. Gov’t]. 2012;59(2):1394–403.
27. Polders DL, Leemans A, Hendrikse J, Donahue MJ, 42. Leemans A, Jones DK. The B-matrix must be rotated
Luijten PR, Hoogduin JM. Signal to noise ratio and when correcting for subject motion in DTI data. Magn
uncertainty in diffusion tensor imaging at 1.5, 3.0, and Reson Med. 2009;61(6):1336–49.
7.0 Tesla. J Magn Reson Imaging. 43. Beaulieu C. What makes diffusion anisotropic in the
2011;33(6):1456–63. nervous system? In: Jones DK, editor. Diffusion MRI:
28. Basser PJ, Pierpaoli C. A simplified method to mea- theory, methods and applications. New York, NY:
sure the diffusion tensor from seven MR images. Oxford University Press; 2011. p. 92–109.
Magn Reson Med. 1998;39(6):928–34. 44. Assaf Y, Pasternak O. Diffusion tensor imaging

29. Jones DK. The effect of gradient sampling schemes (DTI)-based white matter mapping in brain research:
on measures derived from diffusion tensor MRI: a a review. J Mol Neurosci. 2008;34(1):51–61.
Monte Carlo study. Magn Reson Med. 45. Budde MD, Kim JH, Liang HF, Schmidt RE, Russell
2004;51(4):807–15. JH, Cross AH, et al. Toward accurate diagnosis of
30. Jones DK, Cercignani M. Twenty‐five pitfalls in the white matter pathology using diffusion tensor imag-
analysis of diffusion MRI data. NMR Biomed. ing. Magn Reson Med. 2007;57(4):688–95.
2010;23(7):803–20. 46. Filley CM. White matter and behavioral neurology.
31. Jones DK, Williams SC, Gasston D, Horsfield MA, Ann N Y Acad Sci. 2005;1064:162–83.
Simmons A, Howard R. Isotropic resolution diffusion 47. Horsfield MA, Jones DK. Applications of diffusion-­
tensor imaging with whole brain acquisition in a clini- weighted and diffusion tensor MRI to white matter
cally acceptable time. Hum Brain Mapp. 2002;15(4): diseases – a review. NMR Biomed. 2002;15(7-8):
216–30. 570–7.
32. Tournier JD, Mori S, Leemans A. Diffusion tensor 48. Werring DJ, Clark CA, Barker GJ, Thompson AJ,
imaging and beyond. Magn Reson Med. 2011;65(6): Miller DH. Diffusion tensor imaging of lesions and
1532–56. normal-appearing white matter in multiple sclerosis.
33. Froeling M, Tax CMW, Vos SB, Luijten PR, Leemans Neurology. 1999;52(8):1626–32.
A, editors. MASSIVE: multiple acquisitions for stan- 49. Cascio CJ, Gerig G, Piven J. Diffusion tensor imag-
dardization of structural imaging validation and eval- ing: application to the study of the developing brain.
uation. International Society for Magnetic Resonance J Am Acad Child Adolesc Psychiatry. 2007;46(2):
Imaging in Medicine Annual Scientific Meeting & 213–23.
Exhibition; 2014; Milan, Italy. 50. Hüppi PS, Maier SE, Peled S, Zientara GP, Barnes
34. Jeurissen B, Leemans A, Tournier JD, Jones DK,
PD, Jolesz FA, et al. Microstructural development of
Sijbers J. Investigating the prevalence of complex human newborn cerebral white matter assessed
fiber configurations in white matter tissue with diffu- in vivo by diffusion tensor magnetic resonance imag-
sion magnetic resonance imaging. Hum Brain Mapp. ing. Pediatr Res. 1998;44(4):584–90.
2013;34(11):2747–66.
86 K.M. Curran et al.

51. Lebel C, Walker L, Leemans A, Phillips L, Beaulieu atlas: application to normal neonate development
C. Microstructural maturation of the human brain analysis. Neuroimage. 2011;56(1):8–20.
from childhood to adulthood. Neuroimage. 65. Xu D, Mukherjee P, Barkovich AJ. Pediatric brain
2008;40(3):1044–55. injury: can DTI scalars predict functional outcome?
52. Partridge SC, Mukherjee P, Henry RG, Miller SP, Pediatr Radiol. 2013;43(1):55–9.
Berman JI, Jin H, et al. Diffusion tensor imaging: 66. Song SK, Sun SW, Ramsbottom MJ, Chang C, Russell
serial quantitation of white matter tract maturity in J, Cross AH. Dysmyelination revealed through MRI
premature newborns. Neuroimage. 2004;22(3): as increased radial (but unchanged axial) diffusion of
1302–14. water. Neuroimage. 2002;17(3):1429–36 [Research
53. Wozniak JR, Lim KO. Advances in white matter
Support, Non-U.S. Gov’t Research Support,
imaging: a review of in vivo magnetic resonance U.S. Gov’t, P.H.S.].
methodologies and their applicability to the study of 67. Field AS, Alexander AL, Wu Y-C, Hasan KM, Witwer
development and aging. Neurosci Biobehav Rev. B, Badie B. Diffusion tensor eigenvector directional
2006;30(6):762–74. color imaging patterns in the evaluation of cerebral
54. Mabbott DJ, Noseworthy M, Bouffet E, Laughlin S, white matter tracts altered by tumor. J Magn Reson
Rockel C. White matter growth as a mechanism of Imaging. 2004;20(4):555–62.
cognitive development in children. Neuroimage. 68. Wheeler-Kingshott CAM, Cercignani M. About

2006;33(3):936–46. “axial” and “radial” diffusivities. Magn Reson Med.
55. Nagy Z, Westerberg H, Klingberg T. Maturation of 2009;61(5):1255–60.
white matter is associated with the development of 69. Cercignani M, Alexander DC. Optimal acquisition
cognitive functions during childhood. J Cogn schemes for in vivo quantitative magnetization trans-
Neurosci. 2004;16(7):1227–33. fer MRI. Magn Reson Med. 2006;56(4):803–10.
56. Schmithorst VJ, Wilke M, Dardzinski BJ, Holland 70. Sled JG, Pike GB. Quantitative imaging of magneti-
SK. Cognitive functions correlate with white matter zation transfer exchange and relaxation properties
architecture in a normal pediatric population: a diffu- in vivo using MRI. Magn Reson Med. 2001;46(5):
sion tensor MRI study. Hum Brain Mapp. 923–31.
2005;26(2):139–47. 71. Kolind SH, Deoni SC. Rapid three-dimensional mul-
57. Hüppi PS, Dubois J. Diffusion tensor imaging of
ticomponent relaxation imaging of the cervical spinal
brain development. Semin Fetal Neonatal Med. cord. Magn Reson Med. 2011;65(2):551–6 [Research
2006;11(6):489–97. Support, Non-U.S. Gov’t].
58. Zhai G, Lin W, Wilber KP, Gerig G, Gilmore
72. MacKay A, Laule C, Vavasour I, Bjarnason T, Kolind
JH. Comparisons of Regional White Matter Diffusion S, Madler B. Insights into brain microstructure from
in Healthy Neonates and Adults Performed with a the T2 distribution. Magn Reson Imaging.
3.0-T Head-only MR Imaging Unit. Radiology. 2006;24(4):515–25.
2003;229(3):673–81. 73. Leemans A. Visualisation of diffusion MRI data. In:
59. Sullivan EV, Pfefferbaum A. DTI in aging and age-­ Jones DK, editor. Diffusion MRI: theory, methods and
related neurodegenerative disorders. In: Jones DK, applications. New York, NY: Oxford University
editor. Diffusion MRI: theory, methods and applica- Press; 2011. p. 354–79.
tions. New York, NY: Oxford University Press; 2011. 74. Dubois J, Dehaene-Lambertz G, Perrin M, Mangin
60. Huang H, Zhang J, Wakana S, Zhang W, Ren T,
J-F, Cointepas Y, Duchesnay E, et al. Asynchrony of
Richards LJ, et al. White and gray matter develop- the early maturation of white matter bundles in
ment in human fetal, newborn and pediatric brains. healthy infants: quantitative landmarks revealed non-
Neuroimage. 2006;33(1):27–38 [Research Support, invasively by diffusion tensor imaging. Hum Brain
N.I.H., Extramural]. Mapp. 2008;29(1):14–27.
61.
Maas LC, Mukherjee P, Carballido-Gamio J, 75. Giedd JN, Blumenthal J, Jeffries NO, Castellanos FX,
Veeraraghavan S, Miller SP, Partridge SC, et al. Early Liu H, Zijdenbos A, et al. Brain development during
laminar organization of the human cerebrum demon- childhood and adolescence: a longitudinal MRI study.
strated with diffusion tensor imaging in extremely Nat Neurosci. 1999;2(10):861–3.
premature infants. Neuroimage. 2004;22(3):1134–40. 76.
Hulkower MB, Poliak DB, Rosenbaum SB,
62. Ulug AM, Beauchamp Jr N, Bryan RN, van Zijl
Zimmerman ME, Lipton ML. A decade of DTI in
PC. Absolute quantitation of diffusion constants in traumatic brain injury: 10 years and 100 articles later.
human stroke. Stroke. 1997;28(3):483–90. AJNR Am J Neuroradiol. 2013;34:2064.
63. Engelbrecht V, Scherer A, Rassek M, Witsack HJ, 77. Douaud G, Jbabdi S, Behrens TE, Menke RA, Gass A,
Mödder U. Diffusion-weighted MR imaging in the Monsch AU, et al. DTI measures in crossing-fibre
brain in children: findings in the normal brain and in areas: increased diffusion anisotropy reveals early
the brain with white matter diseases. Radiology. white matter alteration in MCI and mild Alzheimer’s
2002;222(2):410–8. disease. Neuroimage. 2011;55(3):880–90.
64. Oishi K, Mori S, Donohue PK, Ernst T, Anderson L, 78. Pierpaoli C, Barnett A, Pajevic S, Chen R, Penix L,
Buchthal S, et al. Multi-contrast human neonatal brain Virta A, et al. Water diffusion changes in Wallerian
5  Quantitative DTI Measures 87

degeneration and their dependence on white matter 87. Chaddock CA, Barker GJ, Marshall N, Schulze K,
architecture. Neuroimage. 2001;13(6):1174–85. Hall MH, Fern A, et al. White matter microstructural
79. Leemans A, Sijbers J, Verhoye M, Van der Linden A, impairments and genetic liability to familial bipolar I
Van Dyck D. Mathematical framework for simulating disorder. Br J Psychiatry. 2009;194(6):527–34.
diffusion tensor MR neural fiber bundles. Magn 88. Emsell L, Chaddock C, Forde N, Van Hecke W,

Reson Med. 2005;53(4):944–53. Barker GJ, Leemans A, et al. White matter micro-
80. Wheeler-Kingshott CA, Ciccarelli O, Schneider T, structural abnormalities in families multiply affected
Alexander DC, Cercignani M. A new approach to with bipolar I disorder: a diffusion tensor tractogra-
structural integrity assessment based on axial and phy study. Psychol Med. 2013;27:1–12.
radial diffusivities. Funct Neurol. 2012;27(2):85–90 89. Cook PA, Bai Y, Nedjati-Gilani S, Seunarine KK, Hall
[Research Support, Non-U.S. Gov’t]. MG, Parker GJ, et al., editors. Camino: open-­source
81. Szczepankiewicz F, Latt J, Wirestam R, Leemans A, diffusion-MRI reconstruction and processing 2006.
Sundgren P, van Westen D, et al. Variability in Proceedings of the International Society for Magnetic
­diffusion kurtosis imaging: impact on study design, Resonance in Medicine. 14th Scientific Meeting,
statistical power and interpretation. Neuroimage. Seattle, WA, May 2006; p. 2759
2013;76:145–54 [Research Support, Non-U.S.
Gov’t].
82. Vos SB, Jones DK, Viergever MA, Leemans A. Partial
volume effect as a hidden covariate in DTI analyses. Suggested Reading
Neuroimage. 2011;55(4):1566–76.
83. Concha L, Gross DW, Beaulieu C. Diffusion tensor
tractography of the limbic system. AJNR Am 90.Jones DK, Cercignani M. Twenty-five pitfalls in the
J Neuroradiol. 2005;26(9):2267–74. analysis of diffusion MRI data. NMR Biomed.
84. Chou MC, Lin YR, Huang TY, Wang CY, Chung HW, 2010;23(7):803–20.
Juan CJ, et al. FLAIR diffusion-tensor MR tractogra- 91.Jones DK, Knösche TR, Turner R. White matter integ-
phy: comparison of fiber tracking with conventional rity, fibre count, and other fallacies: the do’s and don’ts
imaging. AJNR Am J Neuroradiol. of diffusion MRI. Neuroimage. 2013;73:239–54.
2005;26(3):591–7. 92.Tournier JD, Mori S, Leemans A. Diffusion tensor
85. Pasternak O, Sochen N, Gur Y, Intrator N, Assaf
imaging and beyond. Magn Reson Med.
Y. Free water elimination and mapping from diffusion 2011;65(6):1532–56.
MRI. Magn Reson Med. 2009;62(3):717–30. 93.Wheeler-Kingshott CAM, Cercignani M. About
86. Pierpaoli C, Jones D, editors. Removing CSF con- “axial” and “radial” diffusivities. Magn Reson Med.
tamination in brain DT-MRIs by using a two-­ 2009;61(5):1255–60.
compartment tensor model. Proceedings of the 94.Concha L. A macroscopic view of microstructure:
International Society for Magnetic Resonance in using diffusion-weighted images to infer damage,
Medicine. 12th Scientific meeting ISMRM04; 2004. repair, and plasticity of white matter. Neuroscience.
p. 1215 2014;276:14–28.
Survivor’s Guide to DTI Acquisition
6
Eric Peterson and Roland Bammer

Throughout this chapter, we strive to help the


Learning Points reader to best understand DTI acquisitions and DTI
parameters so that one can select and adjust the
• Common acquisition methods used for DTI acquisition that fits best. However, in order to pro-
• Basic imaging considerations in DTI duce the highest quality results for any kind of dif-
• Standard parameters for DTI acquisitions fusion imaging study, it is important to plan and
• Common EPI artifacts and their cause test the acquisition beforehand because there are
• Appearance of common EPI artifacts often complicated image changes from a single
• How to reduce or avoid most DTI artifacts parameter modification. This planning ensures that
• How to diagnose and correct for DTI artifacts the scan goes smoothly, and that costly and poten-
tially impossible reacquisitions are not required.
This chapter is presented in seven major parts:
Introduction Stejskal-Tanner diffusion encoding, an image
acquisition background, echo planar imaging,
In this chapter, we will discuss acquisition meth- fast spin-echo imaging, motion sensitivity of dif-
ods for DTI. This discussion will focus on Echo fusion imaging, image reconstruction, and pedi-
Planar Imaging (EPI) and touch on Fast Spin- atric imaging considerations.
Echo (FSE) acquisition methods. We will address We hope that the information presented here
situations where these methods are particularly about possible DTI acquisition methods and their
well suited, as well as common problems seen associated pitfalls will help you establish reliable
when using these methods in DTI. Please note acquisitions and diagnose acquisition problems.
that many of the image quality aspects are inde- Our aim is that this will enable you to choose the
pendent of whether DTI or DWI is performed. acquisition method with the greatest chances of
Hence for this chapter, unless specifically men- success and get the best performance out of your
tioned, the terms DWI and DTI can be used MRI scanner.
interchangeably.

Stejskal-Tanner Diffusion Encoding


E. Peterson, PhD (*) • R. Bammer, PhD
Department of Radiology, Stanford University,
Diffusion theory was discussed in detail in a pre-
300 Pasteur Drive, Stanford, CA 94305-5105, USA vious chapter (Chap. 3). For this chapter, it is
e-mail: etpeters@stanford.edu important to keep in mind how water molecules

© Springer Science+Business Media New York 2016 89


W. Van Hecke et al. (eds.), Diffusion Tensor Imaging, DOI 10.1007/978-1-4939-3118-7_6
90 E. Peterson and R. Bammer

behave in different tissues. Regions of the rest of the pulse sequence, and in fact, a non-
brain—such as gray matter—are only slightly diffusion-weighted (b = 0) scan is a part of every
structured at the cellular level, and therefore have diffusion protocol and is simply the diffusion
relatively similar diffusion in all directions. sequence with the diffusion gradient amplitudes
However, white matter, which is predominantly set to zero.
axons and other tissues that are structured at the For our purposes, the diffusion tensor acquisi-
cellular level, often have significantly anisotropic tion can be considered an extension of the
diffusion. That is, the diffusion coefficient that Stejskal-Tanner DWI acquisition. The only dif-
one can observe will differ dependent along ference between DWI and DTI is that a DTI
which direction it is measured. acquisition requires a minimum of six or more
This cellular structure can be uniquely probed DWIs, along with at least one image (and often
with MRI using the pulsed gradient acquisition more) without diffusion encoding (i.e., where the
introduced by Stejskal and Tanner [1] (Fig. 6.1). diffusion-encoding gradients are turned off, often
Here, a long-TE spin-echo sequence is modified called a T2w or b = 0 scan) [2, 3]. The acquisition
by straddling the 180° refocusing RF pulse with a of the set of diffusion images is followed by dif-
pair of strong gradients of equal polarity. To fusion post-processing, which is used to calculate
review from the diffusion chapter, these gradi- the directionality and diffusion coefficient, both
ents, often termed diffusion-encoding gradients of which were covered in more detail in the pre-
or motion-probing gradients, reduce the signal ceding chapter (Chap. 4). For the purposes of this
from randomly moving (diffusing) water while chapter, note that the six or more diffusion direc-
leaving the signal amplitude from stationary or tions required to measure the tensor need to be
coherently flowing water unchanged. It is impor- spread out as much as possible in the 3D sphere
tant to remember that the diffusion information of diffusion (Dx, Dy, Dz). The sphere of diffusion
extracted from this sequence is often termed the refers to the diffusion direction (direction from
Apparent Diffusion Coefficient (ADC) as it is an the center of the sphere) and the strength of the
approximation of the diffusion coefficient that is diffusion-encoding gradients (the distance from
made up from the contribution of individually the center of the sphere) and directions that are
diffusing spins (a spin is the individual signal unit spread evenly around the sphere are mathemati-
in MRI; many spins make up a voxel) within the cally best for the diffusion post-processing and
voxel under investigation and certain sequence therefore produce the best diffusion tensor
parameters (i.e., “shutter speed” of the diffusion results. The minimum of six diffusion directions
measurement). These assumptions allow for an is required because there are six unknown ele-
easy evaluation of the ADC, which closely ments of the diffusion tensor (Dxx, Dyy, Dzz, Dxy,
reflects the actual diffusion in the tissues. Dxz, and Dyz).
Also note that one can measure the ADC only The groups of diffusion directions (and some-
along the direction of the applied diffusion- times multiple diffusion sensitivities, called
encoding gradient. This can be in any direction in b-values) are often called “schemes” or “sets.”
the magnet by using a combination of the gradi- These sets are often chosen individually for spe-
ents in the x-, y-, and z-axes. The conventional cific applications. One example of these is
notation for diffusion direction is the so-called schemes, often termed High Angular Resolution
unit vector (i.e., magnitude of the vector equals Diffusion Imaging (HARDI) acquisitions, with
“1”), in which the tip of the vector points along generally more than 60 diffusion-encoding
the direction one wants to measure diffusion. It is directions [4]. A HARDI gradient scheme allows
important to note that the diffusion section of the for better differentiation of complex white mat-
pulse sequence (i.e., the diffusion amplitudes on ter structures and is often used in white matter
the x, y, and z gradients) is independent from the tract tracing applications. The high spherical
6 Survivor’s Guide to DTI Acquisition 91

Fig. 6.1 This figure shows the readout, phase encode, amount (amplitude or b-value) and direction of the diffu-
slice select, and radiofrequency (Gro, Gpe, Gss, and RF sion sensitivity. The hollow arrows indicate the EPI read-
respectively) of a typical Stejskal-Tanner diffusion pulse out, which in this case is a partial Fourier readout. An EPI
sequence with an EPI readout. The Stejskal-Tanner core readout very efficiently covers k-space and the black RF
(the diffusion gradients labeled with arrows) is the bipolar envelope indicates that the spin echo is designed to occur
gradients, which are separated by a 180° RF pulse. These in the center of k-space (hollow arrow) in the readout
gradients are dotted to show that they change to vary the

resolution allows for observation of directional


diffusion differences in highly complex white Acquisition Background
matter structures such as crossing or fiber bun-
dles with other complex patterns (see Chap. 21 In order to better describe DTI acquisitions, it is
for more details about HARDI techniques). important to understand some of the elements of
However, regardless of the direction scheme MRI data acquisition. In this section, we will dis-
chosen (HARDI or otherwise), the rest of the cuss k-space—the form of the image as it is
acquisition (RF pulses, readout method, etc.) is acquired on the scanner—and complex numbers,
often entirely interchangeable and can be modi- both of which are important to keep in mind
fied to best suit the anatomy and physiology when discussing the distortions and artifacts typi-
being imaged. cal in DTI acquisitions.
To review, there are two elements essential to
Stejskal-Tanner DTI:
K-Space
• Paired diffusion gradients, typically spaced
around a 180° RF pulse. The inherent “image” that is acquired by any
• A set of six or more diffusion gradients well MRI scanner is not the image that is shown on the
spaced around the Dx, Dy, and Dz axes. scanner. When mapping the acquired MR signal
92 E. Peterson and R. Bammer

Fig. 6.2 This figure shows the readout in k-space of three reduction factor every second line is skipped. Partial
bidirectional EPI acquisitions. The different readout Fourier imaging is shown in the third panel. In this case,
direction on odd (arrows going left) and even (arrows k-space lines are skipped at the beginning of the readout
going right) lines is the source of EPI “ghosting.” Parallel rather than every other. The acquisition diagram for EPI is
imaging with a reduction factor of 2 is shown, and for this shown in Fig. 6.1

into k-space, it actually becomes the spatial • A Fourier transform can be used to convert
frequency spectrum of the MR image so that a k-space to an image.
simple Fourier transform (after some processing • The traversal of k-space may change signifi-
not discussed here) will yield the MR image and cantly depending on scan parameters, such as
vice versa. In this chapter, we will call the axes in parallel imaging and partial Fourier
k-space kx and ky for the x- and y-axes respec- acquisitions.
tively. The typical acquisition is a Cartesian—
also called a rectilinear or raster—sampling of
k-space, which is shown in Fig. 6.2. For typical Complex Numbers
MR imaging, all the points in k-space are acquired,
and then a 2D or 3D Fourier transform is applied Each point in k-space, and each point in the
to create the 2D or 3D image itself. More details image itself is a complex number—even though
about the transition from k-space to image it is not often shown in the final results. A com-
space—generally called the reconstruction—will plex number is a number that consists of both real
be discussed in detail in a later chapter. and imaginary parts—often called magnitude and
Another tricky detail with EPI is that the gra- angle or magnitude and phase in MRI. The phase
dient polarity changes for every other line in of the k-space points is important during the
k-space, which leads to the well-known “zig-zag” image generation, and unwanted phase is the
pattern of the EPI readout (Fig. 6.2). While the source of most of the image artifacts in EPI.
ky-axis increases linearly for the entire readout, While the signal magnitude is straightforward
the k-space axis reverses for every other line. for most users to understand because it is simply
Thus, the time axis for every other gradient echo the strength of the MR signal in each voxel, the
needs to be reversed to match the MR data sam- concept of having an image where each voxel is a
ple with its location in k-space. If the timing of complex number is not as easily understood. An
the odd and even gradient echo lines is off, ghost- intuitive way to think about it is that if in general
ing artifacts can occur, a phenomenon that will be the spins in a voxel precess (rotate) slightly faster
discussed later. or slower than normal, by the time the sequence
To review, k-space from an EPI acquisition reads out, the angle that is seen is slightly larger
has three important characteristics: or smaller than normal. An analogy to this is a
clock that is slightly faster or slower than a per-
• K-space is the “image” acquired directly from fect clock. If fast, slow, and perfect clocks are set
the MRI scanner. and started at noon, and checked an hour later,
6 Survivor’s Guide to DTI Acquisition 93

Fig. 6.3 This figure shows


the phase or angle in
regions of an image with
different magnetic fields
over time. This phase is
often removed from the
final images; however this
change in phase over time
is the source of shim and
B0 artifacts. In some cases,
the phase can also affect
the combination of data,
which can result in
cancellation of signal

the minute hand of the perfect clock will be The Echo Planar Imaging
pointing up at 12 again. However, the minute Acquisition
hand of the slow clock may only be pointing at
50, and the fast clock may be pointing at 5. In this Echo Planar Imaging
example, the angle (phase) at the time the clock is
checked (TE or echo time in MRI) is the angle of Echo Planar Imaging (EPI) is the clinical and
the minute hand relative to 12. The phase accrual research standard acquisition for DTI. The stan-
can be slowed down or sped up deliberately by dard EPI acquisition is usually a single-shot
applying a magnetic field gradient or uninten- spin-echo acquisition which consists of a 90°
tionally by eddy currents, B0 inhomogeneities, excitation, 180° refocusing pulse (Fig. 6.1), and a
susceptibility differences, and RF. bidirectional Cartesian readout which linearly
The schematic in Fig. 6.3 shows how phase traverses k-space (Fig. 6.2) in one go. The EPI
angles change over time with respect to a magnetic trajectory uses a small “blip gradient” to inch its
field. In fact, phase accrual is the basis for diffusion way up the ky-axis, which is orthogonal to the
imaging—the use of the phase coupled with the readout. The diffusion-encoding pulses, which
random walk (diffusion) of spins across very small are not an inherent part of an EPI readout, are
distances is the indicator that diffusion occurred. placed on either side of the 180° pulse for
The phase is also used in the image acquisition to Stejskal-Tanner imaging.
help form k-space. Many of the artifacts discussed The number one reason why the EPI readout
in this chapter are the result of incorrect phase in is the most popular option for DTI is that any
k-space or image space due to a wide range of fac- DWI method is notoriously sensitive to motion.
tors including motion, diffusion, B0 inhomogene- This motion sensitivity becomes immediately
ities, readout gradient inconsistencies, and others. apparent if one thinks about the minute molecular
To review, complex numbers are important in motion we are trying to capture with this
EPI-MRI for two reasons: sequence. Any bulk motion while these strong
motion-probing gradients are on would pick up
• All MRI data are complex. substantial phase. Due to the unpredictable nature
• In diffusion processing, the phase is typically of bulk motion, these phase changes would most
discarded at the end of the processing, but it is likely differ for each diffusion preparation—
important to understand because it is the readout pair and compete with regular gradient
source of many image artifacts in EPI. encoding and lead to considerable artifacts.
94 E. Peterson and R. Bammer

Fig. 6.4 This figure shows an example of a non-corrected diffusion images have phase differences, and it is there-
and corrected shot combination in non-diffusion and dif- fore often impossible to combine the shots without induc-
fusion imaging. Note how the non-diffusion imaging ing artifacts before phase correction is applied. This case
shots have nearly identical phase patterns, and therefore was a two-shot acquisition, so the artifact from poor phase
can be combined cleanly—without any noticeable arti- combination largely manifests itself as FOV/2 ghosts,
facts—even without phase correction. However, because which will be discussed later
of the sensitivity of diffusion imaging to motion, the

With single-shot EPI the phase errors do not With the increasing demand for ever thinner
disappear but they are the same for the entire slices, the TR of DTI sequences increases along
k-space data set and do not bother us. DWI with the large number of diffusion-encoding
motion sensitivity becomes a problem again directions or intensities for HARDI, Q-ball or
when we want to average multiple measurements q-space imaging. This creates another challenge
together. If the number of signal averages is for diffusion imaging, which is the lengthening of
larger than one, the typical method to perform the overall scan time. Single-shot EPI is currently
averaging is to directly average the acquired MR the fastest and most time-efficient diffusion imag-
signals as they come in. This standard form of ing technique, as it is capable of acquiring a whole
MR signal averaging does not work for diffusion 2D image in a single excitation and therefore a
images as the individual measurements may inter- full DTI dataset in a very short amount of time
fere destructively (Fig. 6.4). A simple remedy is to (Fig. 6.5). Only recently, a further speed-up has
reconstruct each acquisition separately and aver- emerged through simultaneous multi-band acqui-
age the magnitude images afterwards. However, sition, but at the time of this writing, this tech-
averaging in magnitude mode is a challenge in nique is not yet commercially available and is
itself. As the noise in MR magnitude data is no currently only used in research settings [7].
longer Gaussian distributed (for the nerds amongst In summary, acquiring the entirety of k-space
you: it is a Rice-Nakagami distribution), the in a single excitation confers two major advan-
image becomes hazy in regions with low sig- tages in DTI. The first is simply the scan effi-
nal-to-noise ratio (SNR) as the signal effectively ciency—the speed—of the acquisition. For each
“bounces” off the nonzero mean noise floor. The slice in DTI, a minimum of seven images—and
interested reader is referred to the literature for often many more—must be acquired to fully cal-
methods used to resolve this problem [5, 6]. culate the diffusion tensor parameters. This is a
6 Survivor’s Guide to DTI Acquisition 95

Fig. 6.5 This figure shows


DTI results from a medial
slice in a “typical” EPI
acquisition. It shows the
non-diffusion-weighted
(b = 0), mean diffusion
(b = 1000), mean apparent
diffusion coefficient
(ADC), and the color FA
map. The mean diffusion is
simply the average signal
intensity from all of the
diffusion images acquired.
The mean ADC is
calculated using the
average of the ADC in all
three directions (for those
of you interested, it is the
average of eigenvalues
1–3), and higher values
reflect more diffusion in
those regions. The color FA
indicates the major
unidirectional regions of
diffusion color coded based
on the direction (red is
left-right, blue is superior-
inferior, and green is
anterior-posterior)

significant increase in scan time from a single • The full image does not require complicated
anatomical scan; and by acquiring each image in acquisition and reconstruction tricks to avoid
a single—albeit longer—TR, it allows DTI to image phase cancellation.
remain achievable within a clinical setting. The
second advantage of EPI for DTI is the whole
image coverage means that it does not require a Scan Parameters and Their Effects
complicated reconstruction involving data com-
bination in order to prevent phase cancellation In DTI, several scan parameters are changed from
between acquisitions. Despite these strengths, standard anatomical imaging in order to work best
and the standard use of EPI in DTI, single-shot with the constraints of DTI. A list of the basic
EPI is not without its disadvantages, which will parameters, typical values, and comments about
be discussed in the following sections. them is in Table 6.1. This table is intended as a
To review, EPI is used for DWI and DTI for general guide rather than rules for DTI acquisi-
two major reasons: tions and we hope this will aid your understand-
ing of the parameters and the EPI acquisition.
• It is fast because a single image can be Generally, a DTI acquisition is slower and has
acquired for each excitation. more images than anatomical imaging because of
96 E. Peterson and R. Bammer

Table 6.1 Typical DTI parameters


Parameter Value Comment
Flip angle 90° 90° for the excitation and 180° for the spin echo(es)
to achieve maximum signal
TR >3 s At least 2 × T1 of tissue to allow full signal
recovery—keep large number of slices within
1 TR by simply increasing TR
TE 60–110 ms As short as possible—depends on maximum gradient
strength available for a given b-value as well as
whether or not partial Fourier imaging is used
Partial Fourier Yes 20 overscan lines for 128 × 128
7/8 partial Fourier
70 % partial Fourier
Go higher on this factor to avoid “worm hole”
artifacts. When in doubt use full echo but this
increases TE significantly
Matrix size 128 × 128 96–192 are typical, mostly isotropic in plane
rFOV 100 % Cannot do rFOV because PE is along A/P
Scan % 100 % Keep an isotropic in-plane resolution
FOV 24 × 24 cm Large enough to prevent aliasing
Phase-encode direction A/P To keep distortions L/R symmetric on axial scans
Frequency-encode direction L/R
Parallel imaging factor 2–3 Highly dependent on the RF coil
Some coils afford 4—if reduction factor is too
high the parallel imaging noise enhancement
impairs image quality
# of b = 0 images 1–3 About 1 per 6 diffusion images
# of diffusion directions Approx. 21 6+ required, more than 100 possible
Slice thickness 3 mm 1–6 mm
Slice gap 0 mm To avoid gaps and better fiber tracking—use
interleaved slice acquisition to minimize SNR
penalty from slice cross-talk
b-value 1000 s/mm2 500–5000+
For HARDI acquisitions 2500
For q-space even higher, e.g., 5–8000
Bandwidth Maximum for bandwidth Almost always the maximum (minimum for
Bandwidth per pixel water-fat shift) possible for the system
Water-fat shift Minimum for water-fat shift
Imaging axis Axial/Oblique Axial or oblique close to axial
Lipid/fat suppression Yes SPIR/SPSP/SPAIR/CHESS
Note that this is only intended as a guideline for the most important parameters for standard single-shot EPI sequences.
For all acquisitions there are more parameters to consider, and some sequences may typically have parameters well
outside of these ranges

the time needed to acquire diffusion-weighted sometimes calling for more than 100 diffusion-
images in at least six directions plus at least a sin- weighted images per slice. Multiple diffusion
gle non-diffusion-weighted (T2w or b = 0) image. magnitudes (b-values) can also be acquired in a
It is important to remember that this is the mini- single scan [9], as in Q-space imaging. Using more
mum number of images to compute the diffusion diffusion directions and more b-values can
tensor. Diffusion acquisitions like HARDI [4] or improve the fidelity of the tensor estimation, espe-
Q-Ball [8] require many more diffusion directions, cially in cases where there are multiple structures
6 Survivor’s Guide to DTI Acquisition 97

Fig. 6.6 This figure shows images with different num- the averaging effects of more images. The bottom row
bers of diffusion directions used (top row) as well as dif- shows (in a separate slice) a single diffusion direction
ferent b-values used (bottom row). The top row shows the (applied left-right) with b-values of 500, 1000, and 1500
fractional anisotropy maps (brighter indicates more direc- s/mm. Note that the signal is higher in the b = 500 s/mm
tionality in the tissue) all with a b-value of 1000, calcu- scan, but it is difficult to differentiate between structures.
lated using 6, 14, and 21 directions from left to right. Note Also note that depending on the direction of the fiber tract,
that even though nothing else is changed between the higher b-values tend to emphasize (thin arrow) or de-
acquisitions, the clarity of the images improves as more emphasize (thick arrow) the structures
diffusion directions are added, which is primarily due to

within a voxel, such as two tensor tracts crossing In clinical practice, acquiring more diffusion
or splitting. These methods use more advanced directions rather than repeating diffusion direc-
processing methods beyond the standard eigen- tions is the preferred method of performing aver-
value DTI processing; however a discussion of aging. This is done because it is a robust way to
these methods is beyond the scope of this chapter. both increase the SNR and ensure more than
A side effect of acquiring many diffusion enough diffusion directions were acquired.
directions and b-values is that the many acquisi- Furthermore, as the b-value increases, the
tions act as a form of averaging, which increases SNR in the individual diffusion images decreases
the signal-to-noise-ratio (SNR) of the diffusion (Fig. 6.6). This becomes a trade-off between the
tensor images (Fig. 6.6). This is advantageous contrast between the diffusion-weighted images
because another consideration is that the reduc- and their SNR; the former potentially showing
tion of the MR signal due to diffusion weighting more detail and the latter providing less-noisy
causes the SNR to be generally lower in the images. A simple solution to counteract the lower
diffusion-weighted images when compared to an SNR is to increase the voxel size compared to
anatomical image with similar scan parameters. that used with anatomical imaging.
98 E. Peterson and R. Bammer

Another consideration of increased b-values is should be at least three times the tissue T1-
increased eddy current distortions, which are relaxation time in order to allow near full relax-
covered in detail later in this chapter. In short, ation of the spins. In the brain, this means that the
higher b-values—stronger diffusion gradients— minimum TR is considered to be at least 3 s, and
can cause distortions in the image. These may not preferably more than 5 s. Often, however, with
be obvious by looking at the images, but they multi-slice scans, the time it takes to acquire all
may cause artifacts around the edge of the brain of the slices—and make it fit into one TR—is
in the processed maps. more than the 3, or even 5-s minimum. While a
The first general MRI scan parameter to dis- TR that is too low reduces the SNR and can
cuss is the echo time (TE). With the inclusion of induce T1-weighting into the diffusion images,
the diffusion-encoding gradients, the minimum anything beyond approximately 5 s does not
TE is increased by the duration of the gradients. improve or degrade the images in any way.
While the minimum TE—or very close to the Another important scan parameter is the reso-
minimum—is often used, the minimum TE is lution of the scan. EPI-DTI is typically acquired
often far longer than what is possible without the at a relatively low resolution compared to ana-
diffusion gradients, as in the non-diffusion- tomical imaging protocols that take approxi-
weighted image. The one caveat for the TE is that mately the same amount of time. The low
it should be the same for all diffusion directions resolution is primarily because of the reduced
and weightings, including the non-diffusion- signal in the diffusion-weighted images caused
weighted image, in order to prevent any T2 decay by the diffusion-encoding gradients, as well as
(or differences thereof) from influencing the cal- the time required to encode the seven or more
culation of the diffusion coefficient. A standard diffusion-encoding directions, which are required
TE for DTI is around 80 ms, although it can vary for a sufficient SNR for the tensor calculation.
greatly depending on the scanner’s gradient hard- Typically, the resolution is as high as possible
ware as well as the requested scan and diffusion while still allowing a high enough SNR as is
parameters. required by the application. The balance between
The increased TE is also a source of decreased resolution, SNR, and the number of diffusion-
SNR relative to anatomical MRI. In conjunction encoding directions in general is an open research
with the diffusion-encoding gradients, which question, and even for specific applications it is
also decrease the signal, DTI acquisitions have not easy to determine without specific testing. It
a lower SNR than anatomical images. It is in often depends on the anatomy under exploration,
part because of the low signal that often more as well as the SNR and resolution that the post-
than 15 diffusion-encoding directions and mul- processing methods require. Because of these
tiple T2-weighted images are used in standard complicated trade-offs, it is important to check
clinical DTI. previous protocols and work to verify what is
Note that unlike in anatomical imaging where required of the technique and to confirm that it is
a long TE generates T2-weighted contrast in the possible with your scanner before scanning
image, the ADC images only have diffusion con- patients.
trast. While the individual images in the series To review, there are three major parameters
have T2-weighted contrast from the TE, this con- that are treated differently for DTI than most
trast is removed in the DTI processing because other MRI scans:
all of the diffusion images have the same TE. This
results in images which are unaffected by image • The TE is set as short as possible to get the
contrast beyond that intentionally created by the highest SNR possible because the contrast is
diffusion gradients. created with the diffusion gradients, not the
Because of the increased TE, the minimum TE and TR.
repetition time (TR) is also increased. An impor- • The TR is set to be as short as possible for
tant point to remember for DTI is that the TR speed, but it must stay longer than three times
6 Survivor’s Guide to DTI Acquisition 99

the expected T1 of the tissue—typically more the readout direction. Do note that different
than 3 s. vendors report either bandwidth in kHz (GE),
• The resolution is set as high as possible while bandwidth in Hz per pixel (Siemens), or water-
still allowing for good SNR. There is no equa- fat shift (Philips).
tion for this; it is all determined by testing. This water-fat shift is caused by the different
resonant frequency of fat compared to water. As
mentioned above, the slow read progression in ky
Consequences of the EPI Trajectory causes the chemical shift artifact as in standard
gradient or spin-echo imaging. However, chemi-
This section will discuss the EPI trajectory and cal shift is not limited to chemical species; the
common artifacts, which are often seen in con- same principle applies to any region where off-
junction with DTI-EPI. The EPI trajectory is resonance is present. In EPI images this causes
unique because it samples multiple Cartesian the signal from the off-resonance regions to be
lines in k-space in a single readout. This is very shifted, which—due to the gradual off-resonance
efficient in terms of scan time and has the nice change—looks like stretching or signal pileup in
property for diffusion-weighted imaging that the the image rather than a distinct shift (Fig. 6.7).
whole image can be acquired in a single excita- This can be caused by off-resonance effects such
tion, which helps prevent problems from as a poor shim, or more commonly by air-tissue
diffusion-encoding induced phase errors which interfaces such as those adjacent to the sinuses or
will be discussed later. other cavities in the head, which cause irregular
However, acquiring data in a single, long read- magnetic fields in the neighboring brain tissue.
out enhances several types of artifacts, which can The B0 (main magnetic field) and chemical
pose problems when reading or analyzing the shift artifacts are caused by off-resonance differ-
images. These consist of: ences or natural chemical frequency in different
regions of the image. These artifacts can be
• Chemical shift artifacts in which fat/lipid sig- reduced with careful shimming and fast scans,
nal is also shifted significantly relative to the and fat saturation pulses respectively. In fact,
rest of the image and can cause masking of the because the resonance frequency of fat is not the
true image in places of overlap. same as water, fat is typically shifted a significant
• Distortions due to off-resonance in the object amount when compared to the rest of the image,
from main magnetic field distortions such as often into the brain itself (Fig. 6.8). Lipids have a
B0 inhomogeneities or a bad shim. very low diffusion coefficient and therefore vox-
• Ghosting, which is due to inconsistent phase els that have fat overlaid appear hypointense
encoding in odd and even lines in k-space. (dark) on ADC maps. Similarly, residual lipid
ghost often confounds diffusion anisotropy maps.
The main consequence of a Cartesian EPI Because of this, a fat saturation pulse, or some
readout with the phase-encode lines acquired lin- other method of suppressing the fat signal, is
early from one edge of k-space to the other is that required. Alternatively, one can use “water-only”
the EPI readout is very fast in the readout direc- excitation pulses (i.e., spectral-spatial RF pulses)
tion (kx), but very slow in the phase-encode direc- that do not excite fat protons within a slice.
tion (ky) (Fig. 6.2). The consequence of this Another artifact called “ghosting” is inherent
readout is that artifacts, which in other scan types to EPI in which the k-space readout is performed
manifest in the readout direction, present very in both directions, which is what is shown in Fig.
strongly in the phase-encode direction in EPI. For 6.2. This is the most common type of EPI used in
example, the fat-water shift can be tens of pixels the clinic. Ghosting is caused by differences
in the phase-encode direction in EPI images, between odd and even (readouts progressing to
whereas in normal gradient or spin-echo imag- the right and left respectively) k-space lines.
ing, the shift is typically less than two pixels in These differences manifest as image “ghosts” at a
100 E. Peterson and R. Bammer

Fig. 6.7 These EPI images


show off-resonance
induced artifacts due to an
inhomogeneous magnetic
field. When comparing the
phase encode (PE) up vs.
down, it is clear that the
image is either stretched or
compressed in regions with
off-resonance, such as the
regions indicated by the
gray and white arrows.
Even though this artifact
affects the whole image,
the gray and white arrows
show regions where the
differences between the up
and down PE artifact are
easy to see. The speed at
which the image is
acquired decreases from
the fastest, (a matrix of)
128 × 128, to 128 × 128
without ramp sampling, to
the slowest, 192 × 192.
Because of this reduction
in the sampling speed, the
artifacts increase, which is
easy to see when
comparing regions near the
gray or white arrows for
the three different sampling
speeds. Note that
decreasing the resolution is
not the only way to
increase the sampling
speed and therefore
decrease the off-resonance
artifacts; however it is a
way to modulate the
distortion

fraction of the field of view. For single-shot EPI, as well as scalar images (e.g., b = 0) will be most
with altering odd and even echoes, the ghost is corrupted when there is a substantial signal
shifted by half the FOV. Ghost artifacts like this intensity difference between the original and the
(called FOV/2 ghosts) are often fixed in the aliased voxel (e.g., brain tissue vs. eyeball).
reconstruction, though this is not always the case There are two different causes of ghosting: the
and ghosting may be more prevalent if the imag- phase difference and shift between the even and
ing plane is oblique. Similar to residual fat over- odd echoes in k-space. Both of these result in
laying on the tissue of interest, the diffusion ghosting, but their appearance is different and
tensor information can be confounded by the they are caused by the intercept and slope of the
aliased FOV/2-ghost. Quantitative measurements phase terms, respectively [6]. Figure 6.9 shows
6 Survivor’s Guide to DTI Acquisition 101

Fig. 6.8 Lipid suppression is very important in EPI and shows how the fat signal is shifted and can disrupt regions
DTI imaging in order to remove the unwanted fat signal in the image. The next two images show how both fat
from the diffusion calculations. The first image, in which saturation and spectral-spatial pulses work well to sup-
both fat and water are excited with a normal RF pulse, press the unwanted fat signal

Fig. 6.9 This figure shows how differences between odd the circles above the first row show the phase of the even
and even echoes cause EPI ghosting. The first row shows echo and odd echo in black and gray respectively. The sec-
how the phase difference between the echoes causes vary- ond row shows how shifts between the odd and even echoes
ing intensity ghosts in the image. Note how (from left to cause bands of aliased and un-aliased regions in the image.
right) the 0° (no ghosting) and 18° vary only slightly, but at Note that (from left to right) the number of ghosted bands is
90° the ghost and original have equal intensities, and at equivalent to an increased echo shift in k-space. For all
180° only the aliased image remains visible. The arrows in these images the phase-encode direction is up-down
102 E. Peterson and R. Bammer

Fig. 6.10 These images show EPI ghosting with different However, when parallel imaging is used, the ghost shift
parallel imaging factors. Note that with the exception of changes relative to the parallel imaging factor: FOV/(2R)
(a, e) these images were generated by turning off the where R is the parallel imaging acceleration. The thick
“ghost correction” portion of the reconstruction, ghosting arrows show the top of a “ghosted” brain in the R = 1, 2,
can also arise when the ghost correction is used, but is and 3 images (b–d), which shows how it shifts with the
simply unsuccessful. This is the case in the 192 × 192 changes in the parallel imaging factor. The two images
resolution ghost corrected image (a, white arrow) where for a parallel imaging acceleration of 2 show how differ-
some slight aliasing is still present, which is similar to ent ghosting can look between similar images. The gray
that in (b). The “ghosts” are shifted by a fraction related arrow in (f) shows a vertical band where no correction is
to the parallel imaging used and are caused by inconsis- needed to produce a good image; however in the same
tent even and odd readout lines (see Fig. 6.2). Traditional image, the hollow arrow shows a band where the image
EPI ghosts (also called N/2 or Nyquist ghosts) are shifted is entirely ghosted and the true image (e) is not
halfway around the image like in (b), i.e., FOV/2. represented

different types of ghosting, and disabling ghost • Ghosting, which is caused by differences
correction as seen in Fig. 6.10 shows combina- between back-and-forth readout lines
tions of both echo phase and echo shift ghosts. (Figs. 6.9 and 6.10).
Another variant of magnetic field distortions
is due to eddy currents (see section “Examples
and Mitigation of DTI-EPI Artifacts”) induced in Hardware Limitations
the conducting elements of the magnet by switch-
ing the strong diffusion-encoding gradients and There are many hardware limitations in MRI that
which cause spatiotemporal variations of the are considerations for DTI; some of which are
effective field “seen” by the protons. imposed by engineering challenges, and some by
To review, there are three common types of the human body itself. There are two major limi-
artifacts which can manifest—typically, but not tations when it comes to the performance of the
always—in the phase-encode direction: magnetic field gradients and one for the use of
the radiofrequency (RF) system.
• Chemical (fat/water) shift (Fig. 6.8). For the gradient systems, which are especially
• Magnetic field (susceptibility and shim) dis- stressed during DTI acquisitions due to the
tortions (Fig. 6.7). extremely strong diffusion gradients, the two
6 Survivor’s Guide to DTI Acquisition 103

Table 6.2 Scan parameter and gradient limitations ents switch, the faster the acquisition becomes
Parameter Gradient “Difficulty” and the smaller the geometric distortions become.
↑ Matrix size ↑ The limitations on the slew rate are twofold. The
↑ FOV ↓ first, like the maximum gradient strength, is
↑ B-value ↑ caused by hardware limitations, as it is difficult
↑ Slice thickness ↓ to increase and decrease the power in the gradient
Here we loosely define difficulty as those applications that coils very quickly. The second is peripheral
require higher gradient performance, which includes both nerve stimulation (PNS), which is nerve stimula-
gradient strength and slew rate
tion caused by rapidly changing magnetic fields
around the scan subject. It actually occurs in
major limitations are the maximum gradient body parts that are located near the edges of the
strength and the rate at which the gradient gradient coil, i.e., far away from isocenter, where
changes strength (slew rate). Table 6.2 lists sev- the temporal change in the effective field pro-
eral scan parameters that can affect the gradient duced by the gradient is highest. PNS is typically
system and influence the maximum gradient felt as a muscle twitch, and is normally only an
strength and slew rate in some way. annoyance, but can be painful and should be
In diffusion imaging, the maximum gradient avoided if possible. The easiest way to avoid PNS
strength is often reached during the diffusion sen- is simply to decrease the intensity of the gradient
sitizing gradients. If the gradient strength can be switching. This can be achieved by decreasing
increased, the duration of the diffusion pulses can the resolution or the readout bandwidth. In fact,
be decreased for the same desired b-value. To even changing the orientation of the scan is often
review the diffusion equation from the diffusion used to help alleviate PNS.
chapter, the b-value for a diffusion-weighted The limitation on the radiofrequency (RF)
(single) spin-echo sequence is defined by system is called the Specific Absorption Rate
(γGDiffδ)2(Δ − δ/3) where γ is the gyromagnetic (SAR), which is in effect the heating of the sub-
ratio, GDiff is the strength of the diffusion gradi- ject by the RF excitations. RF excitations are
ents, Δ is the time from the beginning of the first done through the RF coil and are a vital part of
diffusion gradient to the beginning of the second any MRI pulse sequence. SAR limitations can
diffusion gradient, and δ is the duration of the dif- vary based on the regulations at the institution as
fusion gradient. A rule of thumb is that the well as the body part being imaged. This heating
b-value increases quadratically with the diffusion is not enough for the patient to notice or is noticed
gradient strength, GDiff, and with the third power with delay as the heating occurs inside the body
of diffusion time, δ. A shorter diffusion gradient and not from the outside where the people would
duration equates to a shorter TE and thus higher be able to sense it with the thermoreceptors in the
SNR as well as shorter TR and thus a faster scan. skin, which is why it is very important not to
The gradient coils (i.e., their diameter, induc- exceed the tissue heating limit. Luckily, it is a US
tance, and resistance) and amplifiers (i.e., peak Food and Drug Administration (FDA) require-
power and sustained power) often limit the maxi- ment that all clinical scanners in the United States
mum gradient strength in the system, and beyond track the SAR of the pulse sequences and stop the
changing scanners or using a gradient insert, scan if it exceeds the allowed limits. Similar
there is unfortunately no way to change how fast limits are generally used worldwide, and differ-
the gradients can be switched or their maximum ent countries and regions have their own laws
gradient strength. and regulations regarding limits.
The slew rate is the speed at which the gradi- Because the SAR is caused by the RF pulses,
ents can change strength. A high slew rate is very and higher flip angles cause more tissue heating,
advantageous for an EPI readout, which requires any pulse sequence with many 90° and especially
very fast positive and negative switching of the 180° pulses is prone to having a high tissue
gradients to achieve an efficient back-and-forth heating. Unfortunately, once the maximum tissue
trajectory (Figs. 6.1 and 6.2); the faster the gradi- heating is reached, the only option is to wait until
104 E. Peterson and R. Bammer

the tissue cools. Because waiting during an MRI The first artifact to discuss is “ghosting” as
is highly undesirable, it is important to avoid the explored above (Figs. 6.9 and 6.10). The typical
SAR threshold when prescribing the sequence. EPI readout is the back-and-forth type that is
Sequences that may reach the SAR limits are shown in Fig. 6.2. The consequence of inconsis-
FSE—where there are many 180° pulses—as tent data in the odd and even readout lines is
well as some spin-echo EPI sequences with fast shown in Figs. 6.9 and 6.10. Even though the odd
repetition times. It is important to notice that and even readouts are spaced differently with dif-
SAR also goes up quadratically with field ferent parallel imaging accelerations, the result is
strength. So SAR management is very important always similar. In each case, there are shifted
at 3 T and even more so at experimental high- “ghosts” that are seen in the phase-encode direc-
field systems, such as 7 T scanners. tion. This artifact can be complicated, especially
Another component to the SAR limit is the when high parallel imaging values are used; how-
weight of the subject and even the body part being ever it is often corrected in the reconstruction
scanned. Different body parts have different SAR [10]. If the ghosts are not easily corrected, scans
limits as determined by the FDA, and these limits in the axial plane may have less inherent ghosting
are set per body weight (W/kg). Even though DTI than oblique, sagittal, or coronal scans. This has
is typically performed on the head, the weight of to do with the fact that the physical gradient coils
the subject determines the SAR limit for head are all slightly different, especially the z-gradient
scans. Variations in patient weight typically do coil, and therefore demonstrate different timing
not cause problems, but if the sequence is set very errors that in turn cause the data inconsistencies
close to the SAR limit, a very small patient may that cause “ghosts.” When prescribing oblique
cause the sequence to pause or abort. This is eas- scans, a combination of the physical gradients
ily understood when considering that an arbitrary (with different temporal responses) is used to
RF pulse deposits a certain amount of energy—in generate the logical gradient and corrections are
this case heat measured in Watts—in the body. A no longer simple. Alternatively, if ghosts are still
SAR limit is the amount of energy per amount of present, a flyback EPI trajectory can be used.
tissue (W/kg) over time; therefore heavier patients This trajectory samples all readouts in the same
can be scanned with more energetic pulses than direction, but is slower than the back-and-forth
lighter-weight patients. trajectory and may not be available on all
To review, there are three limitations caused systems.
by MRI hardware, which may cause limitations Ghosting may also be caused by motion in
for EPI-DTI. some cases. In these cases, it can be very difficult
to differentiate between the acquisition ghosts
• Slew rate—the speed at which the gradients discussed above and the motion ghosts. However,
can change strength, which is important dur- if other slices and EPI scans are ghost-free, then
ing the readout. it is likely that it is a motion ghost. Also, if there
• Maximum gradient strength, which is reached is additional distortion like signal smearing or
during the diffusion-encoding gradients. blurring, that is a good indication that the ghost-
• SAR—the maximum tissue heating from RF. ing is due to motion.
Another major source of EPI artifacts are off-
resonance distortions that are regional imperfec-
Examples and Mitigation of DTI-EPI tions in the shim that cause distortions in the
Artifacts phase-encode direction. The influences of off-
resonance distortions can be seen in Fig. 6.7
Even though there are a few fundamental types of where opposite phase-encode directions are com-
EPI artifacts, there are many causes of EPI arti- pared. Typical sources for off-resonances are: B0
facts. It is important to be able to recognize the inhomogeneities (i.e., deviation in the static field
major types and be familiar with the causes of provided by the magnet), susceptibility errors
each in order to reduce or remove the artifacts. (i.e., when spins are magnetized differently due
6 Survivor’s Guide to DTI Acquisition 105

Fig. 6.11 This comparison shows the difference between EPI images are caused by the progressively slower speed
an FSE (a) and single-shot EPI at three different resolu- in which k-space is traversed in the phase-encode direc-
tions (b–d). Note how the off-resonance distortions are tion: from 128 to 288. The distortions are caused by the
much greater in the EPI images when compared to the combination of off-resonance distortions and the EPI
FSE image (red outline). The increasing distortions in the readout

to the differential field they produce at interfaces respective systems. These methods can differ not
of matter with different susceptibility, e.g., air only in the effectiveness of the lipid suppression
and tissue), eddy currents (residual magnetic but also in how much time is spent to acquire
fields from the diffusion gradients), and lipids each slice. Here, spectral-spatial pulses are obvi-
(which are not actually off-resonance, but pre- ously much more effective than inversion (SPIR,
cess at a different frequency, which then looks SPAIR, STIR) or saturation/binomial (CHESS,
similar to off-resonance). These distortions can ProSet). Fat suppression is not only important
be from the aforementioned sources, but they are because of the unpleasant visual appearance of
inherently related to the slow sampling speed of fat rings. Since lipids have very low diffusion
EPI in the phase-encode direction. Figure 6.7 coefficients, any residual lipid artifact will affect
highlights these differences by showing the same the ADC in the voxel that contain the fat ghost.
slice, sampled in different directions and at differ- An EPI image artifact type that is uniquely
ent speeds, and Fig. 6.11 shows the distortion of associated with DTI and diffusion imaging is
different sampling speeds when compared to a fast eddy current artifacts. These are due to the strong
spin-echo (FSE) scan. These are the typical distor- diffusion gradients that are used to visualize the
tions in EPI, and these artifacts are often accepted structures in the brain. These extremely strong
in order to acquire DTI data in clinically accept- gradients can cause residual magnetic field gradi-
able times; however these may cause issues when ents on any combination of the gradient axes dur-
trying to quantify the diffusion in the brain [11]. ing the imaging section of the pulse sequence
For the same reason that susceptibility gradi- [12]. Note that eddy current artifacts also belong
ents, main field inhomogeneities, and eddy cur- to the family of off-resonance artifacts. These
rents all cause geometric distortions, the fat residual currents cause transient changes in the
signal is shifted by a significant amount relative magnetic field that lead to scaling, translation, or
to the brain water signal. Because of this, some shearing in the image depending on the axis in
kind of fat suppression should be used for any which the eddy current occurs. An example of
EPI acquisition. This is shown in Fig. 6.8, which this can be seen in Fig. 6.12, which shows the
illustrates how the fat signal is shifted and over- differences between images with eddy currents in
laps the brain. However, when a fat saturation or three different directions. These distortions gen-
water selective excitation is used, the fat signal is erally increase with increasing diffusion gradient
removed. There are various methods that can be strength and therefore are not significant under
used, and each vendor has their preferred method, typical (non-diffusion) imaging conditions.
which often has the best performance on their A dual spin-echo acquisition, which will be
106 E. Peterson and R. Bammer

Fig. 6.12 The switching of strong diffusion-encoding respectively. As you can see from the outline, there are
gradients leads to eddy currents and associated field per- many smaller differences in the outlines between the
turbations, which in turn can cause different distortions images as well. Eddy current effects can be reduced by
based on the direction of the diffusion-encoding gradi- using a dual spin-echo imaging sequence, reducing the
ents. This is commonly called “eddy current” distortion, strength or slew rate of the diffusion gradients, or by sim-
and is a concern in diffusion-weighted EPI. These images ply increasing the speed at which k-space is traversed
show the distortion caused by gradients along the x-, y-, along the phase-encode direction (e.g., via parallel imag-
and z-axes, which are shear, scaling, and translation, ing, interleaved EPI, or ramp sampling)

discussed later, can be used to mitigate these dis- seen in Fig. 6.13. Typical clinical scanners have
tortions at the cost of slightly reduced SNR. robust auto-shim procedures, but in some cases
Other distortion effects, in the same family the shim may be determined incorrectly, and in
as off-resonance distortions, are caused if the these cases it is possible to re-run the auto-shim
shim is set incorrectly. The difference between or manually shim the volume.
local susceptibility artifacts and distortions A further aspect to the shim and fat-induced
caused by incorrect shim values are that shim- artifacts is that when the off-resonance distor-
related artifacts generally affect the whole tions are severe, fat suppression can cause the
image and are preventable with careful shim- suppression of nonfat signal because the
ming, whereas susceptibility-related artifacts off-resonance distortions cause the frequency of
are local and typically manifest around air-tis- the brain to shift to frequencies near fat. It is also
sue interfaces. clear that in these regions the image is greatly
In fact, the distortions from poor shim values distorted due to the off-resonance distortions.
look like eddy current distortions where the This is shown in Fig. 6.14, which demonstrates
images are stretched, compressed, and sheared; unsaturated and saturated fat in the case of severe
the cause is off-resonance spins due to bad shim off-resonance distortions.
settings. Even though the shims on modern MR To review, there are three major types of arti-
systems are typically very good, in some cases facts that are caused by off-resonance effects in
the available shim gradients may be insufficient EPI images:
to properly account for the major inhomogene-
ities in the imaging volume. Most clinical scan- • Shim and local magnetic field off-resonance
ners only have linear, or at most first-order (Figs. 6.11 and 6.13).
nonlinear shims, so any field changes that are not • Eddy current distortions caused by the diffu-
corrected by those are uncorrectable and the sion gradients (Fig. 6.12).
remaining field changes result in the distortions • Fat suppression errors (Fig. 6.14).
6 Survivor’s Guide to DTI Acquisition 107

Fig. 6.13 This figure


shows how the shim
quality can distort and
change the image. In the
left column, the small red
and large blue arrows
indicate regions that
change significantly inside
the brain due to the shim
changes, with changing
distortion as the shim
progresses from top to
bottom. In the right
column, the red boundary
shows how the brain shape
is changed by the shim.
Also note how the SNR of
the images decreases as the
shim worsens. To fix this,
the shim values may be
manually changed, and
there may also be higher
order shims, which can
help smooth the magnetic
field over larger regions. In
this example, only the
linear shims were changed.
If higher order shims were
used, the distortions would
become increasingly
complex

Considerations When Prescribing EPI Checklist 1 is a generic outline for trouble-


shooting DTI. We intend it to help troubleshoot
There are a number of factors to pay attention to the potential major problems in a clinical set-
when prescribing an EPI scan. Many of them are ting. It is written generically and may not apply
similar in non-EPI imaging, but often their effects well to all applications. We encourage you to
are amplified or altered in some way in EPI. The use this as a basis for an “artifact checklist” that
previous section described these artifacts and this applies to your scans that you can use to quickly
section discusses how to best avoid these issues and efficiently diagnose and fix common DTI
when prescribing scans. errors.
108 E. Peterson and R. Bammer

Fig. 6.14 This image


shows a brain with large
off-resonance
inhomogeneities with
unsuppressed (a, b) and
suppressed fat (c, d)
images. In this case,
because of the off-
resonance inhomogeneities,
the fat suppression
removes signal from the
center of the brain because
its resonance frequency
was too far away from the
base water frequency

If the scan is artifacted: • Try looking at the line width of the


spectrum from a manual prescan to
1. Does it look like motion? Is there ghosting, evaluate the shim.
signal variation, or blurring? • A manual or a local automatic shim
(a) Did the person move? Ask them, or a sim- may help.
ple test for large motion is to re-run the • Use parallel imaging or multi-shot
localizer to check for major motion. Ask regime.
them to hold still and re-run the scan. (c) If the non-diffusion images are not dis-
Alternatively, use better head restraints or torted, but the diffusion images are dis-
turn on motion correction, if available. torted—typically differently from image to
2. Is the image geometrically distorted image—it may be caused by eddy currents.
(squeezed, stretched)? • Try a dual spin-echo acquisition, or a
(a) The shim may be bad, or eddy currents post-processing eddy current correc-
may be distorting the diffusion images. tion. You could also reduce the b-value
(b) If the localizer looks okay, but if both the as eddy currents increase dispropor-
diffusion images and the non-diffusion tionally as you increase the b-value.
images look distorted, it could be a bad • A lower b-value will reduce the distor-
shim. tions, but it is often not possible to
6 Survivor’s Guide to DTI Acquisition 109

change the b-value for typical DTI choose phase-encode direction to be


protocols. Alternatively, try to increase along A/P and readout along L/R.
TE and lower the diffusion gradient 4. Are there “worm”-like artifacts or fluctuating
strength. signal near the ventricles or even the whole
• Use parallel imaging or multi-shot brain?
regimes. (a) The partial Fourier fraction could be too
• Some scanners allow the combination low.
of two gradient coils to increase maxi- • Try a larger partial Fourier setting in
mum gradient strength at the cost of your scan or avoid it entirely.
lower gradient slew rate and hence 5. Is there ghosting?
slower k-space progression. If you (a) Try changing the orientation to be axial, if
desire to have less geometric distor- possible. Also, if possible, a unidirec-
tion, try to avoid these enhanced modes tional (flyback) EPI readout would remove
and go for the higher slew rate. The the ghosting at the expense of increased
lack of gradient strengths can increase off-resonance artifacts, i.e., distortions
TE but can be mitigated by extra etc., because of the slower k-space
averages. traversal.
3. Is the image distorted in local regions or 6. Do the individual images look good, but the
asymmetrically? tensor does not?
(a) Is/are these distortion(s) near the sinuses (a) It could be caused by motion, eddy cur-
e.g., the nose/ears? rents, or low SNR.
• These are common off-resonance dis- • Try to determine if motion occurred by
tortions near the air/tissue interfaces. looking at the images and asking the
Increasing the bandwidth, increasing the par- subject.
allel imaging factor, or reducing the read- • Try a double spin-echo scan or increase
out length will reduce the distortions. the bandwidth along the phase-encode
Multi-shot EPI also often has reduced distor- direction (by parallel imaging, ramp
tions from off-resonance, so even though sampling, or smaller matrix) to reduce
this will lengthen the scan time, it may be the time it takes to read one line of
possible to use it to minimize the k-space and thus reduce eddy current
distortions. artifacts.
(b) If the distortions is/are exceptionally large • It could be the SNR is too low or there
and potentially located in a place that are too few diffusion or non-diffusion-
does not often suffer from large off- weighted images.
resonance distortions, it could be from 7. Is there a distinct ring, or part of a ring, in the
metal in or near the brain. images that may overlap the brain in places?
• Metal can cause extremely large dis- (a) It is likely that the fat suppression method
tortions and a fast spin-echo sequence is turned off or failed. Verify that some
may be required if the methods listed type of fat suppression is on. If it is on and
above for air/tissue interfaces do not failed, this is often due to poor shimming.
work. (b) If possible, use a spectral-spatial pulse,
(c) If the distortions in a brain scan are asym- but test all the available methods as some
metric and oriented in the left/right direc- may work better than others on your
tion, you probably have chosen L/R as system.
phase-encode direction (and A/P as read- (c) If you are using a thin slice, try to increase
out direction). These are the orientations your slice thickness until you can use
for normal brain imaging but for EPI to spectral-spatial water-only excitation
keep distortions symmetric you want to pulses. Spectral-spatial excitation pulses
110 E. Peterson and R. Bammer

are not available for extremely thin slices, (a) Check the b-values, about 1000 s/mm2 are
so if residual fat is visible it may be neces- normal for clinical imaging.
sary to sacrifice slice thickness for (b) For HARDI acquisitions b-values above
improved fat suppression. 2000 s/mm2 are recommended. To get
(d) If possible, change the polarity of slice reasonable SNR back down on your voxel
select gradient for your excitation pulse size, i.e., use a larger FOV, smaller matrix
and the refocusing pulse to improve fat size, and thicker slices.
suppression. Only spins that are on-
resonance will see both the 90° and 180° Checklist 1: How to diagnose common DTI
pulses and show up in the image. artifacts and SNR issues. Use this as a basis for
(e) Band-like artifacts could also be residual troubleshooting DTI artifacts and lack of
aliasing from a poorly calibrated SENSE SNR. Each system and application has their own
or ASSET scan. Particularly at high field, sources of artifacts, so this is only a template con-
there can be a large mismatch between taining common artifacts. This is intended as an
undistorted coil sensitivity calibration example checklist, and you are encouraged to use
scans and the distorted (stretched or com- this as a basis for your own checklist that better
pressed) EPI scans. Try to use calibration suits your acquisition.
scans that have similar levels of distor- The speed of the readout—the bandwidth—
tions as the EPI scan. can be changed in order to speed up or slow down
the acquisition. An increased bandwidth results
If the scan has a low SNR: in a faster scan and reduces off-resonance arti-
facts, but it may alter—positively or negatively—
1. Check the receiver coil eddy current artifacts and ghosting. The
(a) You could have picked the body coil bandwidth has a dual effect on SNR: with a
rather than the head coil. higher bandwidth noise is increased, but the TE is
(b) Some of your coil elements might be shorter. For EPI, generally the best results are
defective. Try to reconstruct them when the bandwidth is close to the maximum so
individually. that the distortions are kept minimal and the TE is
2. Check the parallel imaging value short.
(a) It could be too high if regions at the center As previously discussed, in EPI the phase-
of the image look noisier than the periph- encode direction is the direction in which most of
ery or if this noisy region repeats itself as the artifacts manifest. Therefore, it is important
a fraction of the FOV. to consider how the artifact may affect the image,
3. Check the resolution and slice thickness and which phase-encode direction would be best
(a) The FOV/matrix gives the resolution, and to mitigate the expected artifacts. Typically, for
if this is smaller than 4 mm3 the resolution brain DTI the slices are prescribed axially or
may be too high for DTI. obliquely (although close to axial), and the PE
(b) Try not to be too aggressive and attempt direction is anterior-posterior or another direc-
imaging slices that are too thin. SNR is tion that causes symmetric artifacts in both hemi-
directly proportional to slice thickness. spheres of the brain (Fig. 6.15).
Too aggressive reduction can have signifi- Even though the artifacts predominantly
cant SNR loss as a result. appear in the in-plane direction, reformatting the
4. If the image is also distorted scans can effectively mask the distortions.
(a) It is very likely the shim is bad; try auto- Figure 6.16 shows how multi-slice axially
matically or manually re-shimming. acquired images can be displayed in other axes.
5. If the images look good but the diffusion ten- The distortions are still present in this case, but
sors are noisy this is a good way to view axial and sagittal
6 Survivor’s Guide to DTI Acquisition 111

Fig. 6.15 This figure shows how the phase-encode direc- visually pleasant distortions, where the structures near the
tion affects off-resonance artifacts in the brain. The thick, dashed, thick, and thin arrows are more symmetric when
gray, hollow, and thin arrows show how the distortions the phase-encode direction is up/down rather than left/
change significantly when changing the phase-encode right. The wedge indicates the residual ghosting and
direction in various slices. Even though the distortions are shows how slight ghosting is present in the phase-encode
often in the periphery of the brain, this is not always the direction in all cases. Important Note: For technologists it
case. The thin arrows indicate clear distortions in the mid- is common practice for brain scans to choose left/right as
dle of the brain, which are especially important because the phase-encode direction. However, for EPI the phase-
these distortions can make it difficult for radiologists to encode direction should almost always be anterior/poste-
read the scans. Because of the asymmetry caused by the rior or frequency-encode left/right. Choosing the wrong
left/right phase-encode direction, the phase-encode direc- phase-encode direction is the most frequent technologist
tion is normally chosen to be up/down, which causes the error one encounters with EPI scanning
distortions to be symmetric in the brain. This creates more

images rather than acquiring EPI data in these Magnetic field inhomogeneities greatly affect
axes. While it is possible, the artifacts and distor- EPI acquisitions, and in the brain the sinuses
tions are generally less significant in the axial cause significant distortions. Additionally, if
plane rather than the coronal, sagittal, or oblique there is any metal (e.g., surgical clips) close to
planes. the imaging area there can be significant distor-
In some cases the gradient performance, par- tions and signal dropout close to the object as
ticularly in the z-axis, may be significantly differ- well as distortions away from the object
ent from the other axes. This can induce stronger (Fig. 6.17). Unfortunately, it is difficult to avoid
than normal ghosting, and because of this these artifacts entirely, but increasing the sam-
oblique—or even non-axial—imaging may be pling speed, multi-shot methods (e.g., interleave
discouraged on some systems. However, this is EPI, readout-segmented EPI), or parallel imaging
highly system dependent, and as the gradients are can help reduce them.
improving in more modern scanners, this is However, increasing the speed is not always
increasingly less problematic. possible because of peripheral nerve stimulation
112 E. Peterson and R. Bammer

Fig. 6.16 When scanning in an axial orientation, it is that when one performs multi-planar reformation with the
possible to view multi-slice images in the coronal and slice direction chosen in the phase-encode direction of the
sagittal axes. In this case, the distortions remain primarily acquisition, the reformation algorithm usually uses a slice
in the anterior/posterior direction, so the reformatted thickness of several millimeters and thus reduces the
images are relatively clear of distortions. The arrow visual effect of the distortions by moving the most obvi-
shows a distortion in the axial plane, and in the sagittal ous distortions into the reformatted slice direction
reformat, the arrow shows the same artifact. The trick is

Fig. 6.17 This figure shows common sources of distor- base of the brain (c) such as the ear canals. Because of the
tion in EPI, such as distortions near the base of the brain large image distortion in (c), it is clearly important to
(a), sinus (b), and metal (c). The effects of each are simi- know if the subject has any metal implants in or near their
lar, but the intensity of the artifacts is very different. Note head. Even metal implants that are MR safe—such as den-
how the metal artifact (c) is much more potent than the tal fillings or implants—can cause distortions in large
sinuses (b) and even larger than the similar artifacts in the regions of the brain

(PNS) limitations. PNS is caused by fast gradient FOV can help reduce the effects of gradient
switching, which is essential for EPI, especially switching; however those have the side effects of
as the scan speed is increased. Even though the increased distortion and lower resolution respec-
scanner has a limited slew rate, it is easy to cause tively. It is important to remember that PNS is
PNS in some subjects with EPI-DTI. Simply patient-specific, so scans which cause no PNS in
slowing down the sampling rate or increasing the some people can be uncomfortable for others.
6 Survivor’s Guide to DTI Acquisition 113

At times it may be necessary to abort a scan and used. Having four shorter—rather than two
re-scan with slightly different parameters in order longer—diffusion gradient lobes helps to cancel
to avoid PNS. out eddy currents in the EPI readout. This DSE
No matter the speed, it is always necessary to diffusion preparation greatly reduces eddy cur-
use fat/lipid suppression with EPI. This is because rent effects, but since a second 180° refocusing
of the extremely large fat-water shift (Fig. 6.8), pulse is needed, the longer echo time reduces the
which is an important characteristic of EPI. While SNR of the acquisition. Therefore, if significant
it may seem like there is not much fat in the head, eddy current artifacts are anticipated, a DSE
there is enough subcutaneous fat present that acquisition may be considered, but with the cost
some may shift into the brain during an EPI of reduced SNR due to a longer echo time. The
acquisition, causing difficulties in radiological DSE acquisition is becoming increasingly popu-
reads as well as distortions in the quantification lar, but as the trend is to move to ever-smaller
of the diffusion tensor. Notice that spectral- voxel sizes and is therefore becoming increas-
spatial-based fat suppression also uses a fast ingly SNR starved, the DSE approach may be
slewing, EPI-type waveform—played simultane- abandoned and sophisticated eddy current cor-
ous to the RF pulses—for the slice selection gra- rection methods used.
dient. When very thin sections are used, the Another method for diffusion preparation is
gradient strength might be maxed out and PNS the Stimulated Echo (STE) diffusion preparation.
thresholds might be reached. This is particularly It consists of three non-180° RF pulses. STE
problematic for coronal EPI, such as is used for imaging inherently has half the signal of Stejskal-
hippocampus DTI, as the slice is parallel to the Tanner imaging simply due to the spin physics of
largest cross-sectional area of the body. the three excitations (Fig. 6.19). STE, however,
To remove fat from EPI scans, there are many can have a shorter TE than a Stejskal-Tanner dif-
kinds of lipid suppression methods available, fusion preparation scheme. This makes the STE
such as lipid spoiling (CHESS), lipid pre- preparation more effective for tissues with a short
saturation with an inversion pulse (SPIR, SPAIR), T2 (e.g., liver, muscles), as well as some applica-
inversion of the lipid signal and imaging at a time tions in high-field systems (higher than 3 T)
with no lipid signal (signal null) (STIR), bino- where the T2 times are reduced by the field
mial RF pulses, and selective excitation of the strength. However, because its uses are limited,
water signal without exciting the lipids at all STE is not a common acquisition method, and it
(SPSP). Each of these has its own advantages and is not typically supported on current clinical
disadvantages and each vendor has their pre- scanners.
ferred method, so it is best to evaluate each sys- To review, there are four major considerations
tem and application individually. Generally with when prescribing DTI-EPI:
a good shim, any method may be used; however
STIR is typically avoided with EPI as it inher- • The phase-encode direction should always be
ently decreases the SNR and is additionally sen- anterior/posterior rather than the more typical
sitive to the presence of contrast agents. left/right for non-EPI scans (Fig. 6.15).
Besides the lipid suppression method, other • Magnetic field inhomogeneities (Fig. 6.17)
preparations are used to reduce the artifacts can cause significant artifacts for any EPI-
associated with DTI. A method called Dual Spin- based imaging.
Echo (DSE) diffusion preparation can be used to • Lipid/fat suppression is necessary because of
reduce the effects of the eddy currents (Fig. the large chemical shift of lipids with EPI.
6.18) [13, 14]. Here, rather than performing a • Eddy currents become significant with clinical
typical Stejskal-Tanner diffusion preparation DTI-EPI diffusion gradients and may require
with the gradients spaced around a single 180° DSE or post-processing compensation
pulse, a series of four diffusion gradients are (Fig. 6.18).
114 E. Peterson and R. Bammer

Fig. 6.18 The Double


Spin-Echo (DSE) image
has less shearing and
stretching than the single
spin-echo (Stejskal-Tanner)
image. This is caused by
eddy currents, which can
also be seen in Fig. 6.12.
The distortions are not
typically very large, but in
certain cases, such as
strong diffusion gradients
(high b-values) and single
spin echo, if not addressed,
can disrupt the results. The
major distortion can be
seen where the arrow is
pointing to the FA images
where a band of artificially
high FA is visible in the
single spin-echo image.
This can also be seen in the
color FA image in the same
region. Bands of high
diffusion anisotropy at the
cortex are usually the
hallmark for eddy current
mis-registration. The
advantage of the single
spin-echo imaging method
is a higher SNR, which is
due to a much shorter echo
time than with a double
spin echo

Fig. 6.19 This figure


shows a comparison of a
spin-echo and stimulated
echo diffusion acquisition
with similar imaging
parameters. The lower
SNR of the STE image is
obvious
6 Survivor’s Guide to DTI Acquisition 115

Pros and Cons of Multi-shot EPI when using multi-shot acquisitions, it is neces-
sary to run a phase and motion navigator to cor-
Separating k-space into several groups (called seg- rect for any phase changes as well as motion,
ments or interleaves), each of which is acquired both of which may have occurred between the
with separate excitations, can reduce many of the shots. For many years this has been poorly under-
distortions previously discussed. This results in stood or no effective phase-correction methods
acquisitions that have less distortion from T2*- were in place. Only recently have multi-shot
decay and higher SNR due to a shorter TE time. techniques emerged that can reliably handle this
Depending on how k-space is separated, multi- problem.
shot acquisitions can also reduce off-resonance A method of acquiring multi-shot data with
artifacts. Also, using a multi-shot acquisition can reduced distortions from off-resonant spins and
allow higher resolution images than would other- T2 decay, as well as higher SNR, is called
wise be possible. However, splitting k-space into Readout-Segmented EPI (RS-EPI) (Fig. 6.20)
multiple segments means taking longer to acquire [15]. It splits k-space into segments along the
the whole image, so scan time increases approxi- readout direction rather than the phase-encode
mately by the number of segments. dimension as is traditionally performed in multi-
Another, more difficult, problem arises from shot imaging, e.g., interleaved EPI. As with all
issues of motion and motion-induced phase dif- multi-shot imaging methods, there is a navigator
ferences between the readouts. If there are any acquired before or after the data acquisition in
motion or phase differences between the data, a order to correct for phase and motion differences
simple combination of the data will result in an between the segments [16–18].
artifacted image (Fig. 6.4). This is because the Other methods that are robust to motion and
extra phase accrued due to motion between indi- distortions, such as Short Axis PROPELLER
vidual interleaves is inconsistent between the (SAP) [19], can be used as well; however, they
shots, and this inconsistency results in false sig- generally have similar advantages and disadvan-
nal cancellations and additions. Because of this, tages as RS-EPI [6].

Fig. 6.20 There are several kinds of EPI, and this figure through k-space. However, the distortions are similar
shows some differences between different EPI readout between R = 6 EPI and R = 2 RS-EPI because the splitting
trajectories. These are single shot with low (2) and high of k-space in RS-EPI speeds up the phase encode progres-
(6) parallel imaging factors, and multi-shot Readout- sion through k-space. The SNR of the RS-EPI case is also
Segmented EPI (RS-EPI) with a low parallel imaging fac- similar to the R = 2 EPI because they both have the same
tor of 2. Note how the artifacts are reduced significantly in parallel imaging acceleration and similar echo and repeti-
RS-EPI and R = 6 EPI due to the faster progression tion times
116 E. Peterson and R. Bammer

To review, multi-shot EPI has one major beyond the scope of this chapter. An interested
advantage and two disadvantages: reader may refer to [20]. A single line of k-space
is acquired between each 180° pulse, which
• The advantage is that the distortions are results in a pulse/readout train that is typically
reduced with multi-shot EPI (Fig. 6.20). between 32 and 128 echoes long. The reason for
• A disadvantage is that the SNR is reduced using 180° pulses to refocus the signal between
because of the parallel imaging (Fig. 6.20). each line is that this creates the much longer-
• A second disadvantage is that the processing lived T2-weighting rather than the shorter-lived
becomes more difficult due to phase differ- T2*-weighting observed with EPI. In fact, FSE
ences between shots. readouts can also use much lower flip angles than
180°, which helps to sustain the echo train beyond
the duration of normal T2 times due to contribu-
tions from STE and higher order spin echoes.
The Fast Spin-Echo Acquisition Regardless of the type, FSE is a very robust
imaging method (Fig. 6.11), which is typically
A Fast Spin-Echo (FSE) imaging sequence con- free from significant distortions and artifacts
sists of a single 90° excitation RF pulse followed because each readout is centered on an
by a train of 180° refocusing RF pulses, often RF-refocused spin echo.
called a Carr-Purcell-Meiboom-Gill (CPMG) Despite its advantages, because of technical
pulse train (Fig. 6.21). It is also possible to use difficulties with exciting exactly 180° pulses, a
refocusing pulses less than 180°, which allows certain amount of the signal is lost beyond the
for longer echo trains to be used but introduces expected T2 decay. This results in a decay in the
additional signal modulations and cancellations echo train that is faster than T2 decay alone,

Fig. 6.21 This figure shows a fast spin-echo sequence seen in Fig. 6.1. However, despite the similarities, the EPI
diagram. The readout section is repeated until the desired readout is much faster and has a much lower SAR than the
number of phase-encode lines is completed for each FSE FSE readout due to the lack of refocusing pulses for each
train. Note the similarities to an EPI readout, which is readout line
6 Survivor’s Guide to DTI Acquisition 117

which limits the image resolution. Another issue Motion Sensitivity of Diffusion-
with using a train of 180° pulses is that the tis- Weighted Imaging
sue heating (SAR) from all the RF pulses is
quite high, often requiring an increased TR in Motion is the most common source of artifacts in
order to allow for tissue cooling, thus increasing DTI MRI. It is also one of the few artifacts that
scan time. cannot be reliably avoided with proper setup and
These restrictions often limit the number of testing. Because of this, and the relative sensitiv-
180° pulses to less than what is required for a ity of DTI to motion, motion corruption is the
single-shot acquisition. This necessitates a most common reason to re-run a scan. Motion
multi-shot acquisition, which has the same shot also is often hard to diagnose, as different motions
combination issues as multi-shot EPI acquisi- cause different artifacts. There are too many dif-
tions as well as phase differences between the ferent types of motion artifacts to cover in this
readout lines if non-180° refocusing RF pulses chapter, but it should be the first possibility eval-
are used. A common solution to the shot combi- uated when an image is artifacted.
nation difficulties is a PROPELLER acquisition Because of this variability, this section will
(also called BLADE on Siemens systems), attempt to outline types of motion and generic
which is an acquisition with rotated segments correction methods in a general fashion. This will
[21]. These segments can be ordered and allow you to understand the sources of motion
acquired in many different ways, all of which are artifacts, and use that knowledge to avoid or
designed to reduce the distortion and to improve choose acquisitions that are robust to motion
robustness to motion. An improvement called whenever possible.
SPLICE can be used in PROPELLER to coun- The simplest types of motion in DTI are turns
teract some of the phase inconsistencies between and shifts of the head. These are commonly
readout lines when non-180° pulses are used; called “rigid” motion. These motions can be
however this reduces the SNR and increases the intentional, such as a reaction to the scanner or
scan time [22]. discomfort from lying motionless in the scanner.
Generally, as with segmented k-space acquisi- They can also be unintentional, such as slight
tions using EPI, multi-shot FSE acquisitions— motion from breathing, falling asleep, or an
PROPELLER or standard Cartesian—result in involuntary reaction to the noise of the scan.
significantly longer scan times and residual As with all MRI, motion causes problems in
image phase inconsistencies which result in dis- DTI. Because single-shot EPI or FSE acquires a
tortions and signal loss. Because of these charac- single image in very little time from a single exci-
teristics, FSE acquisitions are not commonly tation, motion does not often occur during the
used with DTI and are often only used in regions image generation. It primarily occurs between
where EPI is too distorted to effectively acquire images, which must then be re-aligned before the
the image. In the brain this is often in the brain- processing is performed.
stem, near the orbits and sinuses, or in regions However, if multi-shot EPI or FSE—where
close to metallic implants. data from several excitations are combined to
To review, the FSE acquisition can be used in form an image—is used, or if the motion occurs
similar situations as EPI, but there are two key during the image generation during single-shot
differences between the techniques to keep in EPI/FSE, signal changes and image distortions
mind: can occur. In this case, rigid motion of the head
during imaging will cause blurring of the brain at
• FSE is more robust to off-resonance best, and signal dropout at worst. An example of
distortions. slight rigid motion can be seen in Fig. 6.22, which
• EPI is faster and is therefore more commonly shows un-corrected and corrected images with
used for DTI. rigid—in this case a turning of the head—motion.
118 E. Peterson and R. Bammer

Fig. 6.22 Rigid motion—


even minor motion as in
this case—can induce
blurring in the diffusion
tensor calculation. When
prospective motion
correction is used, these
artifacts can be reduced or
removed entirely. This
allows low contrast (blue
arrow) and smaller (red
arrow) regions to be
resolved with motion
correction. The FA shows
how directionally intense
the diffusion is while the
color FA incorporates the
direction of the diffusion as
well (red is left-right, blue
is superior-inferior, and
green is anterior-posterior).
Also note that the diffusion
image without diffusion
correction is relatively
crisp, but both the FA and
color FA without motion
correction look noisy and
generally poorly resolved
in contrast with the motion
correction images. This is
typical of motion between
the images, and therefore
the diffusion images do not
properly align for the
tensor calculation

Unfortunately, the motion sensitivity of DTI For all MRI, many imaging parameters, such
does not stop with rigid motion and single-shot as the field of view (FOV), resolution, and read-
EPI. There are many distortions and artifacts out type, are controlled at the physical level by
caused by motion in DTI, and in order to under- the magnetic field gradients in the scanner. The
stand the more complicated multi-shot scans and magnetic field gradients that control the imaging
other effects of rigid and nonrigid motion, it is are the same ones that are used for diffusion.
important to briefly review MRI data Thus the magnetic field gradients used for imag-
acquisition. ing are similar to those used for diffusion, just
6 Survivor’s Guide to DTI Acquisition 119

with different strengths and shapes. This similar- differences can make it difficult to combine data
ity can cause mixed effects—the diffusion gradi- across shots, and often multi-shot scans require
ents can cause imaging-like effects, and the an extra step in the acquisition, called a “naviga-
imaging gradients can cause diffusion-like tor” (echo/image) in order to combine the shots
effects. without signal cancellation and generation arti-
In order to understand these effects, however, facts (Fig. 6.4).
it is important to remember that the acquired While rigid motion artifacts can be greatly
k-space and the images themselves are inherently reduced with navigators as well as more advanced
complex valued. This means that each pixel in motion correction methods, such as prospective
the image—in both k-space and image space— motion correction, nonrigid motion artifacts can
contains a magnitude and phase. be more difficult to remove due to the compli-
The changes in phase due to motion are called cated motion that can occur. Generally, the
motion-induced phase errors and are common in majority of nonrigid phase errors in the brain are
diffusion imaging. This is because the diffusion- caused by pulsation of both blood and cerebral
encoding gradients are designed to be very sensi- spinal fluid. Luckily, this means that most of the
tive to molecular motion, which means that any nonrigid phase errors manifest primarily in the
small motion—a heartbeat, a small shift of the basal regions of the brain near the hippocampus,
head, or even blood flow-induced distortions— cerebellum, thalamus, and hypothalamus.
can cause a large change in image phase and Therefore, if the scan is focused on more apical
potentially the magnitude as well. This motion regions in the head, the effects of nonrigid phase
can be rigid, such as a translation or rotation, or errors can often be ignored. However, if the
nonrigid, such as cerebral spinal fluid or blood region of interest is basal, it is often advanta-
pulsing. geous to use cardiac gating—and potentially
The motion-induced phase error (from either respiratory gating as well—in order to reduce the
rigid or nonrigid motion) can cause interferences effects of nonrigid motion in those regions. It is
and signal cancellation in the image (Figs. 6.4 important to note that while this primarily applies
and 6.23). Importantly, this phase is generated to multi-shot techniques, gating can help improve
by the diffusion gradients, so each shot (excita- single-shot techniques as well improve the con-
tion) can have a different phase error. These sistency between images.

Fig. 6.23 Images of the


brain without and with
cardiac gating show the
signal cancellation that
occurs due to the pulsatile
motion of brain regions
caused by blood or as a
consequence of CSF
spaces. The gating
effectively increases the
signal in the affected
regions as well as helps to
produce consistent signal
across all diffusion
directions. Note that the
pulsatile motion can almost
completely remove the
signal in specific regions
(arrow) or generally dim
the signal from large
regions (wedge)
120 E. Peterson and R. Bammer

As mentioned above, another source of arti- robust, but it requires a good RF coil shape with
facts due to motion in DTI is that if the images an acquisition method that works with—not
themselves are artifact free, but not well aligned, against—the RF coil. In addition to this, all paral-
this can produce significant error in the diffusion lel imaging methods require some kind of coil
calculation. This misalignment can often be rem- calibration in conjunction with the standard
edied through aligning the images before diffu- images.
sion processing, a process called registration. The classic parallel imaging acquisition is
While this process works well, and is often used Cartesian acquisitions performed by skipping
in the standard DTI processing chain, it is best to phase-encode lines in k-space. The skipped lines
try to avoid motion in the first place if at all effectively reduce the FOV of the image, which
possible. results in a “wrapped” or “aliased” image. The
To review, motion is most frequently caused acceleration factor (R) starts at one (which is a
by two sources, both of which can cause major or fully sampled image) and the R number indicates
minor artifacts: that every Rth line is sampled. While it is theo-
retically limited by the number of receive coils, it
• Motion of the head during scanning, either is typically less than four for standard brain DTI
intentional or unintentional. acquisitions for SNR and artifact reasons. The
• Motion of blood or cerebral spinal fluid in the coil geometry—the placement of the subcoils
head due to respiration, the heart, or cerebral around the object—also matters. The coil ele-
spinal fluid pulsations. ments (subcoils) may be more or less suited for
parallel imaging in different directions, depend-
ing on the construction of the coil as a whole.
While the specifics of any parallel imaging
The Reconstruction method can be very complicated, the advantages
are significant. The acquisition speed is increased
The reconstruction takes the acquired data and by the acceleration factor, as is the speed at which
produces images from that data. The acquisition k-space is traversed in an EPI acquisition. This
and reconstruction need to work together to pro- not only speeds up the acquisition itself, resulting
duce the final images. In this section we will in shorter TE and TR times, but for EPI it also
briefly discuss the reconstruction techniques that reduces off-resonance distortions by the acceler-
have a direct link to how the acquisition is ation factor as well.
performed. However, these benefits do not come without a
price: the SNR of a parallel imaging acquisition
is lower than the same acquisition when fully
Parallel Imaging sampled (Figs. 6.24 and 6.25). Also, the SNR is
no longer homogeneous across the image; it is
Parallel imaging was a significant development coil and region dependent, which often results in
in MRI because it allows the acquisition of less a lower SNR at the center of the coil, which
data than is normally required for the image. unfortunately is also often the center of the
Parallel imaging is possible through the use of object. The inherent lower SNR can be offset to
multi-channel receive coils and pulse sequences some extent by a shorter TE time, but generally
which are designed to best take advantage of the the SNR of a parallel imaging acquisition is
speed-up characteristics of parallel imaging. lower than that of the same acquisition performed
The parallel imaging reconstruction can be without parallel imaging.
done in many different ways, but the two major Another previously mentioned limitation of
groups of methods operate in either k-space parallel imaging is that a coil calibration of some
(GRAPPA) [23] or image space (SENSE) [24]. kind is necessary in order to perform the parallel
Parallel imaging is often relatively fast and imaging reconstruction. The coil calibration
6 Survivor’s Guide to DTI Acquisition 121

Fig. 6.24 This figure shows progressive parallel imaging ues, it is clear how much distortion is present, even in the
values from no parallel imaging (R = 1), to values of R = 2, R = 2 images. However, the trade-off is the reduced SNR
4, and 5. Note how the most distorted image is R = 1—the of the higher parallel imaging factors. It is clear that paral-
one without parallel imaging—which is due to the reduced lel imaging factors of R = 4 and above have increased
scan speed, and the noisiest is the one with a parallel noise values when compared to lower parallel imaging
imaging value of 5. When comparing the geometric dis- values. This demonstrates the balance between distortion
tortions of the images with different parallel imaging val- and SNR that parallel imaging can provide

Fig. 6.25 The demonstration in Fig. 6.24 used GRAPPA when un-aliasing the image. However, when the calibra-
(a k-space based) parallel imaging, but a different set of tion is incorrect—as in the right-most image—the un-
parallel imaging errors can occur with SENSE (an image aliasing does not perform as well. As the arrows indicate,
space based) parallel imaging. SENSE is based on a coil this results in images that are incorrectly un-aliased. In
calibration scan, and this scan can easily become cor- this case, the major artifact is the eye causing a bright spot
rupted through motion or distortion differences between in the brain, which could potentially be confused with
the calibration and the EPI scan. When the calibration is abnormal pathology by a radiologist unaware of the pos-
correct, the SENSE reconstruction performs very well sibility for this error
122 E. Peterson and R. Bammer

Fig. 6.26 This schematic shows common head coil con- 8-element coil (c). Note that this is just an example of a
figurations. It shows a transverse view of a typical 8- or common element setup, and they may vary significantly
32-element head coil (a), a longitudinal view of a from coil to coil
32-element coil (b), and a longitudinal view of an

method varies between parallel imaging tech- geometry when deciding on the imaging plane
niques, but in essence all of the calibration meth- and acceleration factor, in conjunction with the
ods are ways to interpret the sensitivity of the RF previously discussed imaging parameters.
coils, which are then used to form the final image. Typically, clinical scanners will have some
If the calibration is incorrect, serious artifacts can awareness of the parallel imaging capabilities
be produced, which can either mask or appear to and the allowed acceleration, but this is often
be actual medically important information in the intended to prevent scans from being run which
images (Fig. 6.25). As a rule of thumb, the will fail completely.
k-space velocity of the calibration scan should To review, parallel imaging is frequently used
ideally be identical to the k-space velocity of the in MRI for three major reasons:
SENSE scan to be distortion matched.
Often, parallel imaging coils for head imaging • Parallel imaging reduces the distortion from
contain multiples of eight channels (Fig. 6.26). off-resonance with a negligible SNR penalty
These are typically arranged symmetrically in if used conservatively.
rings around the head, with each set of eight • Multi-channel coils are commonly available
channels in a different position respective to the in the clinic.
apex of the coil. Therefore, a typical 8-channel • Parallel imaging reconstructions are robust
head coil has a single ring of coils, and a and readily available.
32-channel head coil has four rings spaced
equally along its length. Because of this, a
32-channel head coil should be able to support Partial Fourier Imaging
accelerations in any direction, whereas an
8-channel coil can only reliably support parallel Partial Fourier acquisition is another method
imaging in the x- and y-axes. With parallel imag- used to reduce the TE and TR. In this case,
ing, it is therefore important to consider the coil k-space lines are also skipped; however the lines
6 Survivor’s Guide to DTI Acquisition 123

Fig. 6.27 This figure shows images with progressively a much higher partial Fourier fraction is required in order
more partial Fourier sampling, with values from 0.5625 to allow a good reconstruction (compare d–f). The sus-
(a, d), 0.625 (b, e) to 0.75 (c, f). Patient 1 shows a case ceptibility of the patients to the partial Fourier fraction is
where a very low partial Fourier fraction causes very little impossible to predict, so a sufficiently high fraction is
artifact (compare a–c). However, patient 2 is a case where necessary to avoid significant artifacts in all cases

omitted are a contiguous block of high-frequency To review, partial Fourier imaging is often
readouts that are often skipped before the echo, used clinically for two reasons:
thereby shortening the TE. The partial Fourier
reconstruction then relies not on multiple RF • It is a straightforward way to shorten the TE
coils but on an assumption about the image—that and TR of scans without inducing significant
the phase angle across the image changes slowly. artifacts if used conservatively (Fig. 6.27).
In essence, if the phase angle change across the • The reconstruction is robust and widely
image is assumed to be slow, the whole k-space is implemented.
not required to correctly reconstruct the image.
Because this is generally a valid assumption, a
partial Fourier approach is often used to decrease Eddy Current Correction
the scan and TE times.
However, as with any imaging technique Eddy currents, as previously discussed, can cause
where k-space is sampled less than it should be, geometric distortions in diffusion-weighted
sampling too few lines in k-space will result in a images (Figs. 6.12 and 6.18). While the Double
distorted image (Fig. 6.27). It is difficult to deter- Spin-Echo (DSE) pulse sequence approach to
mine how much is too much, as it often depends eddy current distortion reduction is one possible
on the scan and the phase angle change in the solution, it is also possible to correct eddy current
object [25]. As with parallel imaging, it is always distortions in post-processing [11, 26]. The post-
better to err on the side of caution rather than processing consists of an alignment of the group
have an artifacted scan. of images in the diffusion series to ensure they
124 E. Peterson and R. Bammer

are all well aligned with one another before the FOV requires stronger gradients than a larger
diffusion tensor processing step takes place. This FOV, so reducing the FOV to better tailor it to the
alignment is typically performed as part of the smaller head asks more of the imaging gradients.
DTI post-processing pipeline in conjunction with It simply may not be possible to achieve the
motion correction (and sometimes even distor- desired scan parameters (e.g., TE and TR) with a
tion correction) because without correcting for smaller FOV, but this is very system dependent.
even mild motion and eddy currents, the tensor Additionally, the SAR limit for children—
computation can become significantly degraded. who are significantly lighter than adults—is sig-
To review, eddy currents can be corrected in nificantly lower. This can cause problems for
the acquisition and reconstruction: FSE or spin-echo sequences, and can be a hidden
surprise when transitioning from adult to pediat-
• In the acquisition a DSE scan can significantly ric imaging. The SAR is difficult to change
reduce the eddy current distortions. without significantly changing the type of imag-
• In the reconstruction, registration can be per- ing, so it may be necessary to change the imaging
formed to reduce the eddy current distortions. type or slow down the acquisition (increase the
TR) in order to perform the desired type of
imaging.
Finally, as children’s brains are still growing,
Pediatric Considerations there is less developed structure to provide diffu-
sion contrast, so the diffusion values need to be
The previous sections of this chapter have been reduced. As values around 1000 s/mm2 are typi-
written generally for any kind of DTI—pediatric cally used in adults, values less than that, typi-
or otherwise. However, as pediatric imaging is cally around 700 s/mm2, are used in pediatric
performed much less often than adult imaging, DTI studies [27].
protocols are often adapted from adult studies for As we have suggested earlier, it is vitally
use in pediatric exams. This conversion is typi- important to test the adapted sequence and proto-
cally minor; however there are few changes that col before using it on a child. This will ensure
should be considered when modifying a standard that it proceeds smoothly and quickly—which is
DTI protocol for pediatric imaging. crucial to ensure high quality results with the
An important consideration in the acquisition least amount of motion in pediatric studies.
for pediatric imaging is that children often are To review, there are four major changes to be
less likely to follow instructions and are more made for pediatric DTI:
likely to move while in the scanner. Because of
this, acquisitions that are robust to motion, such • Because children are more unlikely to follow
as PROPELLER or single-shot EPI, are more instructions and hold still, any motion correc-
commonly used. Other types of prospective and tion or motion restriction is greatly
retrospective motion corrections are beneficial encouraged.
and can be used; however these are beyond the • The head is smaller, so the FOV should be
scope of this section. A simple method to help reduced to better fit the smaller anatomy.
reduce motion is to simply add more pads around • The SAR limit is lower because children are
the patient’s head to help discourage head move- smaller than adults, so it is important to test
ment during the scan session. the SAR of the sequence before scanning a
Another pediatric DTI imaging consideration child.
is the size of the head. Often protocols designed • Because the brain is still developing, the diffu-
for adults will have FOVs that are substantially sion of water is typically higher in children, so
larger than what is required for children. In this a lower b-value should be used when com-
case, it is important to remember that a smaller pared to adults.
6 Survivor’s Guide to DTI Acquisition 125

Conclusion Reson Med. 2012;67(5):1210–24. doi:10.1002/


mrm.23097.
8. Tuch DS. Q-ball imaging. Magn Reson Med.
We hope that this chapter has given you a basic 2004;52(6):1358–72. doi:10.1002/mrm.20279.
understanding of the acquisition for diffusion 9. Assaf Y, Cohen Y. Structural information in neuronal
tensor imaging. Above all, we advise you to test tissue as revealed by q-space diffusion NMR spec-
troscopy of metabolites in bovine optic nerve. NMR
parameters for yourself to see how best to tune
Biomed. 1999;12(6):335–44. Retrieved from http://
them for your application. In order to help you www.ncbi.nlm.nih.gov/pubmed/10516615.
with this process, we have provided basic descrip- 10. Skare ST, Clayton DB, Newbould RD, Moseley ME,
tions and images of the most important parame- Bammer R. A fast and robust minimum entropy based
non-interactive Nyquist ghost correction algorithm.
ters and acquisition methods. Our hope is that
In 14th Annual Meeting of ISMRM, Seattle, WA,
these descriptions and examples will help you 2006.
run the best DTI acquisitions for your 11. Andersson JLR, Skare S. A model-based method for
applications. retrospective correction of geometric distortions in
diffusion-weighted EPI. Neuroimage.
2002;16(1):177–99. doi:10.1006/nimg.2001.1039.
Acknowledgements The authors would like to acknowl- 12. Van Vaals J, Bergman AH. Optimization of eddy-
edge Samantha Holdsworth for the helpful discussions current compensation. J Magn Reson. 1990;70(August
and assistance in collecting some of the images used in 1989):52–70.
this chapter, Didem Aksoy for assistance in acquiring 13. Reese TG, Heid O, Weisskoff RM, Wedeen
some of the images used herein, and Eric Aboussouan for VJ. Reduction of eddy-current-induced distortion in
help in creating some of the figures. diffusion MRI using a twice-refocused spin echo.
Magn Reson Med. 2003;49(1):177–82. doi:10.1002/
mrm.10308.
14. Wider G, Doetsch V, Wuerthrich K. Wider_1994_
References eddy_current_correction.pdf. J Magn Reson.
1994;108:255–8.
1. Stejskal EO, Tanner JE. Spin diffusion measurements: 15. Holdsworth SJ, Skare S, Newbould RD, Guzmann R,
spin echoes in the presence of a time-dependent field Blevins NH, Bammer R. Readout-segmented EPI for
gradient. J Chem Phys. 1965;42(1):288. rapid high resolution diffusion imaging at 3 T. Eur
doi:10.1063/1.1695690. J Radiol. 2008;65(1):36–46. doi:10.1016/j.
2. Basser PJ, Mattiello J, LeBihan D. MR diffusion tensor ejrad.2007.09.016.
spectroscopy and imaging. Biophys J. 1994;66(1): 16. Bammer R, Stollberger R, Augustin M, Simbrunner J,
259–67. doi:10.1016/S0006-3495(94)80775-1. Offenbacher H, Kooijman H, et al. Diffusion-
3. Pierpaoli C, Basser PJ. Toward a quantitative assess- weighted imaging with navigated interleaved echo-
ment of diffusion anisotropy. Magn Reson Med. planar imaging and a conventional gradient system.
1996;36(6):893–906. Retrieved from http://www. Radiology. 1999;211(3):799–806. Retrieved from
ncbi.nlm.nih.gov/pubmed/8946355. http://www.ncbi.nlm.nih.gov/pubmed/10352609.
4. Tuch DS, Reese TG, Wiegell MR, Makris N, Belliveau 17. Butts K, Crespigny AD, Pauly JM, Moseley
JW, Wedeen VJ. High angular resolution diffusion M. Diffusion-weighted interleaved echo-planar imag-
imaging reveals intravoxel white matter fiber hetero- ing with a pair of orthogonal navigator echoes. Magn
geneity. Magn Reson Med. 2002;48(4):577–82. Reson Med. 1996;35(5):763–70.
doi:10.1002/mrm.10268. 18. Butts K, Pauly J, Crespigny AD, Moseley M. Isotropic
5. Bammer R, Holdsworth SJ, Aksoy M, Skare ST. Phase diffusion-weighted and spiral-navigated interleaved
errors in diffusion-weighted imaging. In: Jones DK, EPI for routine imaging of acute stroke. Magn Reson
editor. Diffusion MRI: theory, methods, and applica- Med. 1997;38(5):741–9.
tions. New York, NY: Oxford University Press; 2011. 19. Skare S, Newbould RD, Clayton DB, Bammer
p. 218–49. R. Propeller EPI in the other direction. Magn Reson
6. Skare ST, Bammer R. EPI-based pulse sequences for Med. 2006;55(6):1298–307. doi:10.1002/mrm.20890.
diffusion tensor MRI. In: Jones DK, editor. Diffusion 20. Busse RF, Brau ACS, Vu A, Michelich CR, Bayram
MRI: theory, methods, and applications. New York, E, Kijowski R, et al. Effects of refocusing flip angle
NY: Oxford University Press; 2011. p. 182–202. modulation and view ordering in 3D fast spin echo.
7. Setsompop K, Gagoski BA, Polimeni JR, Witzel T, Magn Reson Med. 2008;60(3):640–9. doi:10.1002/
Wedeen VJ, Wald LL. Blipped-controlled aliasing in mrm.21680.
parallel imaging for simultaneous multislice echo pla- 21. Pipe JG. Motion correction with PROPELLER MRI:
nar imaging with reduced g-factor penalty. Magn application to head motion and free-breathing
126 E. Peterson and R. Bammer

cardiac imaging. Magn Reson Med. 1999;42(5): Retrieved from http://www.ncbi.nlm.nih.gov/entrez/


963–9. Retrieved from http://dx.doi.org/10.1002/ query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citati
(SICI)1522-2594(199911)42:5<963::AID-MRM17> on&list_uids=10542355.
3.0.CO;2-L. 25. Jaermann T, Pruessmann KP, Valavanis A, Kollias S,
22. Deng J, Omary RA, Larson AC. Multishot diffusion- Boesiger P. Influence of SENSE on image properties
weighted SPLICE PROPELLER MRI of the abdo- in high-resolution single-shot echo-planar DTI. Magn
men. Magn Reson Med. 2008;59(5):947–53. Reson Med. 2006;55(2):335–42. doi:10.1002/
doi:10.1002/mrm.21525. mrm.20769.
23. Griswold MA, Jakob PM, Heidemann RM, Nittka M, 26. Horsfield MA. Mapping eddy current induced fields
Jellus V, Wang J, et al. Generalized autocalibrating for the correction of diffusion-weighted echo planar
partially parallel acquisitions (GRAPPA). Magn images. Magn Reson Imaging. 1999;17(9):
Reson Med. 2002;47(6):1202–10. doi:10.1002/ 1335–45.
mrm.10171. 27. Sundgren PC, Dong Q, Gómez-Hassan D, Mukherji
24. Pruessmann KP, Weiger M, Scheidegger MB, SK, Maly P, Welsh R. Diffusion tensor imaging of the
Boesiger P. SENSE: sensitivity encoding for fast brain: review of clinical applications. Neuroradiology.
MRI. Magn Reson Med. 1999;42(5):952–62. 2004;46(5):339–50. doi:10.1007/s00234-003-1114-x.
Strategies and Challenges in DTI
Analysis 8
Wim Van Hecke and Louise Emsell

• Many software packages and tools are avail-


Learning Points able to process and analyse DTI data, which
vary considerably in functionality.
• DTI analysis forms only one part of a DTI • The broad range of analysis approaches and
study and is mutually dependent on other stages heterogeneous functionality in software pack-
in the DTI pipeline, such as data acquisition. ages contributes to a lack of standardization
• There are many different approaches for ana- that complicates the analysis of DTI data and
lyzing DTI and the most optimal method the interpretation of results.
depends on the goal(s) of the DTI
investigation.
• DTI analysis methods can be categorized into Introduction to DTI Analysis
three main classes: whole-brain, regional, and
voxel-based approaches. Since its introduction, DTI has been used to
• There are pros and cons in all DTI analysis study microstructural tissue changes in a wide
approaches, and there is no single best or range of neurologic and psychiatric disorders, as
worst analysis method, but a range of tech- well as in normal development and ageing [1].
niques that are more or less suited to any given Many approaches have been proposed to extract
application. DTI measures from the data and compare them
across subjects. As each of these methods have
some advantages and limitations, the most opti-
W. V. Hecke, PhD (*) mal analysis approach will depend on the clinical
icometrix, Tervuursesteenweg 244, and research questions that need answering.
3001 Leuven, Belgium Furthermore, the limitations of the selected
Department of Radiology, Antwerp University Hospital, method should be considered during the interpre-
Wilrijkstraat 10, Edegem, 2650 Antwerp, Belgium tation of the results. This chapter provides a brief
e-mail: wim.vanhecke@icometrix.com
overview of the different options that are avail-
L. Emsell, PhD able for the analysis of DTI data. In the following
Departments of Translational MRI and Radiology,
chapters, more detailed information is provided
KU Leuven and University Hospitals Leuven,
Leuven, Belgium about three main analysis techniques, i.e., region
of interest analysis (Chap. 9), voxel-based analy-
Department of Old Age Psychiatry,
Universitair Psychiatrisch Centrum (UPC) - KU sis (Chap. 10), and tractography and connectivity
Leuven, Leuven, Belgium analysis (Chap. 11).

© Springer Science+Business Media New York 2016 153


W. Van Hecke et al. (eds.), Diffusion Tensor Imaging, DOI 10.1007/978-1-4939-3118-7_8
154 W.V. Hecke and L. Emsell

Fig. 8.1 Prototypal DTI study pipeline. Whole-brain differences in euthymic bipolar I disorder: a combined
tractogram and connectivity matrix. [Reprinted from magnetic resonance imaging and diffusion tensor imaging
Caeyenberghs K, Leemans A, Leunissen I, Gooijers J, voxel-based study. Bipolar Disord. 2013 Jun;15(4):365–
Michiels K, Sunaert S, et al. Altered structural networks 376. With permission from John Wiley & Sons.]Axon
and executive deficits in traumatic brain injury patients. micrograph. [Reprinted from Beaulieu C. The basis of
Brain Struct Funct. 2014 Jan;219(1):193–209. With per- anisotropic water diffusion in the nervous system—a
mission from Springer Verlag]. Voxel-based analysis fig- technical review. NMR Biomed. 2002 Nov–Dec;15
ure. [Adapted from Emsell L, Langan C, Van Hecke (7–8):435–455. With permission from John Wiley &
W,Barker GJ, Leemans A, Sunaert S, et al. White matter Sons, Inc.] Corrected DTI maps. [Courtesy of A. Leemans]

Analyzing DTI data is only one part of the detail in the chapters of Section 2, many choices
whole DTI processing pipeline. Figure 8.1 sum- have to be made at each step of this pipeline.
marizes a prototypal DTI pipeline, from the goal Note that these different steps are not indepen-
of the DTI study, through to data acquisition, data dent of each other; for example, the most optimal
analysis, and interpretation of the results. As analysis technique will depend on the quality of
highlighted in Chap. 2 and discussed in more the data and how it is acquired.
8 Strategies and Challenges in DTI Analysis 155

Why Do We Need to Analyze techniques available for analyzing DTI data com-
DTI Data? pared to DWI. In clinical practice, DWI informa-
tion, typically the DWI and ADC maps, is
Diffusion-weighted imaging (DWI) is widely interpreted visually by a radiologist. It has been
used in clinical practice as it provides unique, demonstrated that DTI can be useful in evaluat-
rapidly accessible information that can be used in ing changes in the normal appearing white mat-
the assessment of ischaemic stroke, to differenti- ter. However, qualitative assessment of DTI
ate vasogenic versus cytotoxic oedema and to information, such as FA maps, may be more dif-
characterize intracranial lesions such as pyogenic ficult there.
abscess, infections, tumors, and trauma [2]. To illustrate the challenge of qualitatively
However, whilst the processing of DWI data is assessing scalar DTI maps, consider the axial
relatively easy, the analysis of DTI data is signifi- color-encoded FA maps in Fig. 8.2. This random
cantly more complex. For example, the need for assortment of images comprises seven pairs of
more diffusion-weighted images makes the axial slices generated from patients with pathol-
acquisition longer and more challenging. In addi- ogy that has been associated with changes in
tion, motion correction becomes more important, white matter microstructure, and two healthy
and the tensor estimation is more complex com- subjects. There are two patients with tinnitus, two
pared to ADC calculations. There are also more with cerebral palsy, two with multiple sclerosis,

Fig. 8.2 A matching


puzzle with DTI. Match
the axial colour-encoded
FA maps with the correct
pathology. In addition to
two healthy subjects, there
are two images of patients
with tinnitus, cerebral
palsy, multiple sclerosis,
schizophrenia, Alzheimer’s
disease, spinocerebellar
ataxia, and amyotrophic
lateral sclerosis
156 W.V. Hecke and L. Emsell

two with schizophrenia, two with Alzheimer’s analysis, tractography, histogram analysis, atlas-
disease, two with spinocerebellar ataxia and two based segmentation, quantification of graph-based
with amyotrophic lateral sclerosis. Is it possible connectivity networks, and voxel-based analysis
to match the FA maps with the correct pathology to name but a few. Each of these techniques has
and identify the healthy controls? its own strengths and limitations and there is no
For all subjects, a similar axial slice was single technique that can be regarded as superior
selected. Data from the subjects with the same to all the others. The most optimal analysis
pathology were acquired using the same protocol approach depends on many factors, including:
in the same study, whereas data from subjects
with different pathologies were acquired in differ- • The purpose of the analysis (e.g., to delineate
ent studies (and therefore mostly with different a known fibre bundle, to explore the data)
acquisition protocols). Hence, it may be possible • Whether it is for a single subject or group
to match some subjects based on image quality or comparison
based on prior knowledge about the presence of • If there is a hypothesis about the location and
neurodegeneration and ventriculomegaly in some extent of change or difference in DTI measures
of these disorders. However, when these factors • The data acquisition protocol (e.g., # of direc-
are excluded from the visual assessment of the tions, b-value, voxel size)
data, it becomes very difficult to match the pathol- • The data quality
ogy to the DTI data. This demonstrates firstly, that • …
changes in FA that occur due to pathology are not
always readily visualised on colour FA maps, and For simplicity, the different techniques that
secondly, that such FA changes are not specific to are available to analyze DTI data sets can be clas-
one particular disorder. Although visual assess- sified into three categories:
ment of colour FA maps can be useful, in general,
there is a need for reliable quantitative analysis • Whole-brain analyses
methods that allow meaningful conclusions to be • Region-specific analyses
drawn from the DTI data. • Voxel-based analyses

This subdivision of analysis techniques is


DTI Analysis Techniques based on the scale that is used to evaluate the DTI
measures in the brain. As shown in Fig. 8.3, DTI
Many different DTI analysis techniques and analysis can be performed at the level of the whole
approaches have been applied to study a range brain (Fig. 8.3a), at a regional level (Fig. 8.3b), or
of pathologies and include region of interest at the smallest scale, i.e., the voxel (Fig. 8.3c).

Fig. 8.3 Subdividing DTI analysis methods into three parts: whole-brain analysis approaches (a), region-specific anal-
ysis methods (b), and voxel-based analysis methods (c)
8 Strategies and Challenges in DTI Analysis 157

In most voxel-based analysis approaches, DTI


measures are evaluated at the voxel level, but at
the same time in every voxel of the brain. As
such, this method can also be regarded as a
whole-brain analysis technique. This next section
provides a brief overview of each of these major
classes of DTI analysis methods.

Whole-Brain Analysis Techniques

The general concept of whole brain DTI analysis


techniques is to obtain quantitative DTI measures
from all the voxels that include brain white mat-
ter, and can thus be subdivided into two parts (see
Fig. 8.4):

• An approach to define which voxels are part


of the brain white matter
• An approach to extract relevant DTI informa-
tion from these voxels

The selection of the voxels to be included in


the analysis can be done using brain segmenta-
tions from anatomical MRI data sets (located in
the same image space as the DTI data) or by per-
forming whole-brain tractography. In whole-
brain tractography, all brain voxels are used as
seed regions to start the tractography process.
Using specific parameter constraints such as an
FA and curvature threshold, the tracts will mainly
traverse white matter voxels, as the FA is lower in
grey matter and cerebrospinal fluid. Fig. 8.4 An example of whole-brain analysis of DTI
Once the voxels are selected, the DTI infor- measures. Brain or white matter voxels are defined by a
mation can be extracted. If anatomical MR mask created from either an anatomical MRI segmenta-
tion or by performing whole brain tractography. A histo-
based segmentations are used, it is important to gram of the diffusion values in these voxels is obtained
ensure that the anatomical image and the DTI and relevant information can be extracted and compared
data set are located in the same space. It is there-
fore necessary to register both images to each tion, see Chap. 7), the calculated tensors and
other (image registration is introduced in Chap. diffusion metrics will also be aligned with the
10). Usually, the anatomical image is trans- anatomical MRI. Extracting the diffusion infor-
formed to the non diffusion-weighted image mation after whole-brain tractography doesn’t
using a rigid-body or affine transformation. As involve image registration with an anatomical
all the diffusion-weighted images should already MR image. DTI measures from voxels that contain
be in the same space as the non-diffusion- a streamline from the whole-brain tractography
weighted image (done during the motion correc- result will be selected.
158 W.V. Hecke and L. Emsell

Histogram Analysis Region-Specific Analysis Techniques


Once the DTI measures have been extracted from
the selected voxels of interest, they can be sum- In region specific analysis techniques, diffusion
marized using a histogram (see Fig. 8.4). This measures are obtained in one or more predefined
histogram is a frequency distribution that dis- areas of the brain. DTI measures, such as the FA
plays the number of voxels with a specific value and MD are thus statistically evaluated in an ana-
of the diffusion measure (e.g., FA). From this his- tomical region or white matter tract reconstruc-
togram, the following parameters can be tion. There are two main approaches:
extracted:
• Region of interest analysis
• Mean or median of the diffusion measure • Tractography analysis
values
• The peak height of the histogram: voxel count Region of Interest Analysis
of the value that is present the most In region of interest (ROI) analysis, diffusion
• The peak location of the histogram: the diffu- measures are obtained from a specific brain
sion measure value that is present the most in region, which is defined by manual delineation or
the data set by automated segmentation or parcellation. As
automated segmentations are less observer
Usually, studies will only obtain the mean or dependent and thus more reproducible, they have
median value of the diffusion measure. The some clear advantages over manual delineations.
resulting values can then be statistically com- However, automated segmentations are not
pared across groups of subjects or correlated with always appropriate, for example due to ill-defined
other variables, such as clinical, neuropsycho- boundaries in regions of pathology.
logical or other test scores. Manual delineation of ROIs is typically per-
Whole-brain analysis of DTI data has the fol- formed by freehand drawing of the region or by
lowing strengths and limitations: placing basic shapes such as circles or squares on
2D slices. Due to the manual interaction that is
Strengths needed, the results are observer dependent. In
• Does not require prior knowledge of where addition, manually delineating specific regions
hypothesized differences could be found in a group of subjects is time consuming. This is
• Less reliant on user intervention than other especially the case when white matter fibre bun-
approaches dles need to be delineated, as they run through
• Results obtained quickly, without labor- several slices, and thus many 2D ROIs need to be
intensive interventions drawn in order to delineate as much of the bundle
• Fewer statistical tests (i.e., multiple compari- as possible. Ideally, ROIs should be drawn on
sons), compared to other techniques as only maps that are independent of the diffusion mea-
one set of diffusion measures is obtained for sures of interest. For example if FA maps are
the whole brain used to delineate regions, and the FA is a mea-
sure of interest, a bias can be introduced in the
Limitations results because ROIs are typically drawn around
• Regional information is lost as DTI measures regions with a higher FA. However, FA might be
are averaged over the whole-brain white lower in areas of pathology, which could then be
matter excluded from the analysis, thereby artificially
• Results are sensitive to partial volume effects decreasing differences with the control group. In
due to atrophy contrast, regions delineated on an anatomical
• Results can depend on segmentation/registra- MR (T1/T2) image are drawn independently of
tion accuracy or whole-brain tractography the diffusion measures that will be analyzed.
parameters However, this approach also has some potential
8 Strategies and Challenges in DTI Analysis 159

Fig. 8.5 Axial, sagittal, and coronal slices of a T1 weighted image and the color-encoded FA maps of a healthy
subject

limitations, as the anatomical MRI data set needs Instead of delineating regions and structures
to be registered accurately to the DTI data set, manually, automatic segmentation methods can
which is not always straightforward due to dif- be used. Such automated methods are especially
ferent distortions in both images [3]. An alterna- useful when structures or lesions can be accu-
tive approach is to delineate the regions on the rately segmented on the anatomical MRI. As an
non-diffusion-weighted image, which should be example, T2 lesions could be segmented in a
in the same space as the quantitative diffusion patient with multiple sclerosis. After registering
maps after motion correction. However, the the T1/T2 MR image to the DTI data set and
delineation of white matter bundles on either the applying the deformation field to the segmented
anatomical scans or non-diffusion-weighted lesion masks, DTI measures can be derived from
images is confounded by the lack of orientational these lesions. Bear in mind that these results will
contrast (which is provided by the color FA map). strongly depend on the segmentation and regis-
This is illustrated in Fig. 8.5, which shows axial, tration accuracy, especially when some of the
sagittal, and coronal slices of a T1-weighted lesions are small. In addition, the resolution of
image and corresponding color-coded FA slices the DTI image is typically lower than the resolu-
of a healthy subject tion of the anatomical MRI that is used for the
In the presence of lesions, ROI analysis (actu- segmentation, leading to partial volume effects.
ally all DTI analyses) can become challenging. Finally, note that it is not easy to obtain automatic
This is demonstrated in Fig. 8.6, which illustrates segmentations of white matter tracts based on
axial non-diffusion-weighted and color-encoded anatomical MR images.
FA slices from five patients with cerebral palsy. Region-specific analysis of DTI data by using
The lesions in the left hemisphere clearly affect the ROI approach has the following advantages
the visualization of the corticospinal tract (CST). and limitations:
Reliably comparing diffusion values from the left
CST with the contralateral CST in this popula- Strengths
tion or of a healthy population would be difficult. • In comparison to whole-brain analyses, more
For example, delineating the ROI based on the regionally specific information is obtained
color-encoded FA maps can be biased by the • Manual delineation is closer to the original
lower FA values in the lesion. However, drawing data than other techniques which require more
the ROI on the non-diffusion-weighted image, complex modeling and image processing
which is independent from the diffusion mea- • ROI analysis is less dependent on parameter set-
sures, is also challenging. tings than tractography or voxel-based analysis
160 W.V. Hecke and L. Emsell

Fig. 8.6 Axial non-diffusion-weighted and color-encoded FA slices in five patients with cerebral palsy. The presence
of lesions make ROI delineation challenging

Limitations • Requires clear guidelines that describe how


• Requires a prior hypothesis about where dif- the ROI should be defined (e.g., size, anatomi-
ferences could be found, as that is where the cal location, boundaries).
ROI will be placed. • The selection of many ROIs increases the
• Intra- and inter-observer reproducibility of number of statistical tests that are performed
results should be assessed, as manual delinea- and therefore correction for multiple compari-
tion is subjective. sons is required.
8 Strategies and Challenges in DTI Analysis 161

• Results can be biased if ROIs are drawn on the


parameter map of the measure of interest, e.g.,
drawing an ROI on a color FA map when
investigating FA.
• Results can depend on segmentation/registra-
tion accuracy when ROIs are delineated on
anatomical MR images.
• Delineating regions manually is very time
consuming and laborious.
• Excludes (potentially valuable) information
from regions that are not selected/studied.
• Drawing an ROI or segmenting a structure can
be challenging in the presence of pathology.

Fig. 8.7 Connecting the dots: by following the alphabet


Tractography Analysis and drawing a line between subsequent letters, the global
The delineation of white matter tracts using only picture (i.e., a house) becomes clear
2D manually drawn ROIs or anatomical MR
images is not optimal for the reasons outlined
previously. However, by using the inherent direc- (Chaps 3–5), the orientation of the estimated
tional diffusion information in the DTI data set, tensor is assumed to relate to the underlying
virtual representations of white matter fibre bun- white matter architecture, as the amount of diffu-
dles can be reconstructed, using tractography (or sion along the axonal bundles will be greater
“fibre tracking”). Tractography refers to the compared to the amount of diffusion perpendicu-
mathematical reconstruction of white matter lar to them. DTI tractography is based on the
fibre bundle representations by integrating the assumption that by following the maximal
local diffusion tensor information from every amount of diffusion in a given direction (i.e., the
voxel. In its simplest form, tractography can be longitudinal axis of the ellipsoid) in each voxel,
compared with a puzzle “connecting the dots.” the orientation of axon bundles can be followed,
As shown in Fig. 8.7, by following the letters and hence the tensors provide an indirect, sim-
alphabetically, and drawing lines between subse- plistic, discrete representation of white matter
quent letters, one can complete the drawing and fibre pathways, as shown in Fig. 8.8c. In prac-
the global picture. tice, these assumptions suffer major flaws, which
How diffusion tractography relates to “con- are discussed in detail in several other chapters
necting the dots,” is shown in Fig. 8.8. Instead of (see especially Chaps. 5, 11 and 21).
the alphabet and the natural sequence of letters, If the tractography process is started in the
the orientational diffusion information can be green voxel A (referred to as the seed voxel), the
followed and connected to create a more global main direction of diffusion is followed, until a
picture of the white matter bundle. Consider two new voxel is reached (voxel B in Fig. 8.8d). This
voxels in the brain, i.e., the green and blue vox- process is then repeated until a certain stop crite-
els that are shown in Fig. 8.8a. The DTI data can rion is reached. Typical tracking initiation and
be used to estimate tensors in every voxel. Recall termination criteria are based on selection and
that these tensors can be represented by an ellip- exclusion ROIs, and FA, fibre length and curva-
soid whose longest axis represents the direction ture thresholds. For example, tracking may be
of maximal diffusion. For visualization pur- stopped when the FA in a voxel is below 0.2, to
poses, only the relevant tensors between the prevent streamlines going into low anisotropy
green and blue voxels are displayed, as shown in grey matter or CSF. These ROIs and thresholds
Fig. 8.8b. As explained in Section 2 of this book determine the number of streamlines and how
162 W.V. Hecke and L. Emsell

Fig. 8.8 A simplified


example or diffusion tensor
tractography. Two voxels
are selected in the brain
(a) and the relevant tensors
in between the voxels are
visualized (b). As these
tensors are representations
of the underlying white
matter axonal bundles (c),
they can be used to
mathematically reconstruct
virtual representations of
these bundles (d and e)
[Courtesy of A. Leemans]

they travel through the data, and hence the final mathematical reconstructions that bear some
tract reconstruction. It is therefore important to resemblance to parts of axonal bundles. Therefore
realize that tractography is both operator and the thickness, length or number of these recon-
parameter dependent, and there is no ‘ground- structed tracts cannot be directly related to the
truth’ solution to validate tracking results. underlying microstructure or anatomy.
In the example of Fig. 8.8, tracking ends in the As tractography uses directional diffusion
blue voxel, as shown in Fig. 8.8d. The pathway information to reconstruct connections in the
from seed voxel to end point can be represented brain, it is an elegant technique for obtaining dif-
by a streamline. In this simplified case, a single fusion measures from specific white matter bun-
the streamline represents a fibre tract (see the dles. One of the most useful and common
orange line in Fig. 8.8e, representing part of the applications of tractography is the noninvasive,
cingulum); however in practice, many stream- virtual dissection of fibre bundles in 3D, i.e., seg-
lines make up a fibre tract. mentation. The segmented tract is equivalent to a
It is worth noting that some of the terminology 3D ROI from which diffusion measures can be
used in tractography can be confusing. In tractog- calculated. This obviates the need to delineate the
raphy, a fibre, streamline or track is not synony- bundle manually by using 2D ROIs on different
mous with an actual nerve fibre in the biological slices or to apply segmentation methods to ana-
sense, and a fibre tract is not synonymous with an tomical MR images, which contain less specific
anatomical fibre bundle (even in the case of fibre white matter tract information. Typically, the
bundles that include “tract” in their anatomical average of the DTI measure, e.g., FA, is calcu-
name, such as the corticospinal tract!). These lated from all voxels that are part of the delin-
concepts are explained in more detail in Chap. eated tract. A less commonly used, but useful
11. In this context, it is very important to under- strategy is to also measure the value at predefined
stand that the resulting fibre tracts are virtual points or along the length of the bundle. Such
8 Strategies and Challenges in DTI Analysis 163

Fig. 8.9 An example of a


tractography analysis. A
sagittal slice is selected (a)
to draw a seed region for
tractography (b). Diffusion
measures can then be
calculated from the
resulting tracts (c)

tract profiles or distributions may reveal more • As typically only very few ROIs are necessary
localized differences that are lost when averaging to calculate the tracts, it is in general more
over the length of the tract. Some people refer to reproducible compared to ROI-based methods.
this as “tractometry” [4].
An example of a tractography analysis is
shown in Fig. 8.9. Starting from a sagittal color- Limitations
encoded FA slice (Fig. 8.9a), a region of interest • Requires a prior hypothesis about where dif-
is drawn as a seed region for tractography (Fig. ferences could be found as DTI measures
8.9b). The resulting tracts, in this case a represen- will only be analyzed in the tracts that are
tation of the splenium of the corpus callosum, are reconstructed.
shown in Fig. 8.9c. Diffusion measures can then • Tract reconstructions depend on many
be extracted from these tracts and compared parameters.
across subject groups or correlated with clinical • Tractography results are often affected by the
or neuropsychological scores. “crossing-fibre” problem.
Region specific analysis of DTI data using • In non-automated methods, the use of manu-
tractography has the following strengths and ally defined ROIs for tract selection means
limitations: that tractography results are observer depen-
dent. Ideally, clear guidelines should be fol-
Strengths lowed regarding ROI placement.
• In comparison to whole brain tractography or • Noise and other artifacts affect tract recon-
histogram analyses, more regionally specific struction, and therefore the selection of voxels
information is obtained. that will be used in the analysis.
• Tractography provides an intuitive way of • The selection of many tracts increases the
reconstructing 3D virtual representations of number of statistical tests that are performed
white matter bundles in vivo using diffusion and therefore correction for multiple compari-
information. sons is required.
164 W.V. Hecke and L. Emsell

• Pathology can affect the tractography result, Typically, a voxel-based analysis pipeline
again potentially creating a bias. consists of the following steps:
• There is no ground truth to validate tractogra-
phy results. 1. Selection of the atlas/template space to which
all data will be aligned
Note that region-specific analyses can also be 2. Alignment of all data to this atlas using global
performed using automated approaches and and local registration methods
templates/atlases. This will be discussed in the 3. Smoothing of the aligned data sets
next section on voxel-based analysis. 4. Statistical analysis in every voxel

For each of these steps, there are a number of


Voxel-Based Analysis choices to be made, both in terms of selecting the
appropriate approach as well as choosing the
One of the advantages of region-specific analyses specific parameters that will be used. As it has
compared to a whole-brain analysis is that infor- been shown that voxel-based analysis results
mation can be obtained from specific brain areas depend on these choices, every step of the pipe-
of interest. As such, the obtained DTI measures line should be considered with care and parame-
have the potential of being more sensitive (not ter selections should be justified.
averaged out over the whole brain) as well as spe- An overview of VBA is provided in Fig. 8.10.
cific (localized changes might be related to a cer- Voxel-based comparison of FA values is per-
tain pathology). Voxel-based analysis techniques formed for two groups of subjects, each consist-
take this idea further by evaluating and compar- ing of five subjects. In Fig. 8.10c, d. Although
ing DTI measures at the smallest imaging scale these images are warped during spatial align-
possible, i.e., the individual voxel. At the same ment, the registration process ensures that voxels
time, DTI measures are compared in all voxels, in the spatially aligned images retain the same
so this analysis method could also be regarded as quantitative diffusion values as in the original
a whole-brain analysis technique. data, thereby allowing statistical comparisons to
One of the main challenges of any group be made. Depending on the type of VBA imple-
analysis, and particularly voxel-based analysis, mentation used, the warped images may be
is selecting spatially corresponding voxels smoothed, for example to increase signal-to-
across subjects to compare the DTI values. If noise in the parameter maps (smoothing is dis-
this condition is not satisfied, it does not make cussed in detail in Chap. 10). A specific voxel
sense to compare the voxel measures. The pro- with the same x, y, and z coordinates in the atlas
cess of aligning corresponding voxels in differ- space is then selected across subjects and subject
ent data sets is referred to as image registration, groups (as shown by the blue and green lines in
and is an important step in the voxel-based anal- Fig. 8.10c, d). The FA values from the different
ysis pipeline. Between-subject image registra- subjects in that voxel can be visualized by a his-
tion is especially challenging because the brains togram (as shown in Fig. 8.10e). FA values in
of different subjects can vary in size and shape at that specific voxel can then be compared statisti-
the global as well as local level. However, when cally between the groups. When statistical sig-
correspondence between images can be achieved nificance is reached, the voxel can be given a
at the voxel level, voxel-based analysis is a pow- colour, as shown in white in Fig. 8.10f). This
erful tool to analyze DTI data. Since it is highly process of statistical testing of FA values between
automated, there is no need for an a priori groups is repeated for every voxel, resulting in a
hypothesis about the location of anticipated VBA map that displays the voxels and regions in
changes, and the observer dependence of the which a statistical difference is found between
results is minimized. the groups.
8 Strategies and Challenges in DTI Analysis 165

Fig. 8.10 An example of a voxel-based analysis of two statistically (e). Statistically significant voxels are then
groups of five subjects. The original data sets (a and b) are highlighted, for example by labeling with a specific color
transformed from their native space to the atlas space (c (here, white) or by coloring according to a test statistic.
and d). Within each voxel of the registered data sets the This provides a visual map of group differences (f)
diffusion measures, such as the FA value, can be evaluated

As there are many thousands of voxels in a These “hybrid” analysis methods combine the
typical DTI parameter image, many thousands of strengths of the different analysis techniques and
statistical tests need to be performed in VBA, try to avoid specific limitations of them.
making it necessary to perform some sort of cor- Voxel-based analysis of DTI data has the fol-
rection for multiple comparisons, to reduce the lowing advantages and limitations:
number of false positive findings. The number of
statistical tests can be reduced by limiting the Advantages
analysis to, for example: • The data is analyzed at the smallest scale, i.e.,
at the voxel level.
• White matter • The whole brain is evaluated as all voxels are
• Manually drawn regions in atlas space included in the analysis.
• Specific regions, as derived from atlas • No a priori hypothesis about the location of
parcellations the expected differences is needed.
• Specific white matter tracts, by performing • The manual observer interaction and therefore
tractography in atlas space (tensor informa- the observer dependence of the results is
tion should then be available in the atlas) minimized.
166 W.V. Hecke and L. Emsell

Fig. 8.11 Capabilities and limitations of different analysis approaches

Limitations
• Results depend on the parameters that are cho- Choosing an Optimal Analysis
sen in the voxel-based analysis pipeline. Approach
• As statistical analysis is performed in every
voxel, there is a chance of false positive Unfortunately, there is no single DTI analysis
findings and multiple comparison correction approach that is optimal for evaluating diffusion
should be applied. MRI measures for all studies and purposes. As
• Diffusion measures are compared in every different analysis techniques each have their own
voxel, not in specific tracts. strengths and weaknesses, and rely on various
• Results are only meaningful when accurate assumptions, choosing the most optimal analysis
image registration can be achieved. approach for a given purpose is an important step
• Pathology and lesions can affect the results, in the DTI pipeline. For example, Fig. 8.11 pro-
especially when the location of the lesions is vides a summary of what can and cannot be done
variable across subjects. using different analysis approaches.
8 Strategies and Challenges in DTI Analysis 167

In this section, a short and non-exhaustive • Region-specific DTI analysis methods can be
overview of factors and guidelines is provided to used to evaluate the diffusion measures in areas
help select the best analysis technique(s) for dif- where changes are expected. When differences
ferent applications. Ideally, these considerations are hypothesized to be present in specific
should be made before acquiring the data. A white matter bundles, fibre tractography can
much more detailed overview of factors that need be used to reconstruct virtual approximations
to be considered when using DTI in clinical pop- of these pathways. To evaluate the diffusion
ulations can be found in Chap. 13. It is important measures in lesions or specific parts of a white
to stress that these guidelines are not prescriptive matter bundle, region of interest analysis can
and the choice of which methodology to choose be applied.
ultimately rests with the DTI user. The important • Voxel-based analysis can be used for explor-
point is that each choice should be appropriately atory studies or if no clear hypothesis can be
reasoned and justified. made about the location of the expected dif-
ferences in diffusion parameters. Recall that
in a voxel-based analysis, it is assumed that
Things to Consider before Starting the changes in the diffusion measures occur in
DTI Data Analysis similar regions of the brain in different
patients. This is unlikely to be the case in
Goal and Hypothesis many clinical populations, e.g., traumatic
brain injury.
The choice of which DTI analysis technique to • Whole-brain analysis methods can be applied
apply will depend on the general goal of using if more global diffusion changes are expected
DTI, i.e. whether the data will be used for a group or if the location of diffusion changes is het-
study in a research setting or for individual patient erogeneous between patients.
analysis in clinical practice. For example:

• In research studies, typically, a longer DTI The Study Population


acquisition can be performed compared to the
clinical routine, which can impact the selec- With regard to the study population, the follow-
tion of a DTI analysis approach. For example, ing factors should be considered:
some of the more advanced tractography tech-
niques (see Chaps. 11 and 20) require the • Population composition: Can the patient group
acquisition of a large number of diffusion- be regarded as one homogeneous group, or
weighted images acquired along different gra- does it need to be subdivided into different
dient directions. subgroups? Is there a need for a matched
• Not all DTI analysis techniques can be easily healthy control group?
applied in individual patients, e.g., voxel- • Population size: How many subjects should be
based analyses. included in each group in order to be able to
• The use of DTI for an individual patient in draw meaningful conclusions? This will depend
clinical practice requires the use of CE/FDA on the magnitude of expected differences or
approved software, thereby limiting the pos- changes in diffusion parameters. For example
sible DTI analysis options. is the amount of change likely to be statistically
or visually detectable given the unavoidable
The presence or absence of a specific hypoth- presence of noise or artifacts in the data?
esis about the nature and/or location of the • Population characteristics: Different factors,
expected diffusion changes can also influence the such as age, gender, IQ, handedness, etc. may
selection of an appropriate analysis technique. affect the diffusion measures. Different subject
168 W.V. Hecke and L. Emsell

groups should therefore be carefully matched on the data quality, which depends on which DTI
with respect to these factors. For example, if acquisition parameters are chosen. Some types of
children or elderly subjects are scanned, the analysis techniques, such as tractography are
choice of which DTI analysis technique to indeed more suited to acquisition schemes with
apply can be affected, because: more gradient directions. In longitudinal or mul-
– The DTI acquisition time may be shorter ticenter studies, the scanner performance and
and data quality may be affected by acquisition parameters should be monitored.
increased subject motion. A DTI hardware phantom is useful for quantify-
– Of differential rates of brain structural change ing data quality over time and across centers.
due to development or neurodegeneration.
– Image registration of DTI data from chil-
dren or elderly to an adult atlas can The Resources
introduce errors, which will therefore
impact analysis techniques that don’t make The following resources should be considered
use of appropriate population atlases [5]. with regard to DTI analysis:
• Population pathology: The presence and
nature of brain lesions can complicate DTI • People: Which clinical and technical/software
analysis by distorting normal anatomy (see expertise is present or needed to perform the
Fig. 8.6), and hindering image registration and analysis and interpret the results?
tractography. The degree to which analysis • Time: How much scan time is available to
will be affected or the affect on method selec- obtain the DTI data? Is there time to evaluate
tion will depend on: different analysis approaches and compare
– If the lesions are focal or diffuse the results, or perform labour-intensive anal-
– The size and location of the lesion/s ysis methods such as a region of interest
– The number of lesions analysis or non-automated fibre-tracking? Is
– Variability of location across patients there time to run complex computational
– In addition to the presence of lesions, neuro- processes that may take hours or several
degeneration can affect DTI analysis and days to complete, or are results required
interpretation, because of: immediately?
– The increased presence of partial volume • Software and hardware for analysis: Will the
effects data be processed on the scanner or off-line on
– The challenges associated with image reg- a separate computer or server? Which soft-
istration to a healthy adult atlas ware will be used?
• Money: Is there money available to buy specific
software packages or licenses, to acquire enough
The Data Acquisition data sets, or to outsource part of the analysis?

Data quality and data analysis are affected by the


choice of DTI acquisition parameters such as the:
Selecting an Optimal DTI Analysis
• Number of diffusion directions Approach
• Image resolution
• b-Value When faced with so many factors to consider, it
• Number of b-values is easy to become overwhelmed with choices
• Number of averages and lose sight of the reason for acquiring DTI
data in the first place. The decision scheme in
For example, the tensor estimation, tractogra- Fig. 8.12 therefore aims to guide the DTI user in
phy result and image registration result depend choosing which type of analysis technique to
8
Strategies and Challenges in DTI Analysis

Fig. 8.12 A decision scheme to assist with DTI analysis method selection based on the aim of the DTI study
169
170 W.V. Hecke and L. Emsell

use according to the initial goal of the DTI developers have integrated the continuously
investigation. evolving theoretical methods that underlie DTI
It is important to stress that this decision data processing into their applications, as well
scheme does not provide a complete and compre- as the way their code interacts with different
hensive overview of all the relevant questions software programs and operating systems. Not
that could be asked when choosing a DTI analy- only does this add to the challenges of interpret-
sis method, nor does it provide formal solutions ing findings, but means that it is extremely
or strict answers. Figure 8.12 should be inter- important to use the same package and software
preted as an example of how knowledge about version for the analysis of all the datasets in the
the different analysis options and their pitfalls same study. This is particularly important in lon-
can be incorporated into an informed decision gitudinal investigations and may require analyz-
process, which can assist in the selection of a spe- ing new data with older software, or preferably,
cific analysis approach. all the data with the most up-to-date software
version.
In this context, it is also important to become
Selecting a Software Package familiar with the different parameter settings and
to Analyze the Data how changing them affects the final results.
Although most packages provide reasonable
Many DTI software packages are available, all default settings, the most optimal results may
with different functionalities, ranging from data require some empirical parameter adjustment.
import, basic image viewing and processing, This is particularly relevant in VBA and
image quality correction, registration, automatic tractography-based analysis, and indeed in any
segmentation, and DTI tractography to higher analysis employing image registration (including
order diffusion modeling and advanced tractogra- data correction strategies). Chapter 11 provides
phy. Most of the packages can perform the funda- some compelling visual examples of how chang-
mental pre-processing needed for DTI analysis, ing just a single parameter can drastically alter
such as tensor estimation, and visualization of tractography results.
scalar diffusion maps and glyphs. However, the New DTI software packages and tools are
specific approaches for preprocessing, e.g., the continuously being released whilst older ones are
mathematical model for tensor estimation, and being developed to incorporate new features, bug
motion and artifact correction methods, can dif- fixes, and enhancements. For this reason, specific
fer. In addition, there are a wide range of different DTI software tools and their functionality are not
options and approaches for tractography, which listed in this chapter. Instead, we recommend
vary according to the algorithms used in the soft- consulting The Neuroimaging Informatics Tools
ware package and the parameters that can be cho- and Resources Clearinghouse (www.nitrc.org)
sen to control them. and ‘I do Imaging’ (www.idoimaging.com) web-
This inconsistency between different DTI sites which list many of the latest noncommer-
analysis tools is further complicated by the use cially developed DTI software. Details about
of different terminology for both the same and proprietary DTI vendor software can be obtained
different operations across packages. It is from the respective MRI scanner manufacturer
(unfortunately) possible to perform (apparently) applications specialists. Further details about
the same analysis using (apparently) the same DTI analysis software can be found in Chap. 13,
parameters on the same dataset and obtain dif- including topics such as data storage, export/
ferent results when using different packages [6], import and file formats, version control, and
or even using different software versions of the licensing. Figure 8.13 provides a summary
same package. This is because of (sometimes checklist of considerations related to choosing
subtle) differences in the way the software DTI analysis software.
8 Strategies and Challenges in DTI Analysis 171

Fig. 8.13 Checklist of


considerations and features
to DTI analysis software

be made. The most optimal analysis approach


Conclusion will therefore depend on the decisions made in
earlier stages of the DTI pipeline.
In conclusion, the analysis of DTI data sets forms There are many options available for analyz-
only one part of a DTI study. In each phase of a ing DTI data sets, ranging from whole brain to
DTI investigation, choices and decisions have to regional and voxel-based analysis. Knowing the
172 W.V. Hecke and L. Emsell

advantages and pitfalls of each analysis tech- Smeets for providing us with some of the images and data
sets that were used to create the figures in this chapter.
nique can help with selecting the best strategy for
a given application.
The following chapters in this section provide
a more detailed overview of the most commonly Appendix: Memory Game Solution
used DTI analysis techniques.
In Fig. 8.14, the solution to the memory game in
Acknowledgments We would like to thank Ann Van De Fig. 8.2 is provided. Was it possible to match the
Winckel, Thibo Billiet, Alexander Leemans, and Dirk pathologies with the FA maps?

Fig. 8.14 Solution to the DTI memory game


8 Strategies and Challenges in DTI Analysis 173

References 6. Burgel U, Madler B, Honey CR, Thron A, Gilsbach J,


Coenen VA. Fiber tracking with distinct software
tools results in a clear diversity in anatomical fiber
1. Soares JM, Marques P, Alves V, Sousa N. A hitchhik-
tract portrayal. Cent Eur Neurosurg. 2009;70(1):
er’s guide to diffusion tensor imaging. Front Neurosci.
27–35.
2013;7:31.
2. Mukherjee P, Berman JI, Chung SW, Hess CP,
Henry RG. Diffusion tensor MR imaging and fiber
tractography: theoretic underpinnings. AJNR Am
J Neuroradiol. 2008;29(4):632–41. Suggested Reading
3. Jones DK, Cercignani M. Twenty-five pitfalls in the
analysis of diffusion MRI data. NMR Biomed. 7. Jones DK, Knösche TR, Turner R. White matter integ-
2010;23(7):803–20. rity, fibre count, and other fallacies: the do’s and don’ts
4. Jbabdi S, Johansen-Berg H. Tractography: where do of diffusion MRI. NeuroImage. 2013;73:239–54.
we go from here? Brain Connect. 2011;1(3):169–83. 8. Cercignani M. Ch 29: Strategies for patient-control
5. Van Hecke W, Leemans A, Sage CA, Emsell L, comparison of diffusion MRI Data. In: Jones DK, edi-
Veraart J, Sijbers J, et al. The effect of template selec- tor. Diffusion MRI: theory, methods & applications.
tion on diffusion tensor voxel-based analysis results. New York, NY: Oxford University Press; 2010.
Neuroimage. 2011;55(2):566–73. p. 485–99.
DTI Analysis Methods: Region
of Interest Analysis 9
Martijn Froeling, Pim Pullens,
and Alexander Leemans

commonly has to be manually drawn, but in


Learning Points some cases it can be obtained by (semi-)auto-
mated segmentation. The chosen region can be a
• Different strategies to define regions of inter- geometrical shape (i.e., sphere, cube) or be
est (ROI) defined by the shape of the anatomical structure
• Advantages and limitations of a ROI analysis of interest. The first is faster but less precise,
• Effects of ROI size, co-registration, and statis- whereas the second option is more time consum-
tical analysis methods on results ing but in general gives more accurate results, as
will be discussed further on.
Overall the ROI analysis method is rela-
Introduction tively easy to use and is supported by most dif-
fusion tensor imaging data analysis software
The region of interest (ROI) analysis method is [1]. Its main benefit is the high sensitivity to
based on the delineation of predefined areas of small changes of the parameters of interest [2].
the image and is a commonly used method for Additionally the method requires only little
quantitative analysis of diffusion tensor imaging technical know-how compared to other tech-
data. A ROI is defined as a selected area of an niques discussed later on in this and following
image from which the individual or average pixel chapters. However, it also has numerous draw-
values are extracted for further analysis. The ROI backs. ROI analysis is very time consuming
and a clear hypotheses about the location of
pathology is needed. Therefore it does not
M. Froeling, PhD (*) allow for full brain coverage and requires at
Image Sciences Institute, University Medical least a moderate knowledge of the anatomy.
Center Utrecht, Heidelberglaan 100, 3584 CX
Furthermore, even with expert knowledge of
Utrecht, The Netherlands
e-mail: m.froeling@umcutrecht.nl the anatomy and precise ROI definition the
technique is very susceptible to inter- and intra-
P. Pullens, PhD
icometrix, Leuven, Belgium user variability.
This section covers the basis of ROI analysis,
Department of Radiology, Antwerp University
Hospital, University of Antwerp, Antwerp, Belgium when to use and more importantly when not to
use the technique. When applying ROI analysis a
A. Leemans, PhD
PROVIDI Lab, Image Sciences Institute, University clear hypotheses is needed and the ROIs have to
Medical Center Utrecht, Utrecht, The Netherlands be accurately defined. Important considerations

© Springer Science+Business Media New York 2016 175


W. Van Hecke et al. (eds.), Diffusion Tensor Imaging, DOI 10.1007/978-1-4939-3118-7_9
176 M. Froeling et al.

that will be discussed are the effect of the position on a mid-sagittal slice and extended 2–3 slices
and size, ROI normalization, image registration, laterally in both directions.
and statistical analysis. ROIs can also be defined on FA or ADC
images, especially when white matter structures
are being investigated, where contrast is minimal
When to Use ROI Analysis on a T1- or T2-weighted image. In this approach
care must be taken not to fall into the trap of cir-
Because of its good sensitivity, ROI analysis is cular reasoning, because drawing of the ROI is
best performed when a clear hypothesis is pres- not independent of the studied data. This
ent about the expected differences in white approach can also be taken when investigating
matter in a well-defined region of the brain. As pathologies on ipsi- and contralateral sides of the
stated in the introduction, the region can be brain as is shown in the next paragraph.
defined by anatomical structure (e.g., corpus Some pathologies, stroke for instance, are
callosum, amygdala), pathology (stroke, lesion, clearly visible on a trace or ADC map, but not on
tumor, etc.), geometry (sphere, cube, etc.) or other modalities, as is shown in Fig. 9.1. In this
input from another modality, e.g., fMRI. The case the trace or ADC map is the obvious choice
ROI should not be too large in size, because of for ROI definition. By mirroring the ROI to the
statistical reasons explained further on in this contralateral side of the brain, DW metrics can be
chapter. ROI analysis is especially useful in studied in both affected and healthy tissue.
regions where there are lesions, e.g., tumors. In When the researcher is interested in a sub-part
these cases tract based analysis (TBA) might of a certain structure and wants to have control
be impossible due to the lack of normal fiber over the size of the ROI, a geometrical ROI (cir-
pathways. Furthermore, registration of a brain cle/sphere, square/cube, etc.) could be used. The
with lesions to a standard brain atlas may be ROI is then placed in the center of the structure
difficult or flawed due to deviations from the and has the same size in each subject, in contrast
normal anatomy.

When Not to Use ROI Analysis

ROI analysis can clearly not been used if there is


no hypothesis about the location of the effects in
the brain. If structural data are absent or of poor
quality, other methods such as extraction of diffu-
sion metrics from fiber bundles, histogram analy-
sis [3, 4], voxel-based analysis [5], or TBSS [6]
might be better suited, as will be discussed in the
next chapters.

Well-Defined Regions Fig. 9.1 Multimodality data from a stroke patient, 7 h


since last seen normal (LSN). Note the difference in con-
trast between different modalities. In this case, the DWI
To define a ROI usually the region is drawn by and ADC maps provide excellent contrast for ROI defini-
hand on a structural MRI image (T1/T2 tion of the stroke area: hyperintense on DWI, hypointense
weighted). To the investigator (“anatomist”) it on ADC. DWI diffusion-weighted image, ADC apparent
should be clear what the borders of the regions diffusion coefficient, T2WI T2-weighted image, CBF cere-
bral blood flow, CBV cerebral blood volume, MTT mean
are and to what extent it should be included. For transit time, Tmax time to max in perfusion [adapted from
example, the corpus callosum could be outlined Wu et al. [7]. With permission from Wolters Kluwer Health]
9 DTI Analysis Methods: Region of Interest Analysis 177

to manual ROI delineation where ROI size is dif- with the data under study, as misalignment may
ferent for each subject. obscure region location. Once the data is correctly
The sites of activation of a BOLD fMRI aligned, diffusion measures such as FA or ADC
study can also serve as a ROI, for example to can be easily extracted from predefined regions
start tractography, and it may lead to a very-well- such as corpus callosum, fornix etc.
localized ROI in each subject. Because the main
signal is in the gray matter, it may be necessary
to dilate the ROI into the white matter [8–10].
ROI Definition

While relatively easy to implement, there are


Atlas-Based ROIs some important things to keep in mind when per-
forming ROI analysis. As mentioned before, a
There are multiple predefined atlases of white ROI can be best defined on high quality T1 or T2
matter available, and these may serve as good weighted anatomical reference images. This is to
starting points for ROI analysis. Well-known avoid bias of defining the ROI on the parameter
atlases are the JHU atlas [11] and the probabilis- map of interest, which may influence the posi-
tic Juelich atlas [12]. Data from multiple subjects tion and boundaries or the ROI. However, when
is used to create a 3-D overview of well-defined choosing this approach one has to take great care
brain regions. A key advantage of using an atlas that the ROI position on the reference images
is that it is created in a standard space (i.e., MNI, and the parameter map of interest are aligned as
Talaraich), which makes it easy to compare illustrated in Fig. 9.2. In this figure the contour
between subjects or studies. A common approach of the brain derived from the reference scan is
is therefore to register the subject’s data to the overlaid on the diffusion weighted images and
atlas data, or vice versa. When using atlases, care the FA map. Both images were acquired during
must be taken to check overlap of the regions the same scanning session, which should assure

Fig. 9.2 Examples of misalignment between different in red. This contour is overlaid on the corresponding un-
image types. Panels (a) and (e) show high-resolution weighted (b = 0, (b) and (f)) and diffusion weighted
T2-weighted images with the contour of the brain outlined (b = 1000 s/mm2, (c) and (g)) images and the FA map (d and h)
178 M. Froeling et al.

a good alignment. At first sight, the un-weighted regions of interest in the corpus callosum were
and the diffusion-weighted images seem to defined. Both the regions were based on the ana-
match well with the reference image. However, tomical reference image as well as the FA map
closer examination clearly shows misalignment. (see Fig. 9.3). Furthermore, the size of the two
This same misalignment may not be so apparent different ROIs was varied to illustrate the effect
when just looking at the corresponding FA map of partial volume effects and user bias in defining
(see Fig. 9.2d, h). the regions of interest. The results for the average
The origin of the discrepancy between the dif- FA and MD values from these different ROIs are
ferent images can have multiple reasons but can given in Table 9.1. The variation of the parame-
have a great effect on parameter quantification ters clearly emphasizes the sensitivity of the
and fiber tractography [13]. One common reason technique to ROI definition and positioning [14].
for misalignment is that the diffusion images are Small ROIs will typically be more sensitive to
usually acquired with a single-shot EPI readout, erroneous voxels within the ROI. Increasing the
which commonly has nonrigid geometric distor- ROI size will generally decrease the sensitivity to
tions due to its sensitivity to susceptibility arti- these errors, but will increase contamination by
facts (see Chap. 6). Furthermore, there can be other structures, also known as partial volume
patient motion in between acquisition of the ref- effects, decreasing the sensitivity [15].
erence image and diffusion data within the same This implies that the definition of the ROIs
scan protocol. This motion causes rigid misalign- should be done with great care and accuracy.
ment of the images Although the positioning of circles or rectangles
is fast and easy it is generally better to accurately
outline the ROI according to the shape of the
Effect of Motion and Size structure. The latter is more time consuming but
can greatly help minimize the inclusion of other
Although the distortions and offset might seem structures [16]. Another way to exclude different
negligible, one has to realize that only a small types of tissue is to exclude pixels based on diffu-
misalignment can have serious impact on the sion parameters. For example one can exclude
parameter estimation. To illustrate this point, two cerebrospinal fluid by excluding pixels with high

Fig. 9.3 Two examples of a rectangular ROI selecting the FA map (blue). The selections are shown on the
frontal and middle part of the corpus callosum. For each T2-weighted anatomical image (a and e), the un-weighted
region one ROI was drawn based on the high-resolution diffusion image (b and f), the FA map (c and g), and the
reference scan (red) and one ROI was drawn based on the MD map (d and h)
9 DTI Analysis Methods: Region of Interest Analysis 179

Table 9.1 Average values for two part of the corpus callosum for different sizes of manually drawn rectangular regions
of interest. The ROIs were drawn both on the anatomical reference images and the FA maps
Anatomy FA map
Size (pixels) FA MD (×10−3 mm2/s) FA MD (×10−3 mm2/s)
Front 3×5 0.71 ± 0.20 0.82 ± 0.44 0.81 ± 0.11 0.77 ± 0.17
5×7 0.59 ± 0.33 1.19 ± 0.82 0.77 ± 0.20 0.85 ± 0.49
7×9 0.52 ± 0.35 1.34 ± 0.87 0.63 ± 0.28 0.96 ± 0.58
Mid 7×7 0.75 ± 0.13 0.79 ± 0.11 0.80 ± 0.10 0.76 ± 0.13
9×9 0.75 ± 0.16 0.80 ± 0.17 0.75 ± 0.16 0.85 ± 0.32
11 × 11 0.75 ± 0.19 0.81 ± 0.23 0.71 ± 0.22 0.93 ± 0.47

Fig. 9.4 Color coded FA


maps overlaid on high
resolution anatomical
images to illustrate the
result of non-rigid
registration for EPI
distortion correction. The
images on the left (a and b)
show the uncorrected data,
whereas the images on the
right (c and d) show the
corrected data. The white
arrows indicate locations
where the correction of the
misalignment is clearly
visible

MD and low FA. However, with this method it is Another commonly available method is image
also possible to exclude the tissue of interest with registration (see Chap. 10). This technique is
pathology and thus affected parameters. widely available in data processing software [1].
Figure 9.4 shows an example of nonrigid regis-
tration to correct for the misalignment between
the diffusion tensor imaging data and the corre-
Registration sponding anatomical reference data. In panel A
and B one can clearly see the misalignment
There are multiple strategies to correct for the between the corpus callosum, shown in red on the
distortions of the EPI images, e.g., B0 field map- color-coded FA map, and the lateral ventricles,
ping [17, 18], point spread function mapping shown in white on the anatomical reference
[19, 20], or reversed gradient acquisition [21, image. After nonrigid registration using
22]. However, these correction methods demand ExploreDTI [23] one can appreciate the correct
an extra data acquisition prolonging scan time. alignment of these structures as shown in panel C
180 M. Froeling et al.

Table 9.2 Average values for two part of the corpus callosum based on three manually drawn regions of interest in the
corpus callosum based on T1 images before and after registration
Normal Corrected
FA MD (×10−3 mm2/s) FA MD (×10−3 mm2/s)
ROI1 0.70 ± 0.20 0.82 ± 0.33 0.76 ± 0.14 0.77 ± 0.14
ROI2 0.70 ± 0.18 0.81 ± 0.24 0.78 ± 0.12 0.76 ± 0.14
ROI3 0.67 ± 0.23 0.95 ± 0.38 0.70 ± 0.20 0.95 ± 0.34

and D. The effect of registration on parameters when the hypothesis is less strong and multiple
estimated from ROIs drawn on the anatomical regions are investigated, a correction for multiple
image are shown in Table 9.2. The ROIs were comparisons should be carried out to reduce false
drawn in the regions indicated by the white positives [30]. More specifically, when there is
arrows. In this example the FA increases and MD no effect of the null hypothesis, and a p-value of
decreases after registration. For all parameters 0.05 is used, 5 out of each of 100 comparisons
the standard deviation decreased. will falsely reject the null hypothesis (known as
alpha error or type 1 error). There are numerous
possibilities to correct for the multiple compari-
Spatial Normalization son problem. One of the most commonly used
but also the most conservative is the Bonferroni
Spatial normalization is the process of bringing correction, which treats each comparison as an
the study data into a common stereotaxic space. independent experiment. This implies that the
It is a crucial step for group analysis of MRI data p-value at which the null hypothesis is rejected
and it allows for use of a standard 3D coordinate has to be divided by the number of comparisons.
space for analysis and reporting of neuroimaging So for ten different ROIs the p-value will be
data [24]. It consists of mapping the individual 0.005 instead of 0.05, i.e., 0.05/10. The p-value
subject data to a template, for instance to the becomes even lower when multiple parameters
well-known Talairach brain [25] or the MNI tem- are compared. If FA and MD are evaluated in
plate [26]. Once the data is in common space, these ten regions, the Bonferroni threshold of sig-
ROIs can be easily compared and checked for nificance will even decrease to 0.0025, i.e.,
accuracy in size and location. An example is 0.05/20.
shown in Fig. 9.5. It is recommended not to trans- As stated before, ROI analysis is highly user
fer tensor data into a common space, because dependent as variability in ROI placement is eas-
interpolation of tensor data is not straightforward ily introduced by different observers. Secondly,
and data will be corrupted [28]. Consequently, variability might be introduced when multiple
one should only use scalar maps (FA, ADC, etc.) datasets from the same subject are analyzed by a
for conversion into a standard space. single observer at different time points. It is
therefore good practice to calculate inter- and
intra-observer agreement (for instance, using the
Statistical Analysis κ-statistic) [31]. Agreement can be calculated on
the basis of extracted DTI metrics (FA, ADC) but
The choice of which statistical method to use also, as percent overlap, on the actual ROI coor-
depends on the hypotheses and experimental set dinates. The demonstrated overlap in Fig. 9.5 for
up [29]. When there is a clear hypothesis and a multiple subjects should then be replaced by the
corresponding well-defined anatomical region, inter- or intra-observer overlap.
the ROI analysis can be very sensitive. However,
9 DTI Analysis Methods: Region of Interest Analysis 181

Fig. 9.5 An example of ROI overlap in ten healthy con- mapped into Talairach space and combined to form proba-
trols (top rows in each panel a–c) and patient (bottom bilistic maps of ROI overlap [adapted from Tamietto et al.
rows). The ROIs were drawn in native space first, and then [27]. With permission from Elsevier]

applies for areas of compromised data quality.


Summary: The Pros and Cons of ROI The delineation of ROIs can be very time con-
Analysis suming and both intra and inter-rater measures
are poorly reproducible which impacts on both
In conclusion, ROI analysis is a simple and effec- cross-sectional and longitudinal studies [32–34].
tive means to investigate white matter changes in Furthermore, ROI placement without any prior
small, well defined regions on good quality data. knowledge can lead to inaccurate ROI segmenta-
However, the technique is prone to error and not tion which will result in different degrees of par-
suitable for the investigation of structures with tial voluming. Therefore ROI analysis is highly
complex boundaries or poorly defined changes in user dependent and reliability measures need to
white matter microstructure. This particularly be calculated to assess the quality of the results.
182 M. Froeling et al.

References 17. Chen NK, Wyrwicz AM. Correction for EPI distor-
tions using multi-echo gradient-echo imaging. Magn
Reson Med. 1999;41(6):1206–13.
1. Soares JM, Marques P, et al. A hitchhiker’s guide to
18. Jezzard P, Balaban RS. Correction for geometric dis-
diffusion tensor imaging. Front Neurosci. 2013;7:31.
tortion in echo planar images from B0 field variations.
2. Cercignani M. Strategies for patient–control compari-
Magn Reson Med. 1995;34(1):65–73.
son of diffusion MR data. In: Jones DK, editor.
19. Robson MD, Gore JC, et al. Measurement of the point
Diffusion MRI theory, methods, and applications.
spread function in MRI using constant time imaging.
New York, NY: Oxford University Press; 2011.
Magn Reson Med. 1997;38(5):733–40.
3. Law M, Young R, et al. Histogram analysis versus
20. Zeng H, Constable RT. Image distortion correction in
region of interest analysis of dynamic susceptibility
EPI: comparison of field mapping with point spread
contrast perfusion MR imaging data in the grading of
function mapping. Magn Reson Med. 2002;48(1):
cerebral gliomas. AJNR Am J Neuroradiol.
137–46.
2007;28(4):761–6.
21. Andersson JL, Skare S. A model-based method for ret-
4. Young R, Babb J, et al. Comparison of region-of-
rospective correction of geometric distortions in diffu-
interest analysis with three different histogram analy-
sion-weighted EPI. Neuroimage. 2002;16(1):177–99.
sis methods in the determination of perfusion metrics
22. Chang H, Fitzpatrick JM. A technique for accurate
in patients with brain gliomas. J Magn Reson Imaging.
magnetic resonance imaging in the presence of field
2007;26(4):1053–63.
inhomogeneities. IEEE Trans Med Imaging.
5. Snook L, Plewes C, et al. Voxel based versus region
1992;11(3):319–29.
of interest analysis in diffusion tensor imaging of
23. Leemans A, Jeurissen B, et al. ExploreDTI: a graphi-
neurodevelopment. Neuroimage. 2007;34(1):
cal toolbox for processing, analyzing, and visualizing
243–52.
diffusion MR data. Proceedings 17th Scientific
6. Smith SM, Jenkinson M, et al. Tract-based spatial sta-
Meeting, International Society for Magnetic
tistics: voxelwise analysis of multi-subject diffusion
Resonance in Medicine, Honolulu; 2009.
data. Neuroimage. 2006;31(4):1487–505.
24. Evans AC, Janke AL, et al. Brain templates and
7. Wu O, Dijkhuizen RM, et al. Multiparametric mag-
atlases. Neuroimage. 2012;62(2):911–22.
netic resonance imaging of brain disorders. Top Magn
25. Talairach J, Tournoux P. Co-planar stereotaxic atlas of
Reson Imaging. 2010;21(2):129–38.
the human brain: 3-dimensional proportional system -
8. Kleiser R, Staempfli P, et al. Impact of fMRI-guided
an approach to cerebral imaging. New York, NY:
advanced DTI fiber tracking techniques on their clini-
Thieme Medical Publishers; 1988.
cal applications in patients with brain tumors.
26. Evans AC, Collins DL, et al. 3D statistical neuroana-
Neuroradiology. 2010;52(1):37–46.
tomical models from 305 MRI volumes. Nuclear
9. Mazerolle EL, Beyea SD, et al. Confirming white
Science Symposium and Medical Imaging Conference,
matter fMRI activation in the corpus callosum: co-
1993. 1993 IEEE Conference Record; 1993.
localization with DTI tractography. Neuroimage.
27. Tamietto M, Pullens P, et al. Subcortical connections
2010;50(2):616–21.
to human amygdala and changes following destruc-
10. Preti MG, Makris N, et al. A novel approach of fMRI-
tion of the visual cortex. Curr Biol. 2012;22(15):
guided tractography analysis within a group: con-
1449–55.
struction of an fMRI-guided tractographic atlas. Conf
28. Arsigny V, Fillard P, et al. Log-Euclidean metrics for
Proc IEEE Eng Med Biol Soc. 2010;2012:2283–6.
fast and simple calculus on diffusion tensors. Magn
11. Oishi K, Zilles K, et al. Human brain white matter
Reson Med. 2006;56(2):411–21.
atlas: identification and assignment of common ana-
29. Dupont WD, editor. Statistical modeling for biomedi-
tomical structures in superficial white matter.
cal researchers. Cambridge: Cambridge University
Neuroimage. 2008;43(3):447–57.
Press; 2009.
12. Eickhoff SB, Stephan KE, et al. A new SPM toolbox
30. Miller RG, editor. Simultaneous statistical inference.
for combining probabilistic cytoarchitectonic maps
New York, NY: Springer; 1981.
and functional imaging data. Neuroimage. 2005;25(4):
31. Ozturk A, Sasson AD, et al. Regional differences in
1325–35.
diffusion tensor imaging measurements: assessment
13. Irfanoglu MO, Walker L, et al. Effects of image dis-
of intrarater and interrater variability. AJNR Am
tortions originating from susceptibility variations and
J Neuroradiol. 2008;29(6):1124–7.
concomitant fields on diffusion MRI tractography
32. Astrakas LG, Argyropoulou MI. Shifting from region
results. Neuroimage. 2012;61(1):275–88.
of interest (ROI) to voxel-based analysis in human
14. Pajevic S, Basser PJ. Parametric and non-parametric
brain mapping. Pediatr Radiol. 2010;40(12):
statistical analysis of DT-MRI data. J Magn Reson.
1857–67.
2003;161(1):1–14.
33. Chanraud S, Zahr N, et al. MR diffusion tensor imag-
15. Vos SB, Jones DK, et al. Partial volume effect as a
ing: a window into white matter integrity of the work-
hidden covariate in DTI analyses. Neuroimage.
ing brain. Neuropsychol Rev. 2010;20(2):209–25.
2011;55(4):1566–76.
34. Mukherjee P, Chung SW, et al. Diffusion tensor MR
16. Jones DK, Cercignani M. Twenty-five pitfalls in the
imaging and fiber tractography: technical consider-
analysis of diffusion MRI data. NMR Biomed.
ations. AJNR Am J Neuroradiol. 2008;29(5):843–52.
2010;23(7):803–20.
DTI Analysis Methods: Voxel-Based
Analysis 10
Wim Van Hecke, Alexander Leemans,
and Louise Emsell

between individual images due to anatomical


Learning Points variation and pathology.
• Tract-based spatial statistics (TBSS) is a popu-
• VBA is a technique that evaluates local voxel- lar type of VBA that evaluates changes in a
wise differences across the whole brain based on skeleton comprising a limited amount of white
a multistep pipeline, which includes spatial nor- matter, in order to increase sensitivity by reduc-
malization (or image registration) to a template ing registration error and partial volume effects.
or atlas, smoothing, and statistical analysis.
• VBA is most useful for investigating group
differences in DTI measures in an exploratory
manner, without the need for specific a priori An Introduction to Voxel-Based
hypotheses about the location of potential Analysis
alterations in DTI measures.
• VBA assumes that the spatial location of vox- Voxel-based analysis (VBA) of diffusion tensor
els is equivalent between subjects and is imaging (DTI) data is an exploratory technique
therefore fundamentally dependent on image to evaluate differences/changes of diffusion
registration to correct the inherent mismatch metrics in every voxel of a brain data set. In
essence, VBA investigates DTI measures at the
smallest scale possible, i.e., the voxel level, and
W. V. Hecke, PhD (*)
icometrix, Tervuursesteenweg 244, as these measures are compared in every voxel,
Leuven 3001, Belgium VBA also simultaneously evaluates the data at
Department of Radiology, Antwerp University the largest scale, i.e., the whole brain. As a
Hospital, Wilrijkstraat 10, Edegem, 2650 result, unlike with region-of-interest (ROI) anal-
Antwerp, Belgium ysis, VBA does not require an “a priori” hypoth-
e-mail: wim.vanhecke@icometrix.com esis about precisely where in the brain
A. Leemans, PhD differences may be found. This makes it an
PROVIDI Lab, Image Sciences Institute, University interesting analysis approach when there is a
Medical Center Utrecht, Utrecht, The Netherlands
clear hypothesis that there are potential differ-
L. Emsell, PhD ences in DTI parameters somewhere in the
Translational MRI and Radiology, University
Hospital Leuven, KU Leuven, Leuven, Belgium brain, but the location of such differences is not
known in advance. Although VBA indeed has
Universitair Psychiatrisch Centrum (UPC),
KU Leuven, Leuven, Belgium many advantages compared to standard ROI or

© Springer Science+Business Media New York 2016 183


W. Van Hecke et al. (eds.), Diffusion Tensor Imaging, DOI 10.1007/978-1-4939-3118-7_10
184 W.V. Hecke et al.

tractography-based approaches, it also has many general, the gross anatomy of the brain is very
limitations, which one should be aware of before similar across the (healthy) population, and all
embarking on any VBA study. brain regions are present in more or less the same
The aim of this chapter is to provide the reader spatial position across individuals. Nevertheless,
with an overview of the different processing steps as a result of natural anatomical variation, there
that need to be performed for a voxel-based analy- remain clear differences in the size and shape of
sis of DTI data, whilst emphasizing potential different brain regions (e.g., due to age, gender,
sources of error or specific challenges associated or pathology). Therefore, if we attempt to com-
with each step. Along the way we will address pare the same voxel in one person to the equiva-
common questions posed by those wishing to start lent location in another person without accounting
their first DTI VBA, for example: When is VBA a for this normal variation, we will fail. And if we
good option for the analysis of your data? What fail, we also violate one of the main assumptions
assumptions underlie the method? What are the of the VBA method. Luckily, we can try and
potential pitfalls in each processing step? Why is overcome this problem.
image registration and template selection so impor- The image processing technique that aims to
tant? What is smoothing? What is multiple hypoth- correct for differences in brain structure by chang-
esis testing?, and do I really need to worry about it? ing the size and shape of the brain image as well as
(yes you really do!) What is the difference between its local structure is called image registration. The
VBA and tract-based spatial statistics (TBSS)? end result of the image registration step is thus a
And how do I interpret the resulting findings? brain image that has been warped to match another
It is important to remember that the most opti- image, and in which voxels with the same spatial
mal results will only by obtained by considering coordinates represent the same voxel of the same
the many possible options that are inherent in con- brain structure of both images. If images cannot be
ducting a VBA study of DTI data, before you start aligned to each other well, it makes no sense com-
the analysis. paring quantitative DTI values on a voxel level.
Now one may pose the question: to which
image will we align all our data sets? This image
Summary Points to which all data sets are registered is called the
atlas or template, and many strategies and options
• VBA is a technique that evaluates local voxel- exist for this template selection.
wise differences across the whole brain. Once all data sets are located in the same atlas
• It is most useful for investigating group differ- space, smoothing is typically applied, in order to
ences in DTI measures. increase the power of the statistical tests that are
• It can be used in an exploratory manner, without subsequently performed in each voxel. There are
the need for specific a priori hypotheses about other reasons why you may wish to smooth the
the location of differences in DTI measures. data and different choices have to be made
regarding the type and extent of smoothing used.
This will be discussed later in this section.
From Individual Data Sets to VBA Statistical analysis is then performed on
Group Results: The Different Steps these warped, smoothed images. The results of
these statistical tests, with or without correction
As VBA compares diffusion metrics, such as for multiple comparisons, are then displayed with
fractional anisotropy (FA) or mean diffusivity a color on the different slices of the atlas image,
(MD), between subjects at the voxel level, one of thus providing a global view of where in the brain
the main assumptions of VBA is that the DTI the DTI measures are statistically different
information located at a specific voxel is com- between groups of subjects.
pared equivalently in each individual. In other In the following sections we will dive a bit
words, the anatomical location of a particular deeper into these different steps of the VBA pipe-
voxel should be the same for each subject. In line and highlight some options and limitations.
10 DTI Analysis Methods: Voxel-Based Analysis 185

Fig. 10.1 Overview of different steps in the VBA pipeline of DTI data
186 W.V. Hecke et al.

A theoretical example of the VBA pipeline is Image Registration


displayed in Fig. 10.1. The DTI data of six sub-
jects, three in each group, are registered to a DTI Introduction
atlas. Once all data sets are located in the same
space, FA values can be compared between both One of the main assumptions of VBA is that the
groups in each voxel. Note that in this example, same voxels in different images are aligned to
all data sets are perfectly aligned to the atlas, each other. Only in this case, can DTI measures,
which is an important assumption, but also an such as FA or MD, be compared between the
ideal situation that is not realistic. same voxels of different subjects. Note that the
process of spatially matching different images is
frequently described using different terms, such
Summary Points as normalization, warping, aligning, registration,
coregistration, etc. But in the end, although these
• VBA assumes that the spatial location of vox- terms may differ slightly in their technical defini-
els is equivalent between subjects, tion, essentially they refer to the same concept.
• Natural variation in anatomy and pathology A simplified example of the registration con-
causes an inherent mismatch between indi- cept is shown in Fig. 10.2. The data set we want to
vidual images that can be corrected by regis- register is shown on the top left of Fig. 10.2 and is
tration to a template, also referred to as the float image, as this image
• The VBA pipeline contains three main steps: will change during the registration process. On
spatial normalization (or image registration), the top right of Fig. 10.2, the reference image,
smoothing, and statistical analysis. usually the atlas, is displayed. This is our target

Fig. 10.2 The goal of


image registration is to
match a float image to the
same coordinate space as
the reference image. This
can be done by applying
global warping together
with local alignment of
structures
10 DTI Analysis Methods: Voxel-Based Analysis 187

image for registration, i.e., we want to warp the Image registration algorithms can be subdi-
float image so that it looks like the reference vided into two broad categories: global and local
image. The result of the registration process is registration techniques.
shown on the right bottom of Fig. 10.2. This reg-
istered image is the original float image, but • Global image registration techniques apply
warped into the space of the reference image. You the same deformation field (the matrix of
can note two important characteristics of registra- numbers that defines how much a point is
tion. First, you can see that the float image is shifted), which transforms one data set to
translated (i.e., shifted in position along the cardi- another, to all voxels of that data set. This can
nal axes, up/down, forward/back, and left/right) be done by rotating and translating the data
and rotated, but also that local structures of the set, referred to as a rigid-body transformation.
float image, such as the mouth in this case, are In addition, global shearing (“stretching”) and
deformed to match the reference image. Second, scaling parameters can be added, then result-
you can note from this example that the float ing in an affine transformation.
image is spatially transformed, but that the col- • Local registration techniques determine a
ors—in DTI these are the values of the metrics, local deformation field for every voxel of the
such as FA or MD—are not changed after regis- data set, in order to match every voxel with its
tration. Thus, the goal of image registration is to corresponding voxel in the other data set.
spatially warp images in a way that corresponding
voxels are in the same location, without changing A simplified example is given in Fig. 10.3, in
the original image values of these voxels [1]. which sagittal views of the brain, including the
As the global as well as local morphology of corpus callosum, are shown. In this example, the
the brain can significantly vary between different brain of subject X (shown in red) needs to be
subjects, image registration is a challenging task. transformed to the template or atlas brain shown
In addition to natural inter-subject brain variabil- in blue. As a first step, the whole brain data set of
ity, brain morphology can depend for example, on subject X can be rotated and translated globally,
age, gender, and ethnicity. To make image regis- in order to increase the similarity with the tem-
tration even more challenging in the VBA setting, plate brain. This registration technique, visual-
brain morphology can be significantly altered by ized in Fig. 10.3 by the purple box, is referred to
the pathologies in patients that are studied. as a rigid-body transformation. Subsequently,
the resulting brain image can be scaled and
skewed globally. The combination of the rigid-
Image Registration Techniques body transformation with additional global scal-
ing and skewing is referred to as an affine
The goal of this section is to provide some basic registration (the purple and green boxes in Fig.
background knowledge of image registration. 10.3). However, in order to obtain a better match
Image registration can be considered as an opti- of corresponding voxels in different data sets,
mization problem, for which the similarity local deformation fields need to be applied to the
between two or more images needs to be maxi- globally registered data set of subject X. This
mized iteratively [2]. The image registration transformation is referred to as a non-rigid or
problem can thus be subdivided into: non-affine registration and aims at aligning cor-
responding voxels of different data sets.
• a method or algorithm used to find a maximal As aforementioned, an accurate image regis-
similarity tration result is of paramount importance for a
• an approach to measure similarity between reliable VBA result. For example, if a non-affine
images registration would not be performed, the final
registration result of subject X to the template
would include a mismatch around the corpus
188 W.V. Hecke et al.

Fig. 10.3 Overview of the


combination of global and
local image registration
techniques. To transform
the brain of subject X to
the template brain, both
global and local image
registration techniques are
necessary. The purple box
shows the rigid-body
transformation, including
global rotation and
translation. In the green
box, global scaling and
skewing are added to the
transformation, referred to
as the affine
transformation. The use of
local deformations, as
shown in the orange box
and referred to as
non-affine transformations,
allows one to align both
images on a local level

callosum in this example (see Fig. 10.4). Because


similar registration errors would be present for
the other subjects, it is clear that a voxel-wise
comparison of DTI measures would lead to unre-
liable results. Indeed, the DTI measures in voxels
of subject X are then compared with the same
measures in non-corresponding voxels of subject
Y, which might be more similar to the atlas or
contain different registration errors.
In order to optimize the image registration
algorithms, appropriate image similarity
measures need to be defined. In the end, it will
be this image similarity measure that will be opti-
mized to obtain the most optimal image align-
ment. Examples of similarity measures are the
sum of squared intensity differences (SSD), cross
correlation, and mutual information (MI). In the Fig. 10.4 Example of an affine registration result.
SSD approach, the intensity in corresponding Although both data sets are globally aligned, significant
local image registration errors can be seen in the region of
voxels of two images is subtracted and the abso-
the corpus callosum
lute value of the result is squared. The SSD is
then calculated as the sum of this squared differ- in Fig. 10.4, are visualized in grayscale intensi-
ence over all voxels in the image. A schematic ties. On the top row, the image X that was regis-
example is shown in Fig. 10.5. The sagittal views tered to the template, the template image, and the
of the affine registration result, which was shown overlay between both are displayed, showing
10 DTI Analysis Methods: Voxel-Based Analysis 189

Fig. 10.5 An example of the sum of squared differences voxel and subsequently squared. Then, the total sum in
technique to measure image similarity. The intensities of every voxel is taken. When both images would be perfectly
the registered image and template are subtracted in every aligned, the sum of squared differences would be zero

some misregistration of the corpus callosum. measure how well both images are aligned, in
In the second row, the calculation of the SSD is order to find an optimal registration.
schematically depicted, highlighting the mis- • Rigid-body registration involves only transla-
match regions. If a perfect registration could be tions and rotations, whilst affine registration
performed, the SSD measure would be zero, also includes scaling and shearing.
demonstrating optimal image similarity. • Examples of similarity measures include sum
The SSD is a simple approach to evaluate sim- of squared intensity differences (SSD), cross
ilarity between images. More advanced methods correlation, and mutual information (MI).
that can take into account different intensity val-
ues of similar structures in different images, such
as mutual information, generally produce better Registration of DTI Data
results. In addition, some specific image similar-
ity methods have been developed for DTI data, The registration of DTI data is especially challeng-
using information on tensors, statistical relation- ing. This is mainly caused by the fact that DTI
ships between measures, or anatomical informa- data, unlike anatomical MRI or CT data, contain a
tion. In contrast to typical grayscale MRI images, tensor in each voxel, which also represents orien-
DTI data contain more information in each voxel; tational information. Taking this tensor informa-
therefore similarity measures can be optimized to tion into account can improve the registration
make use of this additional information [3, 4]. result (an overview of DTI registration methods is
provided in [5]). In the following paragraphs, we
will describe several challenges in more detail.
Summary Points
DTI Registration Challenge 1:
• To register two images you need (a) a model Reorientation
(global and/or local) to warp the floating The tensor is directionally dependent and contains
image to the reference image, and (b) a way to orientational information about the underlying
190 W.V. Hecke et al.

white matter microstructure. When transforma- containing three voxels. Consider that in the orig-
tions are applied to align data sets, a correction inal DTI data set of subject X, this bundle runs
(i.e., a tensor reorientation strategy) needs to be vertically. The DWI image intensities of the three
applied to ensure that the directional DTI informa- voxels in that bundle are high for the DWI that
tion is still accurate. was acquired with a diffusion sensitized gradient
The need for tensor reorientation after image perpendicular to the bundle, and low for the DWI
alignment of DTI data or during iterative regis- acquired with a diffusion sensitized gradient par-
tration processes is explained in Fig 10.6. To sim- allel to the bundle. For simplicity, only two DWIs
plify things, the concept of reorientation is are considered here. The corresponding white
explained for only one white matter fiber bundle, matter bundle of the template image, however,

Fig. 10.6 Simplified


overview of the tensor
reorientation problem in
DTI. Tensors indeed need
to be reoriented after image
registration in order to be
aligned with the underlying
microstructure
10 DTI Analysis Methods: Voxel-Based Analysis 191

contains some curvature. As a result, the DWI necessary to reorient the tensors if only the rota-
intensities for the different gradients are different tionally invariant, quantitative DTI information
in these voxels. is used in the subsequent analysis.
It is now assumed that we can align both white
matter bundles perfectly, i.e., the corresponding DTI Registration Challenge 2:
voxels of the white matter bundle are perfectly The Tensor Information
registered. The resulting deformation field is then DTI image registration can be optimized by using
applied to the DWIs and the tensor is recalcu- information from other modalities, such as ana-
lated. Now, if we refer to Fig. 10.6 again, we see tomical MRI, or by incorporating scalar and ten-
that the registration process changes the spatial sor information. Park et al. [6] compared the use
location of voxels in order for them to match, but of different input images on the overall registra-
not their values or image intensity. However, if tion result of DTI data. They evaluated registra-
only the spatial location of the voxels is changed, tion results after using T2-weighted images, FA
and not their image intensities, the directional images, the difference of the first and second ten-
diffusion information, and therefore the tensor, sor eigenvalues, FA and the tensor trace, all three
are not changed compared to the information in tensor eigenvalues, and finally the six indepen-
native space. Indeed, as explained in Chaps. 4 dent tensor components [6]. In this study, it was
and 6, the image intensities of the different DWIs demonstrated that the use of the six independent
and the values of the tensor are related to the ori- tensor components as input channels performed
entation of the white matter bundle. most optimal in aligning the tract morphology
As a result, the tensor information of the reg- and tensor orientation. This was further con-
istered data set of subject X to template space firmed by other studies [7].
thus no longer reflects the underlying microstruc-
tural white matter information, as can be observed Scalar Anatomical MRI Information,
in Fig 10.6. In order to correct for this, Alexander Such as 3D T1-Weighted Images
and Gee [4] proposed different methods, referred Using scalar anatomical MRI information to
to as the “finite strain” and the “preservation of determine the deformation field between DTI
principle directions” approach. The finite strain data sets is similar to the approach used in func-
method decomposes the transformation matrix in tional MRI analysis. First, the DTI data set is
a deformation and a rotation component, where- transformed to the 3D T1-weighted image of the
after only the latter is used to reorient the tensors. same subject, using a rigid-body or affine trans-
However, shearing, nonuniform scaling, and formation. The DTI information used for this
stretching factors affect the orientation as well. registration is normally the b0 or non-diffusion
Together with the rotational component, they are weighted image, as this image mostly resembles
taken into account in the preservation of principal the anatomical image. Thereafter, the 3D
direction strategy. T1-weighted image is aligned to a T1-weighted
When a global, i.e., rigid-body or affine, regis- atlas, such as the Montreal Neurological Institute
tration method is applied, the same reorientation (MNI) template. The resulting deformation field
is applied to all voxels. However, in the case of is then applied to the DWIs, which were trans-
non-rigid registrations, the local transformation formed into the space of the T1, or to the trans-
matrix, which can be different for each voxel, is formed quantitative DTI maps directly.
used to calculate local tensor reorientation. The advantages of this approach are:
It is important to note that tensor reorientation
approaches do not affect the rotationally invariant • T1-weighted atlases can be used.
quantitative DTI measures, such as the eigenval- • Many open-source software packages support
ues, the FA, or MD. As they are rotationally this type of algorithms, such as SPM (www.
invariant, reorienting the tensor does not change fil.ion.ucl.ac.uk/spm), FSL (www.fmrib.ox.
their values. In a VBA analysis, it is therefore not ac.uk/fsl), AFNI (afni.nimh.nih.gov/afni).
192 W.V. Hecke et al.

However, using this approach is not optimal registration process in order to increase registra-
for DTI data, for several reasons: tion quality. For example:

• The unique white matter DTI information is • By including several channels of scalar image
not used to guide the registration. The image information. Guimond et al. [9] and Park et al.
intensity of white matter is uniform on [6] have used different channels of input infor-
T1-weighted images, which can result in mis- mation, such as the T2-weighted image inten-
matching of different fiber bundles [6, 8]. sity, fractional anisotropy, trace of tensor, and
• As DTI data sets are usually acquired using an eigenvalues.
EPI sequence, different artifacts are present • By using the scalar information from the
compared to the T1 image. For example, geo- whole tensor to align two DTI data sets and
metric distortions due to eddy currents and detect correspondences between them [6, 7,
susceptibility. This results in misregistration 10–13]. In addition to using the tensor ele-
between the b0 image and T1-weighted image. ments, DTI feature vectors can be derived and
used to drive the registration ([14–16]).
• By using DTI tractography or other connectivity
Scalar DTI Information, Such as FA or information to guide image alignment [17–20].
MD Maps
In this approach, the scalar DTI information, It has been demonstrated that the use of DTI-
such as contained in FA or MD maps, is used as specific information with multiple channels results
input information to guide the registration. As in more accurate registration of DTI data. As an
a result, no anatomical data sets are involved, accurate image alignment is one of the most impor-
and registration is directly performed based the tant assumptions in VBA, including DTI informa-
DTI information. Compared to using anatomi- tion during the registration will increase the
cal information, this method has several reliability of the VBA results [6, 7]. However, some
advantages: drawbacks of this approach should be mentioned:

• Some white matter information (as present in • As this approach is more complex compared
FA or MD maps) is used to increase the regis- to the scalar registration methods, computa-
tration accuracy. tion time is increased.
• The DTI information is directly aligned to an • There is a need for tensor information in atlas
atlas and no anatomical MRI image is needed. space, as this tensor information is needed to
drive the registration.
Although this will increase registration accu-
racy, this approach has some drawbacks:
Summary Points
• FA and MD values do not always discriminate
fiber bundles that are located close to one • In order to achieve successful DTI registra-
another, potentially resulting in misregistra- tion, the orientational dependence of the diffu-
tion of these bundles. sion data needs to be accounted for, i.e., any
• There needs to be an FA template to align the rotation of the DWIs should be corrected for,
subject data to. e.g., by using tensor reorientation during
affine registration.
• Registration can be improved by incorporat-
Diffusion Tensor Information ing diffusion information, such as scalar DTI
Many approaches have been proposed that measures (e.g., FA/MD), tensor information,
incorporate the specific DTI information into the or a combination of different data types.
10 DTI Analysis Methods: Voxel-Based Analysis 193

Atlas or Template ICBM space, containing fiber orientation maps


and white matter parcellation maps [27]. Peng
The atlas or template is the reference frame to et al. [28] and Zhang et al. [21] created a DTI
which all data are registered and which is used to atlas in the ICBM-152 space by registering high
report the results. It has been demonstrated that quality DTI data sets of 67 healthy subjects using
the choice of this atlas or template can affect the a non-affine registration procedure.
VBA results [21–23]. As a result, the atlas selec- One of the main advantages of using standard
tion is an important step in the VBA processing templates is that they provide the possibility to
pipeline for DTI data. An overview of possible make use of predefined anatomical regions for
DTI atlas selection approaches is provided in subsequent region-of-interest analysis in atlas
Zhang and Arfanakis [23]. space. Furthermore, as the standard templates are
DTI templates can be subdivided into two widely used, results, and coordinates of signifi-
broad categories: cant findings in particular, can be easily com-
pared across studies.
• a standard template, However, there are some drawbacks when
• a population-/study-specific template using a standard template. First, as most standard
templates are created from healthy subject data,
A population- or study-specific template is they do not necessarily represent an average brain
usually constructed based on the data sets that are of the subjects of your study, especially when
analyzed. As a result, in theory, this atlas is the pathology is present in some subjects. A simpli-
best representation of these data sets, and should fied example is given in Fig. 10.7. Assume again
thus result in minimizing the registration errors. that DTI measures are compared between healthy
A standard template, on the other hand, is an atlas subjects and patients with Alzheimer’s disease.
that was created from a group of healthy subjects Although some atrophy can be present in the
and is independent from the subject data sets that healthy control group, it will be more severe in
need analyzing. However, these atlases usually the Alzheimer’s group. As a result, when all data
contain anatomical labels and predefined region is transformed to a standard atlas of a healthy
of interests, which might be of interest for the brain, the registration result will be much better
study. So, again, there is no single correct for the healthy subject data compared to the
approach of selecting an optimal template for Alzheimer subject data. This is especially notable
your study. The optimal choice will depend on at borders with CSF (shown in black in Fig.
your study and data (i.e., goals, hypothesis, 10.7), where the groups will not be matched cor-
patient population, data quality). In the following rectly. Not only will there be registration errors
paragraphs, a more detailed description of the (something we don’t want in VBA), there is also
template selection choice is provided, including a bias towards a certain subject group. As a result,
some advantages and limitations of the different this will create false positive findings, caused by
approaches. more significant misregistration in one of the
subject groups in specific brain regions.
Standard Template Another limitation of some standard templates
Standard templates are typically constructed by is that the diffusion tensor information is not
averaging data from a group of healthy subjects always present, hence limiting the information
that have been registered to a stereotaxic atlas. that can be used to drive the registration process
For example, anatomical T1-weighted templates to this atlas.
were constructed by the Montreal Neurological
Institute (MNI) and the International Consortium Population-Specific Atlas
of Brain Mapping (ICBM) [24–26], and are The general idea of population- or study-specific
widely used in functional MRI research. Mori atlases is to use the data sets that are studied to
et al. created the first standard DTI atlas in the determine an atlas space, to which all data sub-
194 W.V. Hecke et al.

Fig. 10.7 A simplified


example of image
registration of healthy
subjects and subjects with
enlarged ventricles to a
healthy subject atlas.
Registration errors can
occur in the group with
enlarged ventricles, thereby
introducing a potential bias
in the VBA results

sequently are registered. The simplest way to The advantage of selecting an individual sub-
construct a study-specific atlas is to select one ject of the study as the template is that the data
DTI data set from the study population as the quality of the template image is similar to the
template. This subject can be chosen randomly, data quality of all other subjects. In addition, ten-
based on visual inspection of all data or based on sor information is present in this atlas and can
calculations that determine which subject is therefore be used during the registration of all
most representative for the population. In the lat- data sets to this atlas. However, this approach
ter case, all data sets are registered to each other also has some limitations. As discussed, select-
and the deformation fields from one subject to ing the most representative subject is not trivial.
all others are averaged. The subject with the In addition, in the case of an individual subject
smallest average deformation field to all other atlas, there is information on predefined anatomi-
subjects can then be regarded as the most repre- cal regions, as is the case in standard template
sentative subject of the population under study. spaces. Similar as with the standard templates, a
10 DTI Analysis Methods: Voxel-Based Analysis 195

bias can be introduced as the selected subject can • A population-specific atlas is created only
be a patient or a control subject. from subjects under investigation and is less
Instead of selecting an individual subject as subject to misregistration bias.
the atlas space, population-specific atlases can be
constructed based on the whole population that is
studied [29, 30]. The resulting atlas should then Smoothing
be the best average representation of all the data
that is being analyzed. Van Hecke et al. [22] dem- What Is Smoothing?
onstrated that the accuracy of VBA results can be Smoothing involves blurring the data using a fil-
improved when using a population-specific atlas ter, typically a Gaussian kernel. As a result, the
compared to the use of a standard template. image value in each voxel is recalculated, based
As mentioned, the main advantage of the on the weighted values of neighborhood voxels,
population- or study-specific atlas is that it is the as determined by the kernel. Typically, the size of
best representation possible of the data sets that this kernel is defined by the full width at half
are studied (when the appropriate approach and maximum (FWHM). The FWHM is an indication
registration techniques are used). As a result, of the distribution of the kernel values, meaning
image registration accuracy will be maximized that when the FWHM is 4 mm, the kernel is
and unbiased to the subject group. By construct- 4 mm wide at 50 % of its peak value. Consider
ing a study-specific atlas, registration errors to the example given in Fig. 10.8.
the atlas can still be present, but they will be In Fig. 10.8a, an axial FA slice is shown that
unbiased towards the subject groups. Another will be smoothed by a Gaussian kernel. As an
advantage of a study-specific data set is that it can example, we focus on a row of voxels, as shown
be made with all tensor information present. This in Fig. 10.8b. Note that in this example, we
provides the opportunity to use the tensor-based explain Gaussian smoothing in a single row of
information during registration, again improving voxels in the x direction, whereas in practice the
registration accuracy; or to perform tractography voxels in the y and z direction will also be taken
in the atlas space. into account. In Fig. 10.8b, the FA values of the 9
As with the individual subject atlas, an impor- voxels of interest are displayed. The FA value of
tant limitation of the population-specific atlas is the middle voxel is depicted in red, as the value
that it does not contain anatomical labels and of this voxel will be changed during the smooth-
delineated regions, in contrast to the standard ing in this example. Of course, in practice this
templates. In addition, as it is made from the data process is repeated for all voxels. The Gaussian
of a specific study, it is usually (though not neces- kernel that will be used for smoothing is shown
sarily) constructed from fewer data sets com- in Fig. 10.8b. Note that the FWHM of this kernel
pared to the standard templates. is 6 mm, as we assume a voxel to have a width of
2 mm. The FA values of the different voxels will
be weighted, whereby the weighting factor is
Summary Points determined by the Gaussian kernel. The total sum
of the weighting factors thereby equals 1. The
• In order to compare DTI values between resulting weighting factors for the different vox-
groups, individual datasets need to be regis- els for this Gaussian kernel are shown in Fig.
tered to a common template space or atlas 10.8c. Next, the FA value of every voxel is multi-
• Standard atlases are created from large num- plied by the corresponding weighting factor (see
bers of subjects and are useful for reporting Fig. 10.8d), and the resulting sum of these values
results in a commonly used and well-defined will be the FA value middle voxel in the smoothed
space. Standard atlases are less suitable for image, in this case an FA of 0.664. As mentioned,
subjects with gross morphological differences this process is repeated for all voxels, thereby
to the standard template. taking all neighboring voxels (in all dimensions)
196 W.V. Hecke et al.

why would you smooth your data? Why would


you bother blurring images when you pushed
your scanner and patients to the limits to acquire
high resolution data sets? There are several rea-
sons why DTI data sets are smoothed before sta-
tistical testing in VBA:

• It helps to accommodate for imperfect


registration.
• It reduces the noise and increases SNR.
• It makes the data more normally distributed.

However, an obvious limitation of smoothing


is that the resulting data is blurred. In addition, by
smoothing the data, information of different
white matter structures and tissue types (white
matter vs. gray matter vs. CSF) will be averaged.
Although it does make sense to integrate infor-
mation from different neighboring voxels of the
same white matter structure, averaging informa-
tion from other structures or tissue types can
introduce false positive as well as false negative
results.

Determining the Smoothing


Kernel Size
An important parameter related to smoothing is
the smoothing kernel size. However, it is not
straightforward to determine the optimal size of
the smoothing kernel for a specific study. To
complicate matters further, it has been demon-
strated that the choice of the kernel size can sig-
nificantly affect the VBA results [31, 32]. This
stresses the importance of selecting the optimal
Fig. 10.8 A simplified example of the process of image kernel size, or at least having a clear argument for
smoothing in one dimension. An FA map (a) is smoothed using a specific smoothing kernel width.
by a Gaussian kernel with an FWHM of 6 mm (b). In (c)
the different weighting factors are shown. Finally, the So, is there a way of determining an optimal
resulting FA value after smoothing is calculated (d) and smoothing kernel size? According to the matched
the smoothed FA map is displayed filter theorem, the optimum smoothing kernel
width should be similar to the expected extent of
into account. The resulting smoothed FA map is the signal difference, as the SNR then reaches its
also shown in Fig. 10.8d. maximum [33]. In other words, for DTI, an “a
priori” hypothesis is needed on the extent of
Why Should You Smooth (and change in the diffusion metrics that are expected.
Why Not)? But this shifts the problem from not knowing
In Fig. 10.8, the process of smoothing was how to choose the optimal kernel size to the prob-
explained. In this simplified example, an isotro- lem of predicting the size of the hypothesized dif-
pic Gaussian smoothing kernel was used. But ferences. After all, one of the strengths of VBA is
10 DTI Analysis Methods: Voxel-Based Analysis 197

that it is an exploratory approach to search in the


whole brain for unknown group differences.
Although similar problems exist with func-
tional MRI analysis, they may be more straight-
forward to address, as the expected differences
relate to the extent of GM brain activity in fMRI
and the size of anatomical structures in VBM.

Smoothing in DTI
The problems related to smoothing DTI data are
similar to problems in ROI analysis, as for both
methods, the size of the expected differences
should be known for an optimal result. However,
in ROI analysis the location of the differences
should also be known, which is not necessary for
VBA, as all the voxels of the whole brain are
evaluated simultaneously. On the other hand,
smoothing of DTI data in VBA has some specific
issues. These are related to the specific nature of
DTI information, i.e., white matter tract informa-
Fig. 10.9 An example of how voxel values from different
tion. These white matter tracts are aligned along structures and tissue types are taken into account during
a specific orientation and can significantly vary in smoothing with an isotropic Gaussian smoothing kernel
size and width. Smoothing with isotropic with different widths. In this example, the diameter of the
yellow circles reflects the FWHM of the Gaussian smooth-
Gaussian kernels will therefore introduce wide-
ing kernels. As can be observed, different information is
spread averaging of information across different included during smoothing for different FWHM of the
white matter bundles and tissue types. This is not smoothing kernels
desirable, as we know for example that white
matter degeneration is not present in CSF. An
example of how different kernel sizes would structure alone is increased. In addition, this
average information from different structures is approach can cope with the variations between
shown in Fig. 10.9. shape, size and width of white matter tracts
Not only can an isotropic Gaussian smoothing across the brain, as the shape of the smoothing
kernel average out signals from different struc- kernel is locally adapted.
tures and/or tissue types, this effect will also However, signal can still be averaged between
depend on their location, as white matter tracts adjacent white matter structures. In addition, this
and brain structures vary in size, shape, and width still relies on a prior hypothesis of the size and
across the brain. shape of the expected differences in diffusion
To address these problems, anisotropic metrics between groups of subjects.
smoothing kernels were introduced in DTI-based
VBA [32, 34]. In these methods, the smoothing
kernel shape is not isotropic and can vary across Summary Points
the brain. For example, the kernel shape can be
determined based on an FA map. At edges of the • Smoothing is typically performed after image
FA image, for example between the white matter registration to accommodate for imperfect
structure and CSF, the smoothing kernel will registration, to reduce the noise and increase
stop, as shown in Fig. 10.10. As a result, the SNR, and to obtain more normally distributed
chance of averaging signal in the white matter data.
198 W.V. Hecke et al.

The “Multiple Comparisons” Problem


When a statistical test is performed, a threshold—
for example p < 0.05—is used to assess whether
the result is statistically significant or not. However,
for a threshold of 0.05, there is still 5 % chance that
a type I error—i.e., a false positive result—occurs.
Although this is reasonable for a single statistical
test, it becomes problematic when thousands of
statistical tests are performed, all with a 5 % chance
of a type I error. This is known as the multiple
comparisons problem. In order to reduce the type I
error in VBA, some correction for multiple com-
parisons should be performed. When very strict
corrections, such as the Bonferroni correction
(dividing the statistical threshold by the number of
statistical tests that are performed), are applied,
typically no statistically significant differences are
found. However, there is a whole range of other
less strict methods to correct for multiple compari-
sons, the most popular being the theory of Gaussian
Fig. 10.10 Example of an anisotropic smoothing kernel.
As can be seen, the information that is included during Random Fields [35], false discovery rate [36], and
smoothing is limited to the white matter structure that is permutation-based approaches [37]. Unfortunately,
delineated in yellow. The FWHM of the smoothing kernel there is no consensus on the most optimal tech-
will affect the information included during smoothing,
nique to correct for multiple comparisons.
and thus the results, less
Different techniques are used in literature, which
makes it difficult to compare results, and in many
studies no significant findings are reported after
correction for multiple comparisons. For the latter
• The FWHM reflects the smoothing kernel size reason, many studies report uncorrected values. It
of the Gaussian kernel. is therefore important to interpret the results of
• VBA results can be affected by the smoothing studies in the context of the statistical analysis and
that is performed and the FWHM of the correction for multiple comparisons that was used.
smoothing kernel. There are several options to reduce the num-
• Anisotropic smoothing methods were intro- ber of statistical tests that need to be performed in
duced to average out information within white a VBA setting. For example, one can apply a
matter structures during smoothing. white matter mask and only evaluate the voxels
within this mask. In many studies this would
make sense, as the quantitative DTI measures are
Statistical Analysis best characterized in white matter and research-
ers are typically only interested in white matter
As with all VBA approaches, such as VBM and when DTI is used. Other approaches can be even
functional MRI, statistical comparison of DTI more limiting in the amount of voxels analyzed,
metrics is performed in every voxel. Although by deriving masks in atlas space from:
this is one of the strengths of the technique, i.e.,
an exploratory whole brain analysis at the small- • Predefined anatomical labels
est scale, it also introduces a “multiple compari- • Manually drawn regions of interest
sons” problem. • Tractography results in atlas space
10 DTI Analysis Methods: Voxel-Based Analysis 199

Parametric or Nonparametric However, VBA also has some significant


Statistics? limitations:
Parametric statistical tests, such as the typically
used t-test or regression tests, require the residu- • Results are only relevant when perfect image
als of the model to be normally distributed. registration is achieved.
However, Jones et al. demonstrated that this • Results are less observer dependent, but are
assumption only holds in around 60 % of the significantly parameter dependent.
voxels [31]. Most of the voxels in which the
residuals were not normally distributed were The latter point is very important. In every
situated in the gray matter. When DTI metrics of step of the VBA pipeline, choices have to be
smaller groups of subjects are compared, non- made, for example, regarding:
parametric statistics, such as permutation or
bootstrap based testing, should be strongly con- • the registration technique and its parameter
sidered. Jones et al. [31] also demonstrated that settings
Gaussian smoothing reduces the amount of vox- – which affine technique?
els with non-normally distributed residuals, but – which non-affine technique?
the number of voxels with non-normally distrib- – which information to drive the registration?
uted residuals remained high. – which similarity measure for registration?
• the atlas to use
– standard vs. population specific?
Summary Points • the smoothing method and the kernel width
– anisotropic vs. isotropic smoothing?
• As statistical tests are performed in all voxels, – kernel width?
false positive results can be reported. A cor- • the statistical test and method to correct for
rection for multiple comparisons should be multiple comparisons
performed to reduce the number of false posi- – parametric vs. nonparametric tests?
tive findings. – correction for multiple comparison? Which
• Nonparametric statistical analysis should be method?
considered, especially when the subjects
groups that are studied are small. Note that this is a non-exhaustive list of exam-
ples, and at each level there are even more param-
eter settings to consider!

To VBA or Not to VBA?


Why Parameter Settings Are
Pros and Cons Important?

So, given all the aforementioned limitations, The importance of the choices made at different
should we use VBA at all? The answer is not steps of the VBA pipeline has been demonstrated
straightforward. VBA has many advantages: by Jones et al. [38]. In this study, the same DTI
data sets were sent to nine different research
• It is an exploratory technique. groups. Each of these groups performed a voxel-
• DTI metrics are evaluated in the whole brain based analysis of the same DTI data set, using
and at the same time at the smallest scale with their own selected set of methods and parameters.
which one can obtain diffusion measures, i.e., The nine research groups reported different clus-
the voxel level. ters in various anatomical locations despite ana-
• It doesn’t need a lot of manual interaction, lyzing identical DTI datasets. This demonstrates
making it less observer dependent. the sensitivity of VBA to choices in the pipeline.
200 W.V. Hecke et al.

Although most VBA studies follow the proto- eton projection only recovers less than 10 % of
typical pipeline, i.e., image registration, smooth- the registration errors. As an accurate image reg-
ing, and voxel-based statistical analysis, there is istration is as important in TBSS as in classical
currently no standardization with regard to the VBA, similar care must be taken with respect to
methods and parameters that should be used. As the use of a non-affine registration method, tensor
a result, different VBA approaches and parameter information during registration, and population-
settings are used in different studies. In light of specific atlases in case subjects are studied with
the Jones et al. [38] study, comparison of results significant pathology or atrophy. It was indeed
across studies is very difficult. demonstrated by Keihaninejad et al. [41] that the
use of a population-specific atlas outperformed
the standard template or individual subject tem-
Tract-Based Spatial Statistics, plate in the study of Alzheimer’s disease.
an Alternative to VBA Although TBSS is an elegant way of trying to
overcome some of the drawbacks of VBA, as for
With the goal of optimizing VBA for DTI data all methods, there are some limitations, which
sets, tract-based spatial statistics (TBSS) was should be taken into account when performing a
introduced by Smith et al. [39]. Although this TBSS analysis. For example, as only the local
approach can also be regarded as a voxel-based maximal FA values are projected on to the skele-
analysis, some modifications from the standard ton and therefore evaluated, an inherent assump-
VBA pipeline were introduced in TBSS. The tion is made that pathology will mainly affect the
main difference between TBSS and standard local maximal FA values, which is not necessar-
VBA is the construction of “a skeleton.” First, all ily the case. TBSS is also more sensitive to
FA images are aligned to a template by using a changes in DTI measures in diagonally oriented
non-affine transformation and are subsequently tracts, as their skeleton contains more voxels than
averaged to result in a group mean FA map. From horizontal or vertical ones [42]. In addition, the
this image, a skeleton is created by selecting the presence of white matter lesions that reduce FA
locally maximal FA values, which are assumed to values will affect the results, as it is possible that
form the center of the white matter tracts. TBSS some voxels that do not belong to the core of the
then projects the FA values of each registered tract have larger FA than those in the core because
data set onto the skeleton. More specifically, the of the presence of the lesion [43]. Furthermore,
locally highest FA value perpendicular to the by limiting the analysis to local FA maxima on
skeleton in each registered FA map is then pro- the skeleton, which comprises a relatively small
jected onto the skeleton. The projection on the percentage of the total image, a lot of potentially
FA skeleton can, to a certain extent, compensate valuable information is not used in the analysis.
for potential registration errors. In addition, as Sometimes this may not be apparent as some
statistical tests are performed on the skeleton, authors choose to display their findings on an
there is no need for smoothing and less statistical artificially thickened skeleton which appears to
tests are performed compared to a standard VBA. encompass more white matter voxels than were
Although the skeleton projection step in TBSS actually analyzed. This is typically done to
can indeed correct for some local misregistration, emphasize findings, but as with tractography
it cannot compensate for larger registration errors visualizations, it can be misleading to those unfa-
that might occur. As the projection procedure miliar with the techniques (see Chap. 8). Finally,
must search locally for the highest FA value, in in regions of crossing fibers, the FA skeleton can-
order to avoid finding spurious correspondences, not be determined reliably as the FA in these
it will not be able to correct for larger misregistra- regions is typically very low. With 60–90 % of
tions [40]. Indeed, the study of Zalesky and col- white matter voxels containing multiple fiber
leagues [40] used synthetic deformations of populations, this may complicate the interpreta-
ground truth images to demonstrate that the skel- tion of TBSS findings significantly [44].
10 DTI Analysis Methods: Voxel-Based Analysis 201

How Do TBSS and Classical VBA Regardless of which technique is applied how-
Approaches Compare? ever, the quality of the inter-subject registration is
central to determining the sensitivity and accu-
Given the fact that TBSS and classical VBA racy of VBA results.
approaches differ with regard to core aspects of
voxel-based analysis, i.e., registration and
smoothing, and given we know that parameter VBA in Clinical Practice?
selection significantly affects VBA results, it is
worth exploring how TBSS results compare with When applied responsibly, with due consider-
those from classical VBA. Although most studies ation for its limitations, VBA can be a powerful
choose to apply one method or the other, a few tool to analyze DTI data from patient populations
studies have directly compared results of the typ- with neurological and psychiatric disorders.
ical VBA approach with those of TBSS when However, is it the most appropriate tool to use in
analyzing the same dataset. Sage et al. [8], for clinical practice, when a DTI data set from an
example, reported very similar results to TBSS individual patient needs to be analyzed and inter-
when an optimized VBA (in terms of registration preted? Although the most appropriate use of
method and atlas building) was performed. It was VBA is for group analysis, some authors have
also demonstrated that the VBA results were applied the technique to analyze individual
more reliable compared to the results of a non- patient data. For example, in traumatic brain
optimized VBA. Preti et al. [45] compared TBSS injury patients, Lipton et al. [47] used the
results with an atlas-based approach to obtain enhanced Z-score microstructural assessment of
DTI measures in specific tracts of healthy sub- pathology (EZ-MAP) approach to evaluate
jects and subjects with mild cognitive impair- regional FA abnormalities. In this VBA approach,
ment and Alzheimer’s disease. They concluded a patient’s FA value is compared to the FA values
that the comparison of the healthy subjects with of a normal reference group in every voxel.
the patients was similar for the atlas-based However, this requires a large reference group
approach and TBSS, but that the atlas-based and the results can depend on this reference
approach was more sensitive to detecting changes group. Kim et al. [48] suggested some improve-
between patients with mild cognitive impairment ments to overcome these problems. Patel et al.
and Alzheimer’s disease. [49] used VBA to detect FA changes in lesions
Schwarz et al. [46] evaluated the use of more and normal appearing white matter in individual
advanced group-wise registration methods on the MS patients. Although FA reductions were
accuracy of VBA and TBSS. Using synthetic observed in many regions, the authors also
data sets as well as comparing healthy subject reported abnormal FA values due to misregistra-
data with data from Alzheimer’s patients, they tion. Given its underlying assumptions and limi-
showed that the TBSS skeleton projection step tations, we would not advocate the use of standard
lowered the overall accuracy of the results when VBA (or TBSS) to analyze individual patient
the image registration was optimized. data at the present time.
In summary, both classical VBA and TBSS
can be successfully applied to study voxel-wise
differences in DTI parameters at a group level. Conclusion
Despite the widespread adoption of TBSS as a
gold standard VBA approach, it is not without The aim of this chapter was to introduce the VBA
significant shortcomings. There have been insuf- approach for DTI data, to elaborate on the differ-
ficient studies that have compared the accuracy ent steps involved, and to outline its advantages
of TBSS results with classical VBA results on the and limitations. Compared to a standard ROI or
same datasets to determine if one approach even tractography-based analysis, VBA is a more
should be used in preference to the other. automated approach and therefore less observer
202 W.V. Hecke et al.

dependent. However, many choices have to be 9. Guimond A, Guttmann CRG, Warfield SK, Westin
CF. Deformable registration of DT MRI data based on
made in the VBA pipeline with regard to image
transformation invariant tensor characteristics. In:
registration, template selection, smoothing, and International symposium on biomedical imaging.
statistical analysis, which the final VBA results Washington, DC: IEEE; 2002. p. 761–4.
will ultimately depend on. VBA should not be 10. Ruiz-Alzola J, Westin CF, Warfield SK, Alberola C,
Maier S, Kikinis R. Nonrigid registration of 3D tensor
viewed as a generic DTI analysis technique that
medical data. Med Image Anal. 2002;6:143–61.
can be applied without any hypothesis. Whether 11. Rohde GK, Pajevic S, Pierpaoli C, Basser PJ. A com-
or not VBA is a suitable way to analyze your data prehensive approach for multichannel image registra-
will depend on your specific study, the questions tion. Biomedical image registration. Berlin: Springer;
2003. p. 214–23.
you hope to answer, on the number and type of
12. Zhang H, Yushkevich PA, Alexander DC, Gee
patients that are studied, the type of DTI data JC. Deformable registration of diffusion tensor MR
acquisition and data quality, etc. Although VBA images with explicit orientation optimization. Med
has been applied in many DTI studies, the lack of Image Anal. 2006;10:764–85.
13. Chiang MC, Leow AD, Klunder AD, Dutton RA,
a standard approach means that it remains pri-
Barysheva M, Rose SE, McMahon KL, de Zubicaray
marily a research tool, rather than a technique GI, Toga AW, Thompson PM. Fluid registration of
that can be used clinically to assess individual diffusion tensor images using information theory.
patients. IEEE Trans Med Imaging. 2008;27:442–56.
14. Verma R, Davatzikos C. Matching of diffusion tensor
images using Gabor features. In: IEEE international
symposium on biomedical imaging: nano to macro,
References vol. 391. Washington, DC: IEEE; 2004. p. 396–9.
15. Yap PT, Wu G, Zhu H, Lin W, Shen D. TIMER: tensor
1. Fitzpatrick J, Hill DLM, Maurer Jr C. Chapter 8. image morphing for elastic registration. Neuroimage.
Image registration. In: Medical image processing and 2009;47:549–63.
analysis, Handbook of medical image registration, 16. Yap PT, Wu G, Zhu H, Lin W, Shen D. F-TIMER: fast
vol. 2. Bellingham, WA: SPIE Press; 2000. tensor image morphing for elastic registration. IEEE
p. 447–513. Trans Med Imaging. 2010;29:1192–203.
2. Maintz JBA, Viergever MA. A survey of medical 17. Leemans A, Sijbers J, Vandervliet E, Parizel
image registration. Med Image Anal. 1998;2:1–36. PM. Multiscale white matter fiber tract coregistration:
3. Peeters THJM, Rodrigues PR, Vilanova A, ter Haar a new feature-based approach to align diffusion tensor
Romeny BM. Analysis of distance/similarity mea- data. Magn Reson Med. 2006;55:1414–23.
sures for diffusion tensor imaging, visualization and 18. Goodlett C, Davis B, Jean R, Gilmore J, Gerig
processing of tensor fields. New York, NY: Springer; G. Improved correspondence for DTI population
2006. studies via unbiased atlas building. In: MICCAI,
4. Alexander DC, Gee JC. Elastic matching of diffusion 2006. Berlin: Springer; 2006. p. 260–7.
tensor MRIs. Comput Vis Image Underst. 2000;77: 19. Li H, Xue Z, Guo L, Wong SC. Simultaneous consid-
233–50. eration of spatial deformation and tensor orientation
5. Muñoz-Moreno E, Cárdenes-Almeida R, Martin- in diffusion tensor image registration using local fast
Fernandez M. Review of techniques for registration of marching patterns. In: Prince J, Pham D, Myers K,
diffusion tensor imaging, tensors in image processing editors. Information processing in medical imaging.
and computer vision. New York, NY: Springer; 2009. Berlin: Springer; 2009. p. 63–75.
6. Park HJ, Kubicki M, Shenton ME, Guimond A, 20. O’Donnell LJ, Westin CF, Golby AJ. Tract-based
McCarley RW, Maier SE, Kikinis R, Jolesz FA, Westin morphometry for white matter group analysis.
CF. Spatial normalization of diffusion tensor MRI using Neuroimage. 2009;45(3):832–44.
multiple channels. Neuroimage. 2003;20:1995–2009. 21. Zhang S, Peng H, Dawe JR, Arfanakis K. Enhanced
7. Van Hecke W, Leemans A, D’Agostino E, De Backer ICBM diffusion tensor template of the human brain.
S, Vandervliet E, Parizel PM, Sijbers J. Nonrigid Neuroimage. 2011;54:974–84.
coregistration of diffusion tensor images using a vis- 22. Van Hecke W, Leemans A, Sage CA, Emsell L,
cous fluid model and mutual information. IEEE Trans Veraart J, Sijbers J, Sunaert S, Parizel PM. The effect
Med Imaging. 2007;26:1598–612. of template selection on diffusion tensor voxel-based
8. Sage CA, Van Hecke W, Peeters R, Sijbers J, analysis results. Neuroimage. 2011;55(2):566–73.
Robberecht W, Parizel P, Marchal G, Leemans A, 23. Zhang S, Arfanakis K. Role of standardized and
Sunaert S. Quantitative diffusion tensor imaging in study-specific human brain diffusion tensor templates
amyotrophic lateral sclerosis: revisited. Hum Brain in inter-subject spatial normalization. J Magn Reson
Mapp. 2009;30(11):3657–75. Imaging. 2013;37(2):372–81.
10 DTI Analysis Methods: Voxel-Based Analysis 203

24. Collins DL, Neelin P, Peters TM, Evans AC. Automatic 39. Smith SM, Jenkinson M, Johansen-Berg H, Rueckert
3D intersubject registration of MR volumetric data in D, Nichols TE, Mackay CE, Watkins KE, Ciccarelli
standardized Talairach space. Comput Assist Tomogr. O, Cader MZ, Matthews PM, et al. Tract-based spatial
1994;18(2):192–205. statistics: voxelwise analysis of multisubject diffusion
25. Evans AC, Collins DL, Mills SR, Brown ED, Kelly data. Neuroimage. 2006;31:1487–505.
RL, Peters TM. 3D statistical neuroanatomical mod- 40. Zalesky A. Moderating registration misalignment in
els from 305 MRI volumes. In: IEEE nuclear science voxelwise comparisons of DTI data: a performance
symposium and medical imaging conference. evaluation of skeleton projection. Magn Reson
Washington, DC: IEEE; 1993. p. 1813–7. Imaging. 2011;29:111–25.
26. Mazziotta JC, Toga AW, Evans A, Fox P, Lancaster 41. Keihaninejad S, Ryan NS, Malone IB, Modat M, Cash
J. A probabilistic atlas of the human brain: theory and D, Ridgway GR, Zhang H, Fox NC, Ourselin S. The
rationale for its development. The International importance of group-wise registration in tract based
Consortium for Brain Mapping (ICBM). Neuroimage. spatial statistics study of neurodegeneration: a simu-
1995;2(2):89–101. lation study in Alzheimer’s disease. PLoS One.
27. Mori S, Oishi K, Jiang H, et al. Stereotaxic white mat- 2012;7(11):e45996.
ter atlas based on diffusion tensor imaging in an 42. Edden RA, Jones DK. Spatial and orientational het-
ICBM template. Neuroimage. 2008;40:570–82. erogeneity in the statistical sensitivity of skeleton-
28. Peng H, Orlichenko A, Dawe RJ, Agam G, Zhang S, based analyses of diffusion tensor MR imaging data.
Arfanakis K. Development of a human brain diffusion J Neurosci Methods. 2011;201:213–9.
tensor template. Neuroimage. 2009;46(4):967–80. 43. Jones DK, Cercignani M. Twenty-five pitfalls in the
29. Zhang H, Yushkevich PA, Rueckert D, Gee analysis of diffusion MRI data. NMR Biomed.
JC. Unbiased white matter atlas construction using 2010;23(7):803–20.
diffusion tensor images. Med Image Comput Comput 44. Jeurissen B, Leemans A, Tournier JD, Jones DK,
Assist Interv. 2007;10(Pt 2):211–8. Sijbers J. Investigating the prevalence of complex
30. Van Hecke W, Sijbers J, D’Agostino E, Maes F, De fiber configurations in white matter tissue with diffu-
Backer S, Vandervliet E, Parizel PM, Leemans A. On sion magnetic resonance imaging. Hum Brain Mapp.
the construction of an inter-subject diffusion tensor 2013;34(11):2747–66.
magnetic resonance atlas of the healthy human brain. 45. Preti MG, Laganà MM, Baglio F, Griffanti L, Nemni
Neuroimage. 2008;43(1):69–80. R, Cecconi P, Baselli G. Comparison between
31. Jones DK, Symms MR, Cercignani M, Howard skeleton-based and atlas-based approach in the assess-
RJ. The effect of filter size on VBM analyses of ment of corpus callosum damages in Mild Cognitive
DT-MRI data. Neuroimage. 2005;26(2):546–54. Impairment and Alzheimer Disease. Conf Proc IEEE
32. Van Hecke W, Leemans A, De Backer S, Jeurissen B, Eng Med Biol Soc. 2011;2011:7808–11.
Parizel PM, Sijbers J. Comparing isotropic and aniso- 46. Schwarz CG, Reid RI, Gunter JL, Senjem ML,
tropic smoothing for voxel-based DTI analyses: a sim- Przybelski SA, Zuk SM, Whitwell JL, Vemuri P,
ulation study. Hum Brain Mapp. 2010;31(1):98–114. Josephs KA, Kantarci K, Thompson PM, Petersen
33. Rosenfeld A, Kak AC. Digital picture processing 2. RC, Jack Jr CR, Alzheimer’s Disease Neuroimaging
Orlando, FL: Academic; 1982. p. 42. Initiative. Improved DTI registration allows voxel-
34. Lee JE, Chung MK, Lazar M, DuBray MB, Kim J, based analysis that outperforms Tract-Based Spatial
Bigler ED, Lainhart JE, Alexander AL. A study of dif- Statistics. Neuroimage. 2014;94:65–78.
fusion tensor imaging by tissue-specific, smoothing- 47. Lipton ML, Kim N, Park YK, Hulkower MB, Gardin
compensated voxel-based analysis. Neuroimage. TM, et al. Robust detection of traumatic axonal injury
2009;44(3):870–83. in individual mild traumatic brain injury patients:
35. Worsley KJ, Evans AC, Marrett S, Neelin P. A three- intersubject variation, change over time and bidirec-
dimensional statistical analysis for CBF activation tional changes in anisotropy. Brain Imaging Behav.
studies in human brain. J Cereb Blood Flow Metab. 2012;6:329–42.
1992;12:900–18. 48. Kim N, Branch CA, Kim M, Lipton ML. Whole brain
36. Genovese CR, Lazar NA, Nichols T. Thresholding of approaches for identification of microstructural
statistical maps in functional neuroimaging using the abnormalities in individual patients: comparison of
false discovery rate. Neuroimage. 2002;15:870–8. techniques applied to mild traumatic brain injury.
37. Nichols TE, Holmes AP. Nonparametric permutation PLoS One. 2013;8(3):e59382.
tests for functional neuroimaging: a primer with 49. Patel SA, Hum BA, Gonzalez CF, Schwartzman RJ,
examples. Hum Brain Mapp. 2002;15:1–25. Faro SH, et al. Application of voxelwise analysis in
38. Jones DK, Chitnis XA, Job D, Khong PL, Leung LT, the detection of regions of reduced fractional anisot-
Marenco S, Smith SM, Symms MR. In Proceedings of ropy in multiple sclerosis patients. J Magn Reson
the 15th Annual Meeting ISMRM. Berlin, 2007; 74. Imaging. 2007;26:552–6.
DTI Analysis Methods: Fibre
Tracking and Connectivity 11
Matthan W.A. Caan

Learning Points From Local to Global

• Tractography does not represent individual One of the unique properties of diffusion weighted
axons but average trajectories of macroscopic MRI (DWMRI) is its ability to measure the
white matter bundles. microscopic orientation of white matter tissue on
• Different tractography methods are avail- a macroscopic scale. The axonal diameter is in the
able, each with its own strengths and order of 1–10 μm [1]. A single voxel in a DWMRI
weaknesses. scan of 2 × 2 × 2 mm3 thus comprises a diffusion
• Prior knowledge on the trajectory of pathways measurement on an ensemble in the order of 105
is indispensable and is utilized by drawing axons. The compactness and organization of
seeding and masking ROIs. white matter bundles enable the measurement of
• Data quality and tracking parameters highly anisotropic diffusion on a macroscopic scale. This
influence the tracking results. property of white matter organisation is exploited
• Tracking of major pathways can be automated, in tractography, or fibre tracking, which is a
but may require significant processing time. post-processing technique that is used to virtually
• DTI tractography may offer indirect mea- dissect white matter fibre bundles in vivo.
surements of brain structural connectivity, White matter bundles are macroscopic struc-
but quantification based on such metrics is tures facilitating communication between brain
challenging and requires cautious regions over varying distances. Association fibres
interpretation. connect gyri within one hemisphere and can be
both short and long [2]. Short association fibres
connecting adjacent gyri are termed U-fibres.
Connections between both hemispheres are
called commissural fibres. The largest commis-
sural fibre bundle is the corpus callosum, which
connects the hemispheres. Projection fibres con-
nect the cortex of the cerebrum to lower brain
M.W.A. Caan, PhD (*) parts, as well as the spinal cord. An example is
Brain Imaging Center, Academic Medical Center
Amsterdam, 22660, 1100 DD Amsterdam,
the corticospinal tract, which is mainly associ-
The Netherlands ated with motor function. Its fibres arise in the
e-mail: m.w.a.caan@amc.uva.nl precentral gyrus (the primary motor cortex) and

© Springer Science+Business Media New York 2016 205


W. Van Hecke et al. (eds.), Diffusion Tensor Imaging, DOI 10.1007/978-1-4939-3118-7_11
206 M.W.A. Caan

travel through the brainstem and spinal cord, both the orientation and shape of the diffusion
where they synapse and connect with peripheral profile are estimated from the diffusion data. In
nerves travelling to the limbs. deterministic tractography, pathways are inte-
Detailed analysis of neuronal network archi- grated in the principal eigenvector field, which
tecture can only be performed ex vivo, e.g., by can be computed from the diffusion tensor field
individual fluorescent staining of axons [3]. (Chap. 4). Tractography is dependent on user
Tractography aims to reconstruct white matter input to specify locations in the brain through
bundles traversing through multiple voxels, which tracts are to be reconstructed and that,
in vivo. Estimated fibre tracts are, in contrast to dependent on the choice, are known to be ana-
microscopic staining, in no way a direct tomically connected.
representation of single axons. They merely rep- Consider a starting point, called a seed point,
resent an integrative pathway of continuous defined in a particular voxel of interest. From this
smoothly curved diffusion orientation informa- seed point a tract is generated along both direc-
tion. Fortunately, validation in macaques has tions of the diffusion orientation in the seed
demonstrated good correspondence between voxel. The tract direction is continuously updated
tractography and histology in a number of white according to the local eigenvector orientation. As
matter bundles [4]. Nevertheless, there are a a result, a curved tract will be found, representing
number of caveats that leave a high chance of a part of a white matter bundle of interest (see
obtaining either false positive or false negative Fig. 11.1 for an example). By seeding from mul-
findings, as will be shown in this chapter. Note, in tiple points distributed over an imaginary bundle
this context “false positives” and “false nega- intersection, the entire bundle may be represented
tives” represent incorrect estimations of stream- by a series of streamlines.
line trajectories, not the classical statistical Certain criteria are required to generate ana-
concept. Tractography should hence be per- tomically plausible tracking results. Tracking is
formed with care and adequate prior anatomical terminated once a voxel is reached that is
knowledge, such as available in text books [5]. unlikely to belong to the bundle of interest. A
One of the main applications of tractography curvature threshold prevents sharp bending of a
is the segmentation of tracts in individual sub- tract and possible propagation in an adjacent
jects. In the clinic, critical pathways in the neigh- bundle. Thresholding on fractional anisotropy
borhood of a brain tumor may be reconstructed (FA) limits tracking to white matter regions
[6]. In a research setting, tractography allows the where FA is higher. Curvature thresholds of
effect of a pathological process on particular between 30 and 70° are typically used (examples
white matter bundles to be studied. In a way, are illustrated in Fig. 11.2), while FA is typically
tracking serves to define a region of interest in limited to values >0.2 (as can be seen in
which changes in (tensor-derived) measures such Fig. 11.3) [9]. Parameter values are best empiri-
as fractional anisotropy (FA) and mean diffusiv- cally chosen, balancing between false positive
ity (MD) (Chap. 5) are assessed. Some authors and negative tracking results and accounting for
refer to this type of analysis as “tractometry” [7]. the complexity of the structure. By starting at
In this chapter, a bundle refers to the ground more conservative values and slowly proceeding
truth, i.e., the anatomical substrate, while a tract to a more liberal regime, optimal parameter val-
reflects a reconstructed pathway. ues may be determined empirically. Importantly,
when multiple datasets or tracking results need
to be compared, identical settings must be
Principles of Deterministic applied to avoid a biased result.
Tractography Additional masking may be applied to restrict
tracking to specific regions. Most algorithms
Fibre tracts are estimated from measured allow a minimal and maximal tract length to be
DWMRI data using computing algorithms. set, as well as a seed point density, i.e., multiple
Tractography takes advantage of the fact that seed points per voxel.
11 DTI Analysis Methods: Fibre Tracking and Connectivity 207

Fig. 11.1 In vivo


illustration of a single
streamline created in
thecorpus callosum,
superimposed on the
principal orientation field.
The seed point is denoted
by an asterisk. Image
created with ExploreDTI
software [8]

Fig. 11.2 Effect of the


curvature threshold on the
reconstruction of highly
curved U-fibres (language
pathways). No single
optimal curvature threshold
exists, the optimal value
depends on the tract of
interest and software and
must be heuristically
determined. In this case,
40° or 50° may be chosen

Data Requirements ciently high for accurate tract reconstruction, with


minimal systematic errors. In addition, a high sig-
Reliable tractography can only be obtained in data nal-to-noise ratio (mainly determined by the field
of sufficient quality. A correct setting of specific strength, coil design, number of gradient direc-
scanning parameters of the diffusion weighted tions, and parallel or multiband imaging settings)
imaging protocol is therefore crucial. The spatial ensures that the tracking will be precise, with min-
resolution and the sensitivity of orientation encod- imal variation. We will now discuss critical param-
ing (specified by the b-value [10]) must be suffi- eters and their influence on tract reconstruction.
208 M.W.A. Caan

Fig. 11.3 Effect of the FA


threshold on the
reconstruction of the left
corticospinal tract. A low
threshold of FA = 0.1
results in more cortical
tract with the cost of false
positive tracts (indicated by
the red arrow), which may
be removed using an
addition AND-ROI. A high
FA = 0.3 threshold appears
to be too strict, with only
few tracts remaining (see
the red arrow). A threshold
of FA = 0.2 is considered
optimal and is commonly
adopted for most bundles

Resolution

Diffusion-weighted MRI is limited by a relatively


low spatial resolution, compared to other MR
modalities. At 3 T, a resolution of 2 mm in- and
through-plane can reasonably be achieved. In ini-
tial work on multislab imaging, higher isotropic
resolutions of 1.3 mm [11] and 1 mm at 7 T [12]
were reported.
The low resolution has a number of limiting
factors on tractography. First, most white matter
bundles have a thickness of only a few millime-
ters resulting in a coarse sampling and partial
Fig. 11.4 False-positive tracking result. The white
voluming. As a result, one voxel may constitute
arrows point to crossing fibre regions where incorrect
of two or more adjacent but perpendicular tracts. modeling results in corticospinal fibres traversing via the
It was shown that up to 70 % of white matter vox- corpus callosum, contralaterally to the cortex. Anatomical
els contain fibre crossings [13]. Estimating the prior knowledge is a prerequisite for correct tracking
results
principal diffusion orientation is then no longer
possible using the diffusion tensor model. This
problem was recognized more than a decade ago over 180° within the distance of a few voxels (see
[14] and is known in the field as the “crossing Fig. 11.2). A tract thus needs to propagate by steps
fibre” issue. In practice however, it refers not just of 45° or more between adjacent voxels. Such
to configurations where fibre tracts literally cross, strong bends may easily be ignored by tracking
but several other configurations where bundles algorithms in favour of continuous straight con-
fan, bend or “kiss.” This problem and solutions to nections. In addition, the chance of false positive
overcome it are introduced below and covered in tracking results increases by applying a liberal
more detail in Chap. 21. curvature threshold. This manifests as tracts “step-
Second, the low resolution limits the maximum ping over” or “jumping” between different adja-
curvature of a tract that can be reconstructed. cent bundles. A careful ROI placement is essential
U-fibres or other connections between gyri curve for accurate tract selection (see Fig. 11.4).
11 DTI Analysis Methods: Fibre Tracking and Connectivity 209

Fig. 11.5 The effect of


anisotropic resolution on
tractography: an increased
slice thickness from 2 to
6 mm with a constant
in-plane resolution of
2 × 2 mm introduces voids
in the body of the corpus
callosum (top, white
arrow) and reduces the
tract volume in the right
uncinate fasciculus
(bottom)

Tracking algorithms require data to be increasing the b-value from 1000 to 3000 s/mm2
acquired at isotropic resolution, i.e. identical reduced the minimally resolvable angle from 45°
along all three axes. Data generated with suffi- to 30° [20]. In addition to acquiring data at a
cient in-plane resolution but a higher slice thick- higher b-value, a higher order diffusion model
ness does not support accurate tract reconstruction, must be employed to resolve crossing fibres.
as illustrated in Fig. 11.5 [15]. Given the current Multi-tensor models and constrained spherical
hardware supplied by most vendors, an isotropic deconvolution (CSD) are examples of approaches
voxel size of 2.0–3.0 mm is recommended [16]. to this problem [19, 21, 22] (Chap. 21).
Increasing the b-value reduces the measured
signal value such that multiple signal averages
b-Value need to be acquired, or sequences with more gra-
dient directions must be chosen, to achieve suffi-
The strength of diffusion weighting as quantified cient data quality for performing reliable
by the b-value (Chap. 3), should be sufficiently tractography (Chap. 6).
high for a precise orientation estimation.
Although a low b-value of 600 s/mm2 enables the
reconstruction of large, uniform white matter Gradient Directions/Signal Averages
tracts (see Fig. 11.6), a value of b = 1000 s/mm2 is
advised for obtaining reliable results in the DWMRI measures diffusion in a limited number
majority of bundles [17]. of orientations, which are combined to estimate
In case of crossing fibre tracts, an even stron- an arbitrarily oriented diffusion profile. Still, the
ger diffusion weighting is required to unravel angular resolution depends on the number and
multiple diffusion orientations within one voxel configuration of the gradient directions chosen.
(see Fig. 11.7). In simulations it was shown that Figure 11.8 illustrates that the uncertainty in the
210 M.W.A. Caan

estimated orientation decreases when increasing directions are chosen for improved precision in
the number of gradient directions from 12 to 46. the estimation. In Fig. 11.9, the uncinate fascicu-
Unbiased tractography requires an optimal distri- lus can be tracked with at least 24 gradient direc-
bution of gradient directions over the unit sphere tions. In literature, a minimal set of 30 gradient
[23]. Different methods exist for doing so, e.g., directions is advised [23], while 60–80 directions
by tessellations of an icosahedron [24], or a dis- are recommended for minimal variance in trac-
tribution of charges. Some vendors provide pre- tography [16].
defined sets of gradient directions of different In addition, the number of signal averages
numbers (e.g., 12, 32, or 64 directions). Note that (NSA) can be increased. Note that both dou-
these sets may not be optimized for tractography, bling the number of gradient directions and
in which case, if possible, a user-defined gradient choosing NSA = 2 increase scanning time by a
set should be entered. factor of 2. Generally speaking, a higher num-
Theoretically, six directions are required to ber of gradient directions slightly increases
estimate the diffusion tensor. Practically, more the angular resolution and is preferable over
multiple signal averages. Clearly, if time per-
mits choosing NSA = 2 will improve the track-
ing precision even more. However, increasing
the scanning time also increases the likelihood
of motion artifacts, which may detract from
the image quality gain achieved by increasing
the NSA.

Field Strength

Imaging at higher field strengths increases the


measured signal, which in turn reduces the
uncertainty in the estimated diffusion orienta-
Fig. 11.6 Tracking in a dataset with a lower b-value of tion. A field strength of 3.0 T is currently com-
600 s/mm2. The uncinate fasciculus can be accurately mon practice in tractography-based clinical
delineated in this dataset research studies. Tractography can successfully

Fig. 11.7 Tractography


through fibre crossings in a
patient with abnormal
pathway development in
the pons [18]. In addition
to non-diffusion-weighted
images, 92 gradient
directions with a b-value of
1600 s/mm2 were acquired.
Deterministic tractography
with a constrained
spherical deconvolution
(CSD) clearly shows
crossing pathways [19].
Tracking performed in
ExploreDTI software
11 DTI Analysis Methods: Fibre Tracking and Connectivity 211

Fig. 11.8 Cones of


uncertainty of the
estimated diffusion
orientation superimposed
on the FA of a sagittal slice
through the cingulum
(cing) and corpus callosum
(CC) for datasets with 12
and 46 gradient directions
(ng). Notice increased
uncertainty for ng = 12 and
in voxels with low FA

performed at 1.5 T, e.g., by acquiring two signal Tract Selection


averages (NSA = 2), acquiring data along a
greater number of diffusion gradient directions In tractography, prior anatomical knowledge on
and by using multichannel phased-array head- the trajectory of the bundle of interest is indis-
coils (see Chap. 6). In a prospective evaluation pensable. Based on this knowledge, one of more
of the depiction of fibre tracts at 1.5 and 3.0 T, regions of interest (ROIs) through which a bundle
higher visual scores, larger numbers of fibres, traverses need to be defined. By means of logic
and a higher asymmetry index in the CST were combinations, a set of fibre tracts representing
obtained at 3.0 T [25]. the bundle can be obtained. See Section 4, “DTI
High-field scanners (7.0 T and above) enable Tractography Atlas” for further details on how to
smaller anatomical structures to be identified track specific white matter bundles.
[12]. Diffusion-weighted imaging at 7.0 T com-
pared to 1.5 and 3.0 T showed an increased SNR
that was larger than what could be expected Seeding
from field strength alone: improved receive coil
hardware largely contributes to higher image Fibre tracking is initiated from so-called seed
quality [26]. points (see Fig. 11.1). These points may be man-
As a downside, imaging artifacts will have a ually annotated by the user in a single voxel or
higher impact at higher field strength. ROI. A drawback of this approach is that fibre
Inhomogeneities of the static field (B0) spatially tracking is a non-commutative procedure [28],
distort the images. Increasing the acceleration meaning that tracking from a tract end point will
factor partly compensates this effect. Additionally, not automatically result in a pathway back to the
variations in the radiofrequency field (B1) initial seed region, as is illustrated in Fig. 11.10.
increase signal heterogeneity at 7.0 T [27]. An alternative approach to single ROI seeding is
212 M.W.A. Caan

at least result in a more robust tracking of sepa-


rate branches (see Fig. 11.12). If whole-brain
tractography is not supported or practically cum-
bersome such as in probabilistic tractography
(see “Tract Selection”), multiple seed ROIs in all
possible branches need to be defined as will be
explained in the next section.

Logic Combination: OR/AND/NOT

Logic combination applied to multiple ROIs is


commonly needed to delineate a bundle. The
OR-operator on two ROIs returns fibres travers-
ing through one or two ROIs. Applying the AND-
operator is stricter and only returns fibres passing
through both ROIs (see Fig. 11.13 for an exam-
ple). The NOT-operator, removing fibres entering
a specific ROI, must be used sparingly to obtain a
reproducible tracking result.

Good Practice for ROI-Drawing

Tractography requires spatial awareness and


interpretation of a 3D representation on a 2D
screen (although some packages support stereo-
viewing). Hence it requires a number of trial-
and-error experiments before an accurate result is
achieved. Direct feedback from the software aids
in speeding up the training phase. A number of
programs support interactive exploration, provid-
ing instant tracking results after the user updates
Fig. 11.9 Tractography of theuncinate fasciculus (UF) selection regions. There are currently methods
for increasing number of gradient directions. With the available, which allow interactive positioning of
theoretical minimum of six directions, the UF cannot be a single seed point or one or more selection boxes
reconstructed
(Fig. 11.14).
More accurate tracking may be achieved by
whole-brain tractography, in which an algorithm delineating arbitrarily shaped ROIs on anatomical
initiates seeding in all voxels that satisfy certain scans. The fractional anisotropy (FA) map color-
criteria, e.g., all white-matter voxels with FA > 0.2 coded for orientation provides a good reference for
act as seed points, as displayed in Fig. 11.11. annotation. Essentially, this map encodes all infor-
From this whole-brain tractography, the user can mation employed during tracking. Most accurate
select a set of tracts using an ROI. The advantage tracking is achieved by annotating on a plane per-
of this approach is its symmetry, whereby tracts pendicular to the (local) tract orientation. The
starting in the ROI and tracts from other brain color information aids the user to identify such a
regions passing through the ROI are found. This plane. Including low-intensity voxels representing
does not result in commutative tracking, but will low FA will not affect tractography, since these
11 DTI Analysis Methods: Fibre Tracking and Connectivity 213

Fig. 11.10 Illustration of


the non-commutative
property of fibre tracking:
seeding from the end voxel
of the left tract results in a
different pathway

Fig. 11.11 Whole-brain


tractography (sagittal
view), obtained by seeding
from all voxels with
FA > 0.2, colored red on the
left image

voxels are automatically disregarded based on an a spatial distortion correction of the diffusion data
FA threshold by the algorithm (see Box 11.1) may be required by means of acquiring a B0-map
Be aware that annotating ROIs on the FA map or performing nonrigid registration (see Chap. 7).
may create a bias, since the ROIs are drawn on The tractography result is dependent on the
the same maps that will be used for statistical selection criteria as imposed by the user.
comparison. Procedures for reproducible reconstruction of
A high-resolution structural scan provides major white matter tracts with deterministic trac-
additional anatomical reference and can be used tography have therefore been proposed [31]. Even
in addition to the color-coded FA map. One must when following synchronized tracking proce-
keep in mind that the spatial resolution of the dif- dures, the inter-observer agreement in practice has
fusion data is commonly a factor of two lower. been reported as not exceeding 90 % [32]. This
Also, it is necessary to verify that the scans are means that some variability exists in the tract vol-
spatially aligned and a correction for possible ume obtained from ROIs drawn by different users.
head motion will typically be needed. In addition, This variability is illustrated in Fig. 11.15, sum-
214 M.W.A. Caan

Fig. 11.12 Different


seeding strategies: (a)
single-seed ROI versus (b)
whole-brain tractography,
in which case all brain
white matter voxels serve
as seeds. A more dense
tracking is obtained using
whole-brain tractography.
One sagittal ROI was
drawn in the body of the
corpus callosum (not
shown)

Box 11.1: On Estimating Tract Volumeand FA a tract, considering the large variation in the
(Consideration Box for Researchers) number of obtained voxels by the students.
In a research study, there may be hypotheses Indeed, in literature larger variations in tract
of microstructural damage, resulting in volume than tract FA were reported [59]. This
reduced FA in a tract or white matter atrophy work also showed that traumatic volume loss
resulting in reduced tract volume on macro- confounds FA estimates, since FA was reduced
scopic level. Figure 11.15 shows results of a in smaller volume tracts, likely caused by par-
tractography training session of students and tial volume effects. Including tract volume as
suggests that tract volume is measured with covariate in the statistical model is one way of
less precision compared to the mean FA within overcoming this issue [60].
11 DTI Analysis Methods: Fibre Tracking and Connectivity 215

The following guidelines may aid in achiev-


ing a reproducible tracking:

• Draw ROIs as large as possible. This may


seem counter-intuitive, but ensures that any
possible fibre belonging to a bundle will be
included.
• Annotate 2D ROIs in the plane perpendicular
to the bundle orientation.
• Apply as few ROIs as possible. In most situa-
tions, two or three ROIs suffice, i.e., one OR
ROI at one end of the tract, an AND ROI at
another end and another AND ROI along the
pathway to exclude any deviating tracts.
• Use NOT-ROIs sparingly. Only fibres appear-
ing as clear outliers may be removed.
• In case whole-brain tractography is unavailable,
generate two overlapping seed and AND ROIs
at both sides of a tract. By doing so a symmetric
tracking result is obtained (see Fig. 11.17).
• Section 4 of this book discusses normal anat-
omy described by a DTI atlas.

Probabilistic Tractography

The principal orientation of diffusion can only be


estimated with a certain limited precision [33].
The uncertainty increases both with higher noise
levels or fewer gradient directions and lower dif-
fusion anisotropy (see Fig. 11.8).
Tractography is highly sensitive to uncertainty
in the orientation estimation. By each tracking
step into an adjacent voxel, the pathway will fur-
ther deviate from the truth. In other words, the
uncertainties propagate or accumulate, such that
Fig. 11.13 Accurate delineation of a longitudinal fascic- tracts may terminate prematurely and the target
ulus. A single-seed ROI (a) results in many false positive
fibres (b). Adding a second AND ROI (c) removes these pathway may not be found. One must be aware
fibres from the selection, resulting in the desired pathway that this uncertainty is invisible in conventional
deterministic tractography visualizations, which
marizing a training session in which students were tend to be perceived as precise segmentation
assigned to reconstruct the forceps major and cin- while in reality this may not be the case, as is
gulum tracts. Large variations in tract volume are illustrated in Fig. 11.4.
observed, resulting from differences in ROI size Fortunately, knowledge on the amount of
and positioning. One can conclude that training on uncertainty in the orientation estimation can be
multiple datasets is required to converge to repro- used to obtain potentially precisely delineated
ducible findings. Also note that the inter-observer pathways. Tractography as it has been introduced
variability in average FA is relatively low. thus far is characterized as deterministic. Repeated
216 M.W.A. Caan

Fig. 11.14 Interactive


tractography of the
cingulum bundle using two
boxes. Transparent colored
fibres reflect fibres
originating from the
corresponding box. Solid
green fibres represent the
cingulum tract, connecting
both boxes. Image created
with DTI interactive (DTIi)
software [29]

Fig. 11.15 Results of a training session of a group of 15 tracking of the Forceps Major, containing approximately
students. (a) Mean FA and number of voxels of two tracts 4000 voxels. Tracking performed in DTI-Studio software
created by the students. (b) Illustrative image of a correct [30]

tracking instances from a single seed point will (see Fig. 11.8). The mean of this distribution
return identical tracking results, based on an aver- equals the orientation used in deterministic trac-
age orientation estimate per voxel (Fig. 11.16). tography. The width of the distribution is
Probabilistic tractography instead estimates proportional to the uncertainty. Once the distri-
the orientation distribution in each voxel [21, 34] bution is known, multiple tracts in the order of
11 DTI Analysis Methods: Fibre Tracking and Connectivity 217

Fig. 11.16 Deterministic


and probabilistic
tractography. (a)
Deterministic tractography
is based on the principal
eigenvector, while
probabilistic tractography
is based on a distribution of
estimated possible
orientations. (b) From a
seed voxel, a single tract is
computed in deterministic
tracking. Probabilistic
tracking proceeds along
one randomly selected
orientation per voxel. (c)
After probabilistic
tracking, a tract probability
map is computed for the
relative number of tracts
passing through a voxel

10,000 are seeded from each seed point. A tract is Deterministic tractography will be unable to find
computed on an orientation estimate drawn from tracts due to the large distance to be tracked and
the distribution in all voxels. Each tract thus fol- the uncertainty in the principal orientation in gray
lows a different pathway. From the average num- matter. By using probabilistic tractography, spe-
ber of visits of the tracts to each voxel, a tract cific distinct subregions within the thalamic gray
probability map can be derived. When the orien- matter have been identified, which correspond to
tation uncertainty is low, all tracts will traverse histological studies [35].
along a similar pathway. Tracts passing through a Probabilistic tractography is computation-
single area with high uncertainty will spread and ally much more demanding than deterministic
show a diffuse pattern afterwards. tractography. Estimating the initial orientation
Mapping connections from gray matter regions distribution, e.g., by Bayesian estimation of
such as the thalamus to the cortex is challenging. diffusion parameters implemented in FSL
218 M.W.A. Caan

Fig. 11.17 (a) In


probabilistic tractography,
more accurate tract
selection is possible by
adding a waypoint mask,
returning only fibres
passing through this mask.
(b) This type of selection
is asymmetrical, swapping
the seed and waypoint
mask will generate a
different outcome, with the
highest probability in the
seed mask. (c) Using both
ROIs as seeds and
waypoints results in a
more representative
probability map

BedpostX [21] takes approximately 15 h to whole-brain probabilistic tracking is less com-


complete. There are however, other (non- monly performed than deterministic tracking.
Bayesian) based methods available that can To obtain symmetric and precise tracking
perform whole-brain probabilistic tractography results, using combined seed and waypoint
in a shorter time-frame (see [36] for a review). masks is advised as illustrated in Fig. 11.17.
Due to the many seeds per voxel, computa- The obtained probability map may be visual-
tional demands and less intuitive visualization, ized in 3D using a volume rendering such as in
11 DTI Analysis Methods: Fibre Tracking and Connectivity 219

Fig. 11.18 Automated


atlas-based tractography.
Here, ROIs are manually
annotated in standard-
space (a, cross sections
shown in dashed red).
Then, the atlas FA volume
is registered to the
subject’s FA volume (b).
The ROIs are transformed
accordingly. (c) In the
subjects’ dataset,
frontostriatal-thalamic
fibres are reconstructed
using probabilistic
tractography, using all
three ROIs both as seed
and waypoint masks.
Registration and
tractography is performed
using FSL software. A 3D
volume rendering is
created in 3D-Slicer
software

Fig. 11.18c or a volume isosurface such as in Higher Order Model Tractography


Fig. 11.19. Dependent on the application,
deterministic tractography remains the first The diffusion tensor model assumes a single-
method for interactive exploration and selec- fibre bundle traversing through a voxel. It has
tion, while probabilistic tractography can add however been estimated that up to 70 % of all
robustness if required. white matter voxels contain contributions of at
220 M.W.A. Caan

Fig. 11.19 Automated


tractography by means of
TRACULA [37]. A
T1-weighted scan is
segmented by freesurfer
software, the resulting
labels serve as ROIs in the
tractography. Diffusion
weighted MRI data are
processed by FSL
bedpostX software to
compute multiple diffusion
orientations per voxel (to
model fibre crossings).
TRACULA combines these
data to reconstruct 18 fibre
pathway probability maps
11 DTI Analysis Methods: Fibre Tracking and Connectivity 221

least two bundles [13]. Fortunately, despite White Matter Atlas


this model error, it is possible to successfully
reconstruct the core structure of major bundles, Stereotaxic human brain atlases are often
which is sufficient for preclinical research employed in neuroimaging research as an ana-
studies investigating groups of subjects. Note tomical reference. Based on an existing anatomi-
however, this important limitation means that cal template averaged over 152 subjects
DTI tractography is not the most appropriate (ICBM-152), a hand-segmented white matter
method for presurgical planning (see Chaps. 15 parcellation map was created, based on fibre ori-
and 21). entation information [39]. Instead of performing
As stated in Chap. 21, a large number of tractography, such a map may be used to segment
higher order models for complex fibre configu- a novel dataset after spatial alignment by means
rations have been proposed. Estimating higher of registration. The accuracy of the segmentation
order models requires higher quality data with relies on registration errors, mainly caused by
a greater number of diffusion gradient direc- inter-subject anatomical variation. As an exten-
tions and higher b-values compared to classical sion, a white matter parcellation atlas was created
DTI (see Chap. 6). This type of acquisition is by populating tract probability maps of multiple
commonly referred to as high angular resolu- subjects [40]. This atlas allows for automated
tion diffusion imaging or HARDI. A higher tract-specific quantification of DTI measures.
b-value of b > 1500 s/mm2 and >60 gradient
directions are preferred, compared to the typi-
cal b = 1000 s/mm2 and 30 directions for Atlas-Based Tractography
DTI. This increases the scan time beyond 8 min
per average. Previously this has proved an A disadvantage of using white matter atlases is
obstacle for clinical research; however, it is that not all inter-subject morphologic variations
now possible to obtain good quality HARDI can be modeled. Alternatively, seeding and mask-
data in under 15 min. ing ROIs may be selected from an atlas for track-
Figure 11.7 presents an example of tractogra- ing in specific datasets, which is named
phy using CSD in the pons of a patient with an atlas-based tracking [41, 42]. While minor spa-
embryonic defect in axonal guidance [38]. The tial offsets in the ROIs may be expected, the
patient showed symptoms of reduced coordina- tracking results obtained potentially more accu-
tion of muscle movements (ataxia), eye move- rately describe the anatomy in the datasets com-
ments (nystagmus), sensory deafness and pared to atlas-based segmentation alone, as
episodes of hypothermia. Common interhemi- described in the previous section.
spherical connections were diminished in this In the atlas, a set of reference regions of inter-
patient (data not shown); instead an additional est (rROIs) is defined that allow the bundle of
transversal bundle was observed, connected to an interest to be tracked. Next, spatial correspon-
abnormal projection out of the brain stem. dence needs to found between the atlas and the
dataset in which tracking needs to be performed.
In Fig. 11.18, this is achieved by linear (affine)
Automated Tractography registration. The rROIs are transformed accord-
ingly and tracking in the target dataset is per-
Fibre reconstruction relies on manual identifica- formed using the rROIs. Following this approach,
tion of anatomical landmarks of a tract of inter- sufficient agreement with a manual approaches
est. This subjective judgment is a potential source was obtained (κ > 0.8) [42], showing that atlas-
of experimental variability. In addition, manual based automated tracking is relatively robust to
reconstruction in a large number of datasets is registration errors between the atlas and the data-
time consuming. Several methods have therefore set of interest. See Chaps. 10 and 12 for further
been proposed for automated tractography. details about DTI atlases and spatial alignment.
222 M.W.A. Caan

Additional prior information for tractography traverse between two brain regions, the stronger
may be obtained from a high-resolution struc- the connection is. Additionally, changes in the
tural T1-weighted scan. An automated segmenta- local microstructural composition of the myelin
tion and parcellation of subcortical and cortical sheath and cell membranes may affect connectiv-
structures [43] provides accurately defined ROIs ity. Rather than quantifying these neurobiologi-
for seeding and masking. cal features, connectivity in the context of
A method was proposed that allows the auto- tractography refers to the probability of recon-
mated delineation of 18 “tracts constrained by structing a pathway based on diffusion [46]. The
underlying anatomy” (TRACULA). The method question to be answered is if tractography-derived
takes T1- and diffusion-weighted images as input. measures can be used as markers of connectivity.
Spatial distortions that appear in DWIs need to be If so, then variations in these measures must be
corrected by including an additional B0-scan or by related to changes in the known (or hypothesized)
registration. The T1-scan is segmented using connectivity and be minimally driven by mea-
Freesurfer software [43, 44] and diffusion orienta- surement and reconstruction errors. We will
tion distributions are estimated by means of briefly touch upon aspects the reader may
BedpostX (see “Tract Selection”). Based on these encounter when considering a tractography-
data tractography is performed resulting in subject- based connectivity study.
specific tract probability maps. These are displayed
in Fig. 11.19 at a threshold of 20 %. In case of gross
anatomical variation, automated tractography What Were We Measuring Again?
might fail, in which case it is advisable to manually
define more ROIs than are definedby default. It is important to stress that the signal measured
with diffusion MRI is really distant from what
connectivity aims to capture. Random Brownian
Connectivity motion of water molecules causes an orientation
dependent signal attenuation. As discussed
The human brain is a highly interconnected organ before, diffusion MRI is measuring at limited
in which different brain regions are mutually resolution, orders of magnitude higher than the
dependent on one another. This leverages a high axonal scale. For the purpose of tractography, the
potential for studies into large-scale connectivity principal orientations of diffusion are obtained
that may provide novel insight into complex brain from the data using any number of modeling
disorders. DW-MRI has been enthusiastically schemes, among which CSD [19] and compart-
adopted by neuroscientists as a tool for assessing ment models [47]. One must be aware that these
structural connectivity in the human brain. One are the available measures based on which
goal is to associate this structural connectivity research questions are to be defined and answered.
with measures of functional connectivity, e.g.,
derived from correlation measures in functional
MRI data. As an example, in a study into Major What Not to Do
Depressive Disorder, a negative structure-
function relation was identified that was posi- Streamline deterministic tractography as intro-
tively associated with depression severity [45]. duced in this chapter answers the question if a
The purpose of studying white matter connec- connection between regions can be reconstructed
tivity is to gain insight into the function of the from the given data with a clear “yes” or “no.”
brain [28]. The challenging task is to quantify Any other inferences on quality or confidence of
connectivity using certain measures that may be the connection cannot be made. Anatomically
compared over time or between individuals. One implausible connections such as given in
structural aspect of connectivity is defined by the Fig. 11.4 are displayed with equally solid stream-
number of axonal projections: the more axons lines as other connections that are known to be
11 DTI Analysis Methods: Fibre Tracking and Connectivity 223

present. Probabilistic tractography by no means Other Connectivity Measures?


overcomes this issue of inaccuracy: nonexisting
connections are shown with equally large repro- Probabilistic tractography outputs the reproduc-
ducibility as for deterministic tractography [28]. ibility of tracking in a tract visitation map. These
Consequently, one cannot bluntly track fibres numbers must not be interpreted as a probability
between any pair of brain regions and obtain of the local existence of a connection, but instead
massive connectivity matrices without at least as a measure of variation in the obtained result.
introducing anatomical prior knowledge on con- Figure 11.17 shows that the tracking variation is
nections that are known are there. This is not to minimal close to the seeding area and thus opera-
state that, if carefully designed, such matrices tor dependent. Also other topology-induced vari-
cannot be reconstructed, as is shown in [48]. ation is present. This makes it clear that these
probabilistic outcomes cannot be directly inter-
preted as connectivity estimates.
Fibre Count Global tractography (not to be confused with
“whole-brain tractography”) is a more recent
It is tempting to interpret streamline or fibre count development in the field based on HARDI data,
as a measure for the number of axons. However, which models the problem of estimating the trac-
as has been made clear, tractography is measured togram in a different way to the other techniques
on an entirely different measurement scale. Also, discussed so far. In classical approaches, algo-
a number of factors affect the fibre count: rithms try and find a close match between the
local diffusion signal and underlying microstruc-
• Increasing length and curvature will decrease ture in each voxel, and propagate streamlines
the fibre count, since tracts ‘bend off’ and are based on the principal direction(s) of diffusion.
early terminated. This means they are sensitive to cumulative errors
• Brain diseases may locally affect white matter along the fibre trajectory. In global approaches,
bundles such that tractography may primarily algorithms reconstruct the whole tractogram
follow other pathways. It is likely that in such simultaneously by finding the most optimal paths
a situation an increased number of fibres are through the diffusion field that best fit the underly-
found in these other pathways, which clearly ing data. In contrast to classical, local approaches,
does not reflect the underlying configuration. in global tractography, neighboring pathways
• Brain atrophy is highly correlated with DTI influence each other as part of the optimization
measures [49] and tract volume [50] and needs process. The advantage of such global approaches
to be accounted for in statistical analyses. is that they are less influenced by local perturba-
• The fibre count is highly dependent on the tions in the diffusion signal (e.g., due to artefacts,
uncertainty in the estimated diffusion orienta- noise) and the accumulation of errors. This may
tion, which in turn is dependent on FA and make them more appropriate for investigating
data quality (see Fig. 11.8). FA is directly global connectivity than classical approaches. The
related to the diffusion profile and must be disadvantage however, is that they are computa-
assessed if appropriate. tionally very demanding and are therefore pres-
• Track-weighted imaging or tract density imag- ently used only in a research context. For further
ing and allied techniques uses a sophisticated information, the reader is directed to [54].
form of interpolation to visualize diffusion
data at a higher resolution than the acquired
resolution by locally integrating the number of Foresight
visiting streamlines [51, 52]. While visually
attractive (see Fig. 11.20) and reproducible How to proceed in assessing connectivity based
[53], these maps are primarily qualitative and on diffusion MRI? There is increasing evidence
still subject to similar limitations as other that tractography-derived measures correlate
probabilistic tractography based methods. with independent modalities, such as fMRI. Future
224 M.W.A. Caan

Fig. 11.20 (a) Axial FA map colorcoded by orientation same data as (a), with 20 million streamlines on a 0.5 mm
(Fac) at original resolution of 2.5 mm (isotropic). (b) isotropic grid. Note the enhanced detail in deep white
Color-coded track-density image (TDI) generated from matter structures, but the apparent loss of detail in the cor-
the same data as (a) using probabilistic tractography (con- tex due to higher uncertainty in long-range connections in
strained spherical deconvolution), with 20 million stream- these regions. Images generated using MRTrix software.
lines on a 2.5 mm isotropic grid (i.e., original resolution). Courtesy of Thijs Dhollander
(c) Color-coded super-resolution TDI generated from the

research should identify the underlying processes been used for creating figures is mentioned in
that co-occur in these data. Novel imaging modal- the captions wherever appropriate. The reader is
ities such as mapping myelin density [55] may referred to Chap. 13 for further discussion on
shed new light on this highly interesting debate. practical issues related to the analysis of DTI
Meanwhile, tractography-derived measures may data using presently available software. Some
be adopted, as long as the strong underlying vendors provide software for online tracking on
assumptions are recognized and not violated in the scanner console or a connected computer.
analysis and interpretation [56]. The advantage is that data do not have be trans-
ferred and processed off-line (requiring time
and expertise) but can be analyzed on the fly,
Discussion and results can easily be integrated in the
acquired dataset to be archived in the hospital
In this chapter, a number of different tractogra- PACS. An example of online tracking is given
phy methods have been presented, each with its in Fig. 11.21.
own strengths and weaknesses. Data quality and In conclusion, tractography is a powerful
tracking parameters highly influence the tracking method to obtain estimates of global structural
results. Unfortunately, no ground truth is avail- connections between brain regions from local
able to the user for validation. Prior knowledge measurements of diffusion. If performed and
on the global trajectory of pathways is indispens- interpreted correctly, it may provide useful com-
able in drawing seeding and masking ROIs. plementary information in preclinical research
Different software packages exist for per- and has potential future utility in routine clinical
forming tractography. The software that has practice (Fig. 11.22).
11 DTI Analysis Methods: Fibre Tracking and Connectivity 225

Fig. 11.21 Online


tracking result with scanner
software (Fibretrak, Philips
Medical Systems) of the
corticospinal tract.
Multiple ROIs were
defined by the user to
obtain the left corticospinal
tract. An FA map color
coded for orientation is
blended with a T1-weighted
structural scan to aid in
delineating the tract. A
short diffusion weighted
protocol was chosen:
resolution 2.5 mm
isotropic, 30 gradient
directions, b = 1000 s/mm2,
total scanning time 3 min
and 30 s. Head motion was
corrected for prior to
tractography

Fig. 11.22 What is my


tractography goal?
226 M.W.A. Caan

Must Reads 11. Engström M, Skare S. Diffusion-weighted 3D mul-


tislab echo planar imaging for high signal-to-noise
ratio efficiency and isotropic image resolution. Magn
1. One of the first articles that introduced trac- Reson Med. 2013;1514:1507–14.
tography [57]. 12. Heidemann RM, Anwander A, Feiweier T, Knösche TR,
2. Principles and limitations of tractography [15]. Turner R. k-space and q-space: combining ultra-high
spatial and angular resolution in diffusion imaging using
3. Discussion on connectivity and tractography
ZOOPPA at 7 T. Neuroimage. 2012;60(2):967–78.
[28, 36]. 13. Jeurissen B, Leemans A, Tournier J-D, Jones DK,
4. Overview paper discussing challenges to be Sijbers J. Investigating the prevalence of complex
addressed in the future [7]. fiber configurations in white matter tissue with diffu-
sion magnetic resonance imaging. Hum Brain Mapp.
5. A paper providing a distinctive mapping
2013;34(11):2747–66.
between thalamic and cortical regions using 14. Tuch DS, Reese TG, Wiegell MR, Wedeen
probabilistic tractography [35]. VJ. Diffusion MRI of complex neural architecture.
Neuron. 2003;40(5):885–95.
15. Chung H-W, Chou M-C, Chen C-Y. Principles and
limitations of computational algorithms in clinical
diffusion tensor MR tractography. AJNR Am
References J Neuroradiol. 2011;32(1):3–13.
16. Jones DK, Knösche TR, Turner R. White matter
1. Waxman SG, Kocsis JD, Stys PK. The axon: struc- integrity, fiber count, and other fallacies: the do’s and
ture, function and pathophysiology. Oxford: Oxford don’ts of diffusion MRI. Neuroimage.
University Press; 1995. 2013;73:239–54.
2. Jellison BJ, Field AS, Medow J, Lazar M, Salamat 17. Jones DK, Basser PJ. ‘Squashing peanuts and smash-
MS, Alexander AL. Diffusion tensor imaging of cere- ing pumpkins’: how noise distorts diffusion-weighted
bral white matter: a pictorial review of physics, fiber MR data. Magn Reson Med. 2004;52(5):979–93.
tract anatomy, and tumor imaging patterns. AJNR Am 18. Barth PG, Majoie CB, Caan MWA, Weterman MAJ,
J Neuroradiol. 2004;25(3):356–69. Kyllerman M, Smit LME, Kaplan RA, Haas RH, Baas
3. Livet J, Weissman TA, Kang H, Draft RW, Lu J, F, Cobben J-M, Poll-The BT. Pontine tegmental cap
Bennis RA, Sanes JR, Lichtman JW. Transgenic strat- dysplasia: a novel brain malformation with a defect in
egies for combinatorial expression of fluorescent pro- axonal guidance. Brain. 2007;130(Pt 9):2258–66.
teins in the nervous system. Nature. 2007;450(7166): 19. Jeurissen B, Leemans A, Jones DK, Tournier JD,
56–62. Sijbers J. Probabilistic fiber tracking using the resid-
4. Dauguet J, Peled S, Berezovskii V, Delzescaux T, ual bootstrap with constrained spherical deconvolu-
Warfield SK, Born R, Westin C-F. Comparison of tion. Hum Brain Mapp. 2011;32(3):461–79.
fiber tracts derived from in-vivo DTI tractography 20. Ghosh A, Descoteaux M, Deriche R. Riemannian
with 3D histological neural tract tracer reconstruction framework for estimating symmetric positive definite
on a macaque brain. Neuroimage. 2007;37(2):530–8. 4th order diffusion tensors. Med Image Comput
5. Nieuwenhuys R, Voogd J, Van Huijzen C, van Huijzen Comput Assist Interv. 2008;11(Pt 1):858–65.
C, Voogd J. The human central nervous system. 21. Behrens TE, Berg HJ, Jbabdi S, Rushworth MF,
New York: Springer; 2008. Woolrich MW. Probabilistic diffusion tractography
6. Stadlbauer A, Nimsky C, Buslei R, Salomonowitz E, with multiple fibre orientations: what can we gain?
Hammen T, Buchfelder M, Moser E, Ernst-Stecken Neuroimage. 2007;34(1):144–55.
A, Ganslandt O. Diffusion tensor imaging and opti- 22. Caan MWA, Khedoe HG, Poot DHJ, den Dekker AJ,
mized fiber tracking in glioma patients: histopatho- Olabarriaga SD, Grimbergen KA, van Vliet LJ, Vos
logic evaluation of tumor-invaded white matter FM. Estimation of diffusion properties in crossing
structures. Neuroimage. 2007;34(3):949–56. fiber bundles. IEEE Trans Med Imaging.
7. Jbabdi S, Johansen-Berg H. Tractography: where do 2010;29(8):1504–15.
we go from here? Brain Connect. 2011;1(3):169–83. 23. Jones DK. The effect of gradient sampling schemes
8. Leemans A, Jeurissen B. ExploreDTI: a graphical on measures derived from diffusion tensor MRI: a
toolbox for processing, analyzing, and visualizing dif- Monte Carlo study. Magn Reson Med.
fusion MR data. 17th Annual Meeting …, vol. 245, 2004;51(4):807–15.
no. 2, p. 3300, 2009. 24. Akkerman EM. The direct tensor solution and higher-
9. Tournier JD, Mori S, Leemans A. Diffusion tensor order acquisition schemes for generalized diffusion
imaging and beyond. Magn Reson Med. 2011; tensor imaging. J Magn Reson. 2010;206(1):9–19.
65(6):1532–56. 25. Okada T, Miki Y, Fushimi Y, Hanakawa T, Kanagaki
10. Le Bihan D. Molecular diffusion nuclear magnetic M, Yamamoto A, Urayama S, Fukuyama H, Hiraoka
resonance imaging. Magn Reson Q. 1991;7(1):1–30. M, Togashi K. Diffusion-tensor fiber tractography:
11 DTI Analysis Methods: Fibre Tracking and Connectivity 227

intraindividual comparison of 3.0-T and 1.5-T MR novel finding in pontine tegmental cap dysplasia. Eur
imaging. Radiology. 2006;238(2):668–78. J Paediatr Neurol. 2014;18(3):434–8.
26. Polders DL, Leemans A, Hendrikse J, Donahue MJ, 39. Mori S, Oishi K, Jiang H, Jiang L, Li X, Akhter K,
Luijten PR, Hoogduin JM. Signal to noise ratio and Hua K, Faria AV, Mahmood A, Woods R, Toga AW,
uncertainty in diffusion tensor imaging at 1.5, 3.0, and Pike GB, Neto PR, Evans A, Zhang J, Huang H,
7.0 Tesla. J Magn Reson Imaging. Miller MI, van Zijl P, Mazziotta J. Stereotaxic white
2011;33(6):1456–63. matter atlas based on diffusion tensor imaging in an
27. Choi S, Cunningham DT, Aguila F, Corrigan JD, ICBM template. Neuroimage. 2008;40(2):570–82.
Bogner J, Mysiw WJ, Knopp MV, Schmalbrock 40. Hua K, Zhang J, Wakana S, Jiang H, Li X, Reich DS,
P. DTI at 7 and 3 T: systematic comparison of SNR Calabresi PA, Pekar JJ, van Zijl PCM, Mori S. Tract
and its influence on quantitative metrics. Magn Reson probability maps in stereotaxic spaces: analyses of
Imaging. 2011;29(6):739–51. white matter anatomy and tract-specific quantifica-
28. Jones DK. Challenges and limitations of quantifying tion. Neuroimage. 2008;39(1):336–47.
brain connectivity in vivo with diffusion MRI. Imaging 41. Nucifora PGP, Wu X, Melhem ER, Gur RE, Gur RC,
Med. 2010;2(3):341–55. Verma R. Automated diffusion tensor tractography:
29. Blaas J, Botha CP, Peters B, Vos FM, Post FH. Fast implementation and comparison to user-driven trac-
and reproducible fiber bundle selection in DTI visual- tography. Acad Radiol. 2012;19(5):622–9.
ization. IEEE Vis. 2005, pp. 59–64. 42. Zhang W, Olivi A, Hertig S, Van Zijl P, Mori
30. Jiang H, van Zijl PCM, Kim J, Pearlson GD, Mori S. Automated fiber tracking of human brain white
S. DtiStudio: resource program for diffusion tensor matter using diffusion tensor imaging. Neuroimage.
computation and fiber bundle tracking. Comput 2008;42(2):771–7.
Methods Programs Biomed. 2006;81(2):106–16. 43. Fischl B. Automatically parcellating the human cere-
31. Wakana S, Caprihan A, Panzenboeck MM, Fallon JH, bral cortex. Cereb Cortex. 2004;14(1):11–22.
Perry M, Gollub RL, Hua K, Zhang J, Jiang H, Dubey 44. Fischl B, Salat DH, Busa E, Albert M, Dieterich M,
P, Blitz A, van Zijl P, Mori S. Reproducibility of quan- Haselgrove C, van der Kouwe A, Killiany R, Kennedy
titative tractography methods applied to cerebral D, Klaveness S, Montillo A, Makris N, Rosen B, Dale
white matter. Neuroimage. 2007;36(3):630–44. AM. Whole brain segmentation: automated labeling
32. van der Graaff MM, Sage CA, Caan MW, Akkerman of neuroanatomical structures in the human brain.
EM, Lavini C, Majoie CB, Nederveen AJ, Neuron. 2002;33(3):341–55.
Zwinderman AH, Vos F, Brugman F, van den Berg 45. de Kwaasteniet B, Ruhe E, Caan M, Rive M,
LH, de Rijk MC, van Doorn PA, Van Hecke W, Olabarriaga S, Groefsema M, Heesink L, van Wingen
Peeters RR, Robberecht W, Sunaert S, de Visser G, Denys D. Relation between structural and func-
M. Upper and extra-motoneuron involvement in early tional connectivity in major depressive disorder. Biol
motoneuron disease: a diffusion tensor imaging study. Psychiatry. 2013;74(1):40–7.
Brain. 2011;134(Pt 4):1211–28. 46. Kaden E, Knösche TR, Anwander A. Parametric
33. Jones DK. Determining and visualizing uncertainty in spherical deconvolution: inferring anatomical con-
estimates of fiber orientation from diffusion tensor nectivity using diffusion MR imaging. Neuroimage.
MRI. Magn Reson Med. 2003;49(1):7–12. 2007;37(2):474–88.
34. Parker GJM, Alexander DC. Probabilistic anatomical 47. Panagiotaki E, Schneider T, Siow B, Hall MG,
connectivity derived from the microscopic persistent Lythgoe MF, Alexander DC. Compartment models of
angular structure of cerebral tissue. Philos Trans R the diffusion MR signal in brain white matter: a
Soc Lond B Biol Sci. 2005;360(1457):893–902. taxonomy and comparison. Neuroimage.
35. Behrens TEJ, Johansen-Berg H, Woolrich MW, Smith 2012;59(3):2241–54.
SM, Wheeler-Kingshott CAM, Boulby PA, Barker 48. Honey CJ, Sporns O, Cammoun L, Gigandet X,
GJ, Sillery EL, Sheehan K, Ciccarelli O, Thompson Thiran JP, Meuli R, Hagmann P. Predicting human
AJ, Brady JM, Matthews PM. Non-invasive mapping resting-state functional connectivity from structural
of connections between human thalamus and cortex connectivity. Proc Natl Acad Sci U S A.
using diffusion imaging. Nat Neurosci. 2009;106(6):2035–40.
2003;6(7):750–7. 49. Vernooij MW, de Groot M, van der Lugt A, Ikram
36. Lazar M. Mapping brain anatomical connectivity MA, Krestin GP, Hofman A, Niessen WJ, Breteler
using white matter tractography. NMR Biomed. MMB. White matter atrophy and lesion formation
2010;23(7):821–35. explain the loss of structural integrity of white matter
37. Yendiki A, Panneck P, Srinivasan P, Stevens A, Zöllei in aging. Neuroimage. 2008;43(3):470–7.
L, Augustinack J, Wang R, Salat D, Ehrlich S, Behrens 50. Fink F, Klein J, Lanz M, Mitrovics T, Lentschig M,
T, Jbabdi S, Gollub R, Fischl B. Automated probabi- Hahn HK, Hildebrandt H. Comparison of diffusion
listic reconstruction of white-matter pathways in tensor-based tractography and quantified brain atro-
health and disease using an atlas of the underlying phy for analyzing demyelination and axonal loss in
anatomy. Front Neuroinform. 2011;5:23. MS. J Neuroimaging. 2010;20(4):334–44.
38. Caan MWA, Barth PG, Niermeijer J-M, Majoie CB, 51. Calamante F, Tournier J-D, Jackson GD, Connelly
Poll-The BT. Ectopic peripontine arcuate fibres, a A. Track-density imaging (TDI): super-resolution
228 M.W.A. Caan

white matter imaging using whole-brain track-density detection of myelin and prospects for quantitative
mapping. Neuroimage. 2010;53(4):1233–43. imaging of myelin density. Proc Natl Acad Sci U S A.
52. Dhollander T, Emsell L, Van Hecke W, Maes F, 2012;109(24):9605–10.
Sunaert S, Suetens P. Track orientation density imag- 56. Behrens TEJ, Sporns O. Human connectomics. Curr
ing (TODI) and track orientation distribution (TOD) Opin Neurobiol. 2012;22(1):144–53.
based tractography. Neuroimage. 2014;94:312–36. 57. Basser PJ, Pajevic S, Pierpaoli C, Duda J, Aldroubi
53. Willats L, Raffelt D, Smith RE, Tournier J-D, A. In vivo fiber tractography using DT-MRI data.
Connelly A, Calamante F. Quantification of track- Magn Reson Med. 2000;44(4):625–32.
weighted imaging (TWI): characterisation of within- 58. Kurki TJI, Laalo JP, Oksaranta OM. Diffusion tensor
subject reproducibility and between-subject tractography of the uncinate fasciculus: pitfalls in
variability. Neuroimage. 2013;87:18–31. quantitative analysis due to traumatic volume
54. Mangin J-F, Fillard P, Cointepas Y, Le Bihan D, changes. J Magn Reson Imaging. 2013;38(1):46–53.
Frouin V, Poupon C. Toward global tractography. 59. Vos SB, Jones DK, Viergever MA, Leemans A. Partial
Neuroimage. 2013;80:290–6. volume effect as a hidden covariate in DTI analyses.
55. Wilhelm MJ, Ong HH, Wehrli SL, Li C, Tsai P-H, Neuroimage. 2011;55(4):1566–76.
Hackney DB, Wehrli FW. Direct magnetic resonance
Normal Diffusion Tensor Imaging-
Based White Matter Anatomy 12
Bram Stieltjes

Learning Points Representation of White Matter


Anatomy Using Diffusion Tensor
• Normal diffusion tensor imaging-based anat- Imaging
omy of the healthy brain.
• The identification of several major white mat- As diffusion tensor imaging permits estimation
ter fiber bundles on 2D directionally encoded of the main nerve fiber direction within a voxel,
color fractional anisotropy maps in axial, cor- a natural extension is to reconstruct the course
onal, and sagittal views. of whole nerve tracts [1–7]. Figure 12.1 shows
• The identification of common 3D virtual a sketch of a brain with diffusion tensor ellip-
reconstructions of DTI-based white matter soids plotted in several brain voxels. The con-
fiber tracts. sidered person is watching a red-leaved flower.
• Region-of-interest placement in order to virtu- This information is caught by the eye and
ally reconstruct commonly identified DTI- passed to the visual brain cortex through the
based white matter fiber tracts. optical tract. This tract is observable in the ten-
sor ellipsoids: they are elongated in tract voxels
and reflect the main fiber direction. In the
sketch, the diffusion is isotropic in voxels not
containing the optic tract.
Figure 12.1b illustrates a straightforward
approach to reconstruct tracts from the tensor
ellipsoids, the so-called FACT algorithm
(FACT = fiber assignment by continuous track-
ing) [4, 5]. The tract is started in one voxel and it
follows the orientation of the ellipsoid in this
B. Stieltjes, MD, PhD (*) voxel until it hits the border of the voxel. Then,
Department of radiology and nuclear medicine, this process is iterated in the adjacent voxel.
University Hospital Basel,
Basel, Switzerland Usually several starting points—which are also
e-mail: B.Stieltjes@dkfz-heidelberg.de called seeds—are chosen. For example, in

© Springer Science+Business Media New York 2016 231


W. Van Hecke et al. (eds.), Diffusion Tensor Imaging, DOI 10.1007/978-1-4939-3118-7_12
232 B. Stieltjes

Fig. 12.1 The principle of fiber tracking. (a) Schematic ment. The optical tract shows anisotropic (ellipsoid) water
view of a person watching a flower. Information is trans- displacement and the tract can be reconstructed using dif-
ported from the eye to the visual cortex at the back of the fusion tensor imaging
head. (b) Close-up of the voxel-wise diffusion measure-

Fig. 12.1b, the tract starting in the center of the ground truth. Nonetheless, it has been shown
initial voxel does not reach the end of the optic that the main fiber tracts can be reconstructed
nerve tract, since the tract is interrupted when it correctly.
reaches a voxel with isotropic diffusion. The Since the initial works on fiber tracking,
other tract, which starts in the upper part of the many fiber tracking algorithms and strategies
initial voxel, reaches the visual brain cortex. have been developed. Since a detailed review of
Thus, by choosing several seeds instead of only these techniques is beyond the scope of this
one seed, the chance is increased that the actual introduction and covered in Chap. 11 of this
tract is reconstructed. book, here we only present an in-depth descrip-
In Fig. 12.1b, an important property of fiber tion of the algorithm used for the fiber tracking
tracking results can be appreciated: the strong in the “Three-Dimensional Tract Representation”
dependency on the chosen algorithm and its section.
parameters. The termination criterion of the
algorithm depicted in Fig. 12.1b is to stop the
tract if an isotropic voxel is reached. However, Protocol
if the tract were allowed to maintain its direc-
tion in an isotropic voxel under the condition Data Acquisition
that the next voxel is anisotropic, then the
broken fiber would make it to the end of the The data were acquired using a 3 T scanner (TIM
optic tract. In practice, of course, voxels are Trio, Siemens, Erlangen, Germany) equipped
usually not perfectly isotropic, so one would with a 32-channel head coil. A single-shot echo-
rather choose a certain anisotropy threshold to planar imaging (EPI) sequence was applied for
terminate the fiber. The final tracking result will DTI assessment (TR 6400 ms, TE 91 ms, 96 × 96
in any case depend strongly on this threshold. matrix size, field of view 240 mm). Fifty axial
As there is often no unique and perfect choice slices with a thickness of 2.5 mm and no gap, 60
of algorithm and parameters, the obtained gradient directions, and two b-values (0 and
results should not be regarded as a perfect 1000 s/mm2) were obtained.
12 Normal Diffusion Tensor Imaging-Based White Matter Anatomy 233

Data Preprocessing The most important, central parts of the brain are
displayed in the three main radiological planes,
Eddy currents and head motion were corrected axial, coronal, and sagittal.
using FSL flirt (www.fmrib.ox.ac.uk/fsl/) by
affine registration of the baseline and diffusion
weighted volumes to the first baseline volume. Two-Dimensional Anatomy
Gradient directions were corrected according to
the transformation. FSL bet was used in order to Axial slices (Figs. 12.2, 12.3, 12.4, 12.5, 12.6,
estimate brain masks. Tensors were fit using the 12.7, 12.8, 12.9, and 12.10)
teem library (teem.sourceforge.net). Negative Coronal slices (Figs. 12.11, 12.12, 12.13,
Eigenvalues were corrected by adding (to all 12.14, 12.15, 12.16, 12.17, 12.18, and 12.19)
Eigenvalues) the amount by which the smallest is Sagittal slices (Figs. 12.20, 12.21, 12.22,
negative (corresponding to increasing the non- 12.23, 12.24, 12.25, 12.26, and 12.27)
diffusion weighted image value).

Three-Dimensional Tract
Tracking Algorithm Representation

The tractography algorithm that was used to Introduction (3D)


generate the fiber tracking results was originally
introduced by Reisert et al. [8] and is a so-called The three-dimensional part covers the most
global tracking algorithm [9, 10]. This algo- prominent white matter connections within the
rithm ranked first in an evaluation study by cerebral hemispheres. The complete reconstruc-
Fillard et.al. on the performance of tractography tion process is covered in a consistent, step-by-
algorithms [11] and is implemented in MITK- step fashion. First the relevance and anatomy of
Diffusion ([12], www.mitk.org/Difusion). The the tract is discussed and an initial region of
complete procedure is described in more detail interest (ROI) is shown. This ROI is chosen as to
in [13]. yield an optimal final result. By using inclusion
(green) and exclusion (red) ROIs, the result is
further refined. The final result is represented
Two-Dimensional Tract without the ROIs to optimally appreciate the
Representation anatomical location. The color coding of these
tracts is identical to the two-dimensional color
Introduction (2D) maps (section, Introduction 2D). The intricate
anatomy of important adjacent tracts is further
Here we present a diffusion tensor imaging illustrated in combined overviews. Here each
derived color map produced using MITK- individual tract is represented in unicolor to
Diffusion ([12], www.mitk.org/Difusion). In enhance the visualization of the complex, inter-
these maps, the directional orientation of fiber woven anatomy.
tracts is color coded in the following fashion:
tracts moving left-right are coded red (e.g., the
Corpus callosum), anterior-posterior tracts are Three-Dimensional Anatomy
coded green (e.g., the Cingulum), and cranio-
caudal tracts are coded blue (e.g., the cortico- The Corpus Callosum
spinal tract). The intensity or hue indicates the Fiber tracking of the corpus callosum is relatively
fractional anisotropy, a measure of fiber density. straightforward. Select a midsagittal on the sagit-
234 B. Stieltjes

Fig. 12.2 Axial slices of a color-coded diffusion tensor The intensity represents the fractional anisotropy (FA) also
image. The colors represent fiber direction; red = left to indicated in the lower corner. In the upper corners, the
right, blue = cranio-caudal, and green = anterior-posterior. slice section is indicated in the coronal and sagittal plane
12 Normal Diffusion Tensor Imaging-Based White Matter Anatomy 235

Fig. 12.3 Axial slices of a color-coded diffusion tensor The intensity represents the fractional anisotropy (FA) also
image. The colors represent fiber direction; red = left to indicated in the lower corner. In the upper corners, the
right, blue = cranio-caudal, and green = anterior-posterior. slice section is indicated in the coronal and sagittal plane
236 B. Stieltjes

Fig. 12.4 Axial slices of a color-coded diffusion tensor The intensity represents the fractional anisotropy (FA) also
image. The colors represent fiber direction; red = left to indicated in the lower corner. In the upper corners, the
right, blue = cranio-caudal, and green = anterior-posterior. slice section is indicated in the coronal and sagittal plane
12 Normal Diffusion Tensor Imaging-Based White Matter Anatomy 237

Fig. 12.5 Axial slices of a color-coded diffusion tensor The intensity represents the fractional anisotropy (FA) also
image. The colors represent fiber direction; red = left to indicated in the lower corner. In the upper corners, the
right, blue = cranio-caudal, and green = anterior-posterior. slice section is indicated in the coronal and sagittal plane
238 B. Stieltjes

Fig. 12.6 Axial slices of a color-coded diffusion tensor The intensity represents the fractional anisotropy (FA) also
image. The colors represent fiber direction; red = left to indicated in the lower corner. In the upper corners, the
right, blue = cranio-caudal, and green = anterior-posterior. slice section is indicated in the coronal and sagittal plane
12 Normal Diffusion Tensor Imaging-Based White Matter Anatomy 239

Fig. 12.7 Axial slices of a color-coded diffusion tensor The intensity represents the fractional anisotropy (FA) also
image. The colors represent fiber direction; red = left to indicated in the lower corner. In the upper corners, the
right, blue = cranio-caudal, and green = anterior-posterior. slice section is indicated in the coronal and sagittal plane
240 B. Stieltjes

Fig. 12.8 Axial slices of a color-coded diffusion tensor The intensity represents the fractional anisotropy (FA) also
image. The colors represent fiber direction; red = left to indicated in the lower corner. In the upper corners, the
right, blue = cranio-caudal, and green = anterior-posterior. slice section is indicated in the coronal and sagittal plane
12 Normal Diffusion Tensor Imaging-Based White Matter Anatomy 241

Fig. 12.9 Axial slices of a color-coded diffusion tensor The intensity represents the fractional anisotropy (FA) also
image. The colors represent fiber direction; red = left to indicated in the lower corner. In the upper corners, the
right, blue = cranio-caudal, and green = anterior-posterior. slice section is indicated in the coronal and sagittal plane
242 B. Stieltjes

Fig. 12.10 Axial slices of a color-coded diffusion tensor The intensity represents the fractional anisotropy (FA) also
image. The colors represent fiber direction; red = left to indicated in the lower corner. In the upper corners, the
right, blue = cranio-caudal, and green = anterior-posterior. slice section is indicated in the coronal and sagittal plane
12 Normal Diffusion Tensor Imaging-Based White Matter Anatomy 243

Fig. 12.11 Coronal slices of a color-coded diffusion ten- The intensity represents the fractional anisotropy (FA) also
sor image. The colors represent fiber direction; red = left to indicated in the lower corner. In the upper corners, the
right, blue = cranio-caudal, and green = anterior-posterior. slice section is indicated in the axial and sagittal plane
244 B. Stieltjes

Fig. 12.12 Coronal slices of a color-coded diffusion ten- The intensity represents the fractional anisotropy (FA) also
sor image. The colors represent fiber direction; red = left to indicated in the lower corner. In the upper corners, the
right, blue = cranio-caudal, and green = anterior-posterior. slice section is indicated in the axial and sagittal plane
12 Normal Diffusion Tensor Imaging-Based White Matter Anatomy 245

Fig. 12.13 Coronal slices of a color-coded diffusion ten- The intensity represents the fractional anisotropy (FA) also
sor image. The colors represent fiber direction; red = left to indicated in the lower corner. In the upper corners, the
right, blue = cranio-caudal, and green = anterior-posterior. slice section is indicated in the axial and sagittal plane
246 B. Stieltjes

Fig. 12.14 Coronal slices of a color-coded diffusion ten- The intensity represents the fractional anisotropy (FA) also
sor image. The colors represent fiber direction; red = left to indicated in the lower corner. In the upper corners, the
right, blue = cranio-caudal, and green = anterior-posterior. slice section is indicated in the axial and sagittal plane
12 Normal Diffusion Tensor Imaging-Based White Matter Anatomy 247

Fig. 12.15 Coronal slices of a color-coded diffusion ten- The intensity represents the fractional anisotropy (FA) also
sor image. The colors represent fiber direction; red = left to indicated in the lower corner. In the upper corners, the
right, blue = cranio-caudal, and green = anterior-posterior. slice section is indicated in the axial and sagittal plane
248 B. Stieltjes

Fig. 12.16 Coronal slices of a color-coded diffusion ten- The intensity represents the fractional anisotropy (FA) also
sor image. The colors represent fiber direction; red = left to indicated in the lower corner. In the upper corners, the
right, blue = cranio-caudal, and green = anterior-posterior. slice section is indicated in the axial and sagittal plane
12 Normal Diffusion Tensor Imaging-Based White Matter Anatomy 249

Fig. 12.17 Coronal slices of a color-coded diffusion ten- The intensity represents the fractional anisotropy (FA) also
sor image. The colors represent fiber direction; red = left to indicated in the lower corner. In the upper corners, the
right, blue = cranio-caudal, and green = anterior-posterior. slice section is indicated in the axial and sagittal plane
250 B. Stieltjes

Fig. 12.18 Coronal slices of a color-coded diffusion ten- The intensity represents the fractional anisotropy (FA) also
sor image. The colors represent fiber direction; red = left to indicated in the lower corner. In the upper corners, the
right, blue = cranio-caudal, and green = anterior-posterior. slice section is indicated in the axial and sagittal plane
12 Normal Diffusion Tensor Imaging-Based White Matter Anatomy 251

Fig. 12.19 Coronal slices of a color-coded diffusion ten- The intensity represents the fractional anisotropy (FA) also
sor image. The colors represent fiber direction; red = left to indicated in the lower corner. In the upper corners, the
right, blue = cranio-caudal, and green = anterior-posterior. slice section is indicated in the axial and sagittal plane
252 B. Stieltjes

Fig. 12.20 Sagittal slices of a color-coded diffusion ten- The intensity represents the fractional anisotropy (FA) also
sor image. The colors represent fiber direction; red = left to indicated in the lower corner. In the upper corners, the
right, blue = cranio-caudal, and green = anterior-posterior. slice section is indicated in the axial and coronal plane
12 Normal Diffusion Tensor Imaging-Based White Matter Anatomy 253

Fig. 12.21 Sagittal slices of a color-coded diffusion ten- The intensity represents the fractional anisotropy (FA) also
sor image. The colors represent fiber direction; red = left to indicated in the lower corner. In the upper corners, the
right, blue = cranio-caudal, and green = anterior-posterior. slice section is indicated in the axial and coronal plane
254 B. Stieltjes

Fig. 12.22 Sagittal slices of a color-coded diffusion ten- The intensity represents the fractional anisotropy (FA) also
sor image. The colors represent fiber direction; red = left to indicated in the lower corner. In the upper corners, the
right, blue = cranio-caudal, and green = anterior-posterior. slice section is indicated in the axial and coronal plane
12 Normal Diffusion Tensor Imaging-Based White Matter Anatomy 255

Fig. 12.23 Sagittal slices of a color-coded diffusion ten- The intensity represents the fractional anisotropy (FA) also
sor image. The colors represent fiber direction; red = left to indicated in the lower corner. In the upper corners, the
right, blue = cranio-caudal, and green = anterior-posterior. slice section is indicated in the axial and coronal plane
256 B. Stieltjes

Fig. 12.24 Sagittal slices of a color-coded diffusion ten- The intensity represents the fractional anisotropy (FA) also
sor image. The colors represent fiber direction; red = left to indicated in the lower corner. In the upper corners, the
right, blue = cranio-caudal, and green = anterior-posterior. slice section is indicated in the axial and coronal plane
12 Normal Diffusion Tensor Imaging-Based White Matter Anatomy 257

Fig. 12.25 Sagittal slices of a color-coded diffusion ten- The intensity represents the fractional anisotropy (FA) also
sor image. The colors represent fiber direction; red = left to indicated in the lower corner. In the upper corners, the
right, blue = cranio-caudal, and green = anterior-posterior. slice section is indicated in the axial and coronal plane
258 B. Stieltjes

Fig. 12.26 Sagittal slices of a color-coded diffusion ten- The intensity represents the fractional anisotropy (FA) also
sor image. The colors represent fiber direction; red = left to indicated in the lower corner. In the upper corners, the
right, blue = cranio-caudal, and green = anterior-posterior. slice section is indicated in the axial and coronal plane
12 Normal Diffusion Tensor Imaging-Based White Matter Anatomy 259

Fig. 12.27 Sagittal slices of a color-coded diffusion ten- The intensity represents the fractional anisotropy (FA) also
sor image. The colors represent fiber direction; red = left to indicated in the lower corner. In the upper corners, the
right, blue = cranio-caudal, and green = anterior-posterior. slice section is indicated in the axial and coronal plane
260 B. Stieltjes

Fig. 12.28 Tracking of


the Corpus callosum (CC),
first step. (a) Midsagittal
view of the CC with the
initial starting region of
interest (ROI) in white. (b)
Removal of spurious fibers
mainly from the cingulum
using exclusion ROIs (red)

tal plane (Fig. 12.28a). The corpus callosum from this fiber tracking are shown in correspond-
appears as a central large red structure. Outline ing colors.
the structure (white outline) and you should yield
an initial tracking result that appears similar to The Cingulum
Fig. 12.28b. Since the cingulum and the fornix The cingulum is a paired parasagittal structure
run directly adjacent to the corpus callosum in that extends from the septal area to the uncus
the midsagittal plane, parts of these tracts are region of the temporal lobe. Due to its close prox-
included in the initial result. These spurious tracts imity to the corpus callosum, especially while in
can be removed by placement of two ROIs (red proximity of the cingulate gyrus, callosal fibers
ROIs, Fig. 12.28b). The first ROI is placed just are invariably included in the starting ROI. The
posteriorly of the caudal tip of the genu of the ROI outlined in white includes the left and right
corpus callosum (left red ROI) and the cingulum cingulum. The appropriate coronal plane is found
fibers from this ROI are excluded. The second when the large green structure above the corpus
ROI is placed anteriorly of the caudal tip of the callosum is selected in the sagittal plane
splenium of the corpus callosum (right red ROI). (Fig. 12.30a). The callosal fibers can be easily
This ROI excludes fibers from the fornix and the removed by using the midsagittal ROI previously
posterior aspect of the cingulum. used as starting ROI for tracking of the corpus
The previous result can be segmented further callosum. The final result is in Fig. 12.30b.
into three distinct areas of the corpus callosum,
the anterior part or genu, the central part or body, The Fornix
and the posterior aspect or splenium. To achieve Fiber tracking of the fornix is quite challenging.
this segmentation, the first tracking result is taken First, it is a very thin structure with a relatively
and three new ROIs are drawn (Fig. 12.29a). The low fractional anisotropy, most probably due to
red ROI delineates the midsagittal aspect of the substantial partial volume effects of the sur-
genu, the yellow ROI the body, and the green rounding gray matter and cerebrospinal fluid.
ROI the splenium. In Fig. 12.29b, the results This low anisotropy may cause fiber tracking to
12 Normal Diffusion Tensor Imaging-Based White Matter Anatomy 261

Fig. 12.29 Tracking of


the Corpus callosum,
second step. (a)
Midsagittal view of the CC
with the initial starting ROI
in white and the sub-
segmentation in red,
yellow, and green. (b)
Tracking result in colors
corresponding to the initial
ROIs

Fig. 12.30 Tracking of


the bilateral cingulum
bundle (CB). (a) Initial
ROI encompassing the CB
bilaterally (white) in the
coronal plane. (b) Tracking
result overlaid on a sagittal
fiber density map

be aborted. Second, it is a highly curved struc- major tracts related to the hippocampus and
ture that poses additional difficulties for fiber memory function. Its fibers run along the medial
tracking algorithms. And finally, the fornix lies aspect of the hippocampus, forming the fimbria,
in close proximity to other fiber bundles such as that project posterosuperiorly. The bilateral fib-
the corpus callosum and the anterior commissure ria form the crus, run over the thalamus, and then
that further hamper fiber reconstruction. The for- join to form the body. Running frontally, the cor-
nix is of great interest, especially in psychiatric pus separates again to form the column and the
research on Alzheimer’s, since it is one of the majority of the fibers terminate in the mammil-
262 B. Stieltjes

Fig. 12.31 Unilateral


tracking of the fornix (FX).
(a) Initial ROI (white
circle) encompassing the
FX in the coronal plane.
(b) Tracking result overlaid
on a sagittal fiber density
map

Fig. 12.32 (a) Initial ROI


(white circle) for all the
following tracts. (b)
Through the red exclusion
ROI, the Uncinate
Fasciculus (UF) is
excluded

lary body. The body of the fornix is a good start- The Inferior Longitudinal-, Inferior
ing region best found in the midsagittal plane. Fronto-Occipital- and Uncinate
Then, the full fornix can be delineated on the Fasciculus
corresponding coronal slice (Fig. 12.31a). The Fiber tracking of the inferior longitudinal fascic-
initial result contains spurious fibers from the ulus can be started from the temporal lobe. As
corpus callosum and the anterior commissure indicated in Fig. 12.32a, a large, somewhat non-
that can be removed subsequently. The final specific starting region can be chosen to encom-
result as shown in Fig. 12.31b is somewhat frag- pass the temporal lobe on the coronal slice (right).
ile and, depending on your data, may look less This yields a large complex of fiber bundles
well defined. (Fig. 12.32b) that can be further dissected.
12 Normal Diffusion Tensor Imaging-Based White Matter Anatomy 263

Fig. 12.33 Tracking of the


inferior longitudinal
fasciculus (ILF). (a)
Placement of large
occipital inclusion ROI
(white circle). (b)
Depiction of the ILF
overlaid on a fiber density
map

Fig. 12.34 Tracking of


the uncinate fasciculus
(UF). (a) Starting ROI at
the fronto-temporal
junction. (b) Initial
tracking result and green
inclusion ROI

The inferior longitudinal fasciculus is the main tures. Additional spurious fibers, mainly stemming
bundle connecting the temporal and occipital lobes. from the anterior commissure and the corpus cal-
The two other major fiber bundles that are part of losum, can be excluded by a large parieto-occipital
the initial complex (Fig. 12.32b) are the uncinate- inclusion ROI (Fig. 12.33a). Figure 12.33b shows
and the inferior fronto-occipital fasciculus. These the inferior longitudinal fasciculus bilaterally.
two bundles can be easily excluded by the exclu- As mentioned in the previous section on the
sion ROI (red) indicated in Fig. 12.32b at the inferior longitudinal fasciculus, a coronal ROI
fronto-temporal junction. This is also a perfect at the fronto-temporal junction (Fig. 12.32b) is
starting ROI for these two structures. This is the ideal starting region for the reconstruction
described in the next section on these two struc- of the inferior fronto-occipital and uncinate fas-
264 B. Stieltjes

Fig. 12.35 Depiction of


the UF overlaid on a
sagittal fiber density map

Fig. 12.36 Tracking of


the inferior fronto-occipital
fasciculus (IFOF). (a)
Initial ROI as in Fig. 12.34.
(b) Initial tracking result
and green inclusion ROI

ciculus. As can be seen in the initial result in the frontal and occipital lobe, a second inclu-
Fig. 12.34b, no spurious fibers are present, and sion ROI, as depicted in Fig. 12.34a, b (green
in two easy steps the two bundles can be iso- ROI) within the temporal lobe, effectively iso-
lated. To better appreciate the intricate interwo- lates the uncinate fasciculus. The results can be
ven structure of the above named three fiber seen in Fig. 12.35. Note that no additional ROIs
bundles, this section is concluded with a com- are required to remove spurious fibers. Likewise,
bined overview. Since the uncinate fasciculus the inferior fronto-occipital fasciculus can be
connects the frontal and temporal lobe whereas isolated by placing an inclusion ROI directly
the inferior fronto-occipital fasciculus connects posterior to the point where both bundles join
12 Normal Diffusion Tensor Imaging-Based White Matter Anatomy 265

Fig. 12.37 Depiction of


the IFOF overlaid on an
axial fiber density map

Fig. 12.38 Combined


bilateral view of the UF
(red), the IFOF (yellow),
and the ILF (green)

(Fig. 12.36a, b) and the final result is shown in The Superior Longitudinal-, Superior
Fig. 12.37. Fronto-Occipital- and Arcuate
Figure 12.38 is an overview where the previ- Fasciculus
ously reconstructed tracts are shown in a com- The superior longitudinal, superior fronto-
bined array. The uncinate fasciculus is colored occipital and arcuate fasciculus can all be isolated
red, the inferior longitudinal fasciculus is colored from one starting ROI (Fig. 12.39a). The arcuate
green, and the inferior fronto-occipital fasciculus fasciculus especially is of great interest in
is colored yellow. neuroscience and neurosurgery alike, since it
266 B. Stieltjes

Fig. 12.39 Initial ROI


including the superior
longitudinal fasciculus
(SLF), the arcuate
fasciculus (AF), and the
superior fronto-occipital
fasciculus (SFOF). (a)
Initial unilateral ROI
placement. (b) Removal of
spurious fibers from the
internal capsule (white
ROI). (c) 3D overview of
the initial tracking result
and the exclusion ROI as
in (b) (red circle)

Fig. 12.40 Selection of


the SFOF. (a) Initial ROI
displayed on the coronal
color map. (b) Depiction of
the same inclusion ROI
(white circle) on the 3D
tracking result after
removal of the fibers from
the internal capsule

connects the speech areas of Broca and Wernicke as a starting point to isolate the individual fiber
in the dominant (mostly left) hemisphere. bundles.
Spurious fibers from the uncinate and inferior The superior fronto-occipital fasciculus runs
fronto-occipital fasciculus can be removed by slightly more medial and superiorly of the superior
using the starting ROI for these two bundles, as longitudinal fasciculus. Thus, by choosing the bun-
described in the previous section, as exclusion dle that follows this course from the initial result,
ROI. Spurious fibers from the cortico-spinal tract the superior fronto-occipital fasciculus can be iso-
can be removed by placing a large ROI in the pos- lated. A ROI drawn on a coronal section ensures
terior limb of the internal capsule (Fig. 12.39b). the complete inclusion of the tract (Fig. 12.40a, b)
The final result shown in Fig. 12.39c can be used and the result is shown in Fig. 12.41.
12 Normal Diffusion Tensor Imaging-Based White Matter Anatomy 267

Fig. 12.41 Depiction of


the SFOF overlaid on a
sagittal fiber density map

Fig. 12.42 Selection of


the AF. (a) Initial ROI
displayed on the coronal
color map. (b) Depiction of
the same inclusion ROI
(white circle) on the 3D
tracking result after
removal of the fibers from
the SFOF

After isolation and exclusion of the superior ciculus can be obtained by excluding both the
fronto-occipital fasciculus, the result shown in arcuate and the superior fronto-occipital fasciculus
Fig. 12.42b includes the arcuate and superior longi- from the initial result and it is displayed in
tudinal fasciculus. Since the arcuate fasciculus is Fig. 12.44.
the only bundle of the two entering the temporal In Fig. 12.45, the previously reconstructed
plane, a ROI placed to include this region tracts are shown in a combined array. The supe-
(Fig. 12.42a, b) will effectively isolate the arcuate rior fronto-occipital fasciculus is colored red, the
fasciculus. The results of this dissection are dis- arcuate fasciculus is colored green, and the supe-
played in Fig. 12.43. The superior longitudinal fas- rior longitudinal fasciculus is colored yellow.
268 B. Stieltjes

Fig. 12.43 Depiction of


the AF overlaid on a
sagittal fiber density map

Fig. 12.44 Depiction of


the SLF overlaid on a
sagittal fiber density map.
This image was obtained
by removing the AF and
the SFOF from the initial
tracking result

The Cerebral Peduncles importance for motor function in humans. The


The cerebral peduncles contain three major fiber initial result as shown below can be obtained by
bundles, the fronto-pontine, the cortico-spinal, encompassing the cerebral peduncle on an axial
and the temporo-parieto-occipito-pontine tract. slice as indicated in Fig. 12.46a. The initial result
The most renowned of the three is the cortico- (Fig. 12.46b) includes all three tracts.
spinal tract, as it is the main connection from the The fronto-pontine tract can be selected by
motor cortex to the spinal cord and of critical placing a relatively large ROI in the frontal lobe.
12 Normal Diffusion Tensor Imaging-Based White Matter Anatomy 269

Fig. 12.45 Combined


unilateral view of the
SFOF (red), the SLF
(yellow), and the AF
(green)

Fig. 12.46 Initial tracking


of the fronto-pontine-, the
cortico-spinal-, and the
parieto-occipito-pontine
tract (FPT, CST, and POPT
respectively). (a) Initial
ROI overlaid on an axial
color map. (b) Initial
tracking result with a
combined representation of
the FPT, the CST, and the
POPT overlaid on a sagittal
fiber density map

To include all tracts, the coronal plane should be area. To include all tracts, the coronal plane
used. The cortico-spinal tract can be selected by should be used. In the overview, the previously
placing a relatively large ROI to encompass the reconstructed tracts are shown in a combined
corona radiata. To include all tracts, the axial array (Fig. 12.47). The fronto-pontine tract is col-
plane should be used. The temporo-parieto- ored red, the temporo-parieto-occipital-pontine
occipito-pontine tract can be selected by placing tract is colored green, and the cortico-spinal tract
a relatively large ROI in the parieto-occipital is colored yellow.
270 B. Stieltjes

Fig. 12.47 Combined


unilateral view of the FPT
(red), the CST (yellow),
and the POPT (green)

human brain. Proc Natl Acad Sci U S A. 1999;


References 96(18):10422–7.
8. Reisert M, Mader I, Anastasopoulos C, Weigel M,
1. Mori S, van Zijl PC. Fiber tracking: principles and Schnell S, Kiselev V. Global fiber reconstruction
strategies - a technical review. NMR Biomed. becomes practical. NeuroImage. 2011;54(2):
2002;15(7-8):468–80. 955–62.
2. Mori S, Kaufmann WE, Davatzikos C, Stieltjes B, 9. Kreher BW, Mader I, Kiselev VG. Gibbs tracking: a
Amodei L, Fredericksen K, Pearlson GD, Melhem novel approach for the reconstruction of neuronal
ER, Solaiyappan M, Raymond GV, Moser HW, van pathways. Magn Reson Med. 2008;60(4):953–63.
Zijl PC. Imaging cortical association tracts in the 10. Mangin JF, Poupon C, Cointepas Y, Riviere D,
human brain using diffusion-tensor-based axonal Papadopoulos-Orfanos D, Clark CA, Regis J, Le
tracking. Magn Reson Med. 2002;47(2):215–23. Bihan D. A framework based on spin glass models
3. Stieltjes B, Kaufmann WE, van Zijl PC, Fredericksen for the inference of anatomical connectivity from
K, Pearlson GD, Solaiyappan M, Mori S. Diffusion diffusion-weighted MR data - a technical review.
tensor imaging and axonal tracking in the human NMR Biomed. 2002;15(7-8):481–92.
brainstem. NeuroImage. 2001;14(3):723–35. 11. Fillard P, Descoteaux M, Goh A, Gouttard S, Jeurissen
4. Xue R, van Zijl PC, Crain BJ, Solaiyappan M, Mori B, Malcolm J, Ramirez-Manzanares A, Reisert M,
S. In vivo three-dimensional reconstruction of rat Sakaie K, Tensaouti F, Yo T, Mangin JF, Poupon
brain axonal projections by diffusion tensor imaging. C. Quantitative evaluation of 10 tractography algo-
Magn Reson Med. 1999;42(6):1123–7. rithms on a realistic diffusion MR phantom.
5. Mori S, Crain BJ, Chacko VP, van Zijl PC. Three- NeuroImage. 2011;56(1):220–34.
dimensional tracking of axonal projections in the
brain by magnetic resonance imaging. Ann Neurol.
1999;45(2):265–9.
6. Hahn HK, Klein J, Nimsky C, Rexilius J, Peitgen MITK Diffusion Imaging
HO. Uncertainty in diffusion tensor based fibre track-
ing. Acta Neurochir Suppl. 2006;98:33–41. 12. Fritzsche KH, Neher PF, Reicht I, van Bruggen T,
7. Conturo TE, Lori NF, Cull TS, Akbudak E, Snyder Goch C, Reisert M, Nolden M, Zelzer S, Meinzer HP,
AZ, Shimony JS, McKinstry RC, Burton H, Raichle Stieltjes B. MITK Diffusion Imaging. Methods Inf
ME. Tracking neuronal fiber pathways in the living Med. 2012;51(5):441–8.
12 Normal Diffusion Tensor Imaging-Based White Matter Anatomy 271

13. Stieltjes B, Brunner R, Fritzsche KH, Laun Johansen-Berg H, Behrens TEJ. Diffusion MRI; from
FB. Diffusion tensor imaging; introduction and atlas. quantitative measurement to in vivo neuroanatomy.
Heidelberg: Springer; 2012. Amsterdam: Academic; 2009. Chapters 5-7 give a
great overview of the relationship between micro-
structure and DTI-derived parameters.
Stieltjes B, Brunner R, Fritzsche KH, Laun
Suggested Reading FB. Diffusion tensor imaging; introduction and
atlas. New York: Springer; 2012. Gives a more
Nieuwenhuys R, Voogd J, Van Huijzen C. The human detailed introduction to diffusion imaging and rich
central nervous system; a synopsis and atlas. anatomical reference.
New York: Springer; 1988. A great anatomical work of
reference for brain anatomy.
The Role of DTI in Multiple
Sclerosis and Other Demyelinating 16
Conditions

Massimo Filippi, Elisabetta Pagani, Paolo Preziosa,


and Maria Assunta Rocca

disease, correlate with clinical manifestations


Learning Points (locomotor disability and cognitive impair-
ment), and tend to worsen over time.
• Multiple sclerosis (MS) white matter (WM) • Fiber tracking based on DTI and voxel-based
lesions show increased mean diffusivity (MD) approaches for the definition of regional dis-
and decreased fractional anisotropy (FA), with tribution of damage hold promise for the
the most severe abnormalities seen in nonen- assessment of specific WM tracts, whose
hancing T1-hypointense lesions. No clear-cut damage might be critical for the development
differences have been detected between of irreversible disability.
enhancing and nonenhancing lesions. • The role of DTI in the diagnostic workup of
Increased MD may precede the development patients with different demyelinating condi-
of MS lesions visible on conventional mag- tions deserves further investigation.
netic resonance imaging (MRI).
• In MS, DTI abnormalities are detected in the
normal-appearing WM, gray matter, optic Introduction
nerves, and spinal cord.
• MS-related DTI abnormalities are more severe Multiple sclerosis (MS) is the most common
in patients with the progressive forms of the chronic inflammatory demyelinating disease
affecting the central nervous system (CNS) of
young adults in Western countries leading, in
M. Filippi, MD (*) • P. Preziosa, MD most cases, to severe and irreversible clinical dis-
M.A. Rocca, MD ability. Due to its sensitivity in the detection of
Neuroimaging Research Unit, and Department of white matter (WM) lesions, conventional mag-
Neurology, Institute of Experimental Neurology,
netic resonance imaging (MRI) has become a
Division of Neuroscience, San Raffaele Scientific
Institute, Vita-Salute San Raffaele University, paraclinical tool central to the diagnosis of MS
Via Olgettina 60, 20132 Milan, Italy and to monitor its evolution. Despite this, the cor-
e-mail: filippi.massimo@hsr.it relation between patients’ clinical status and MRI
E. Pagani, PhD measures is often weak to moderate. Diffusion
Neuroimaging Research Unit, Institute of tensor (DT) MRI is a quantitative technique with
Experimental Neurology, Division of Neuroscience,
a higher specificity toward the heterogeneous
San Raffaele Scientific Institute, Vita-Salute San
Raffaele University, Via Olgettina 60, 20132 pathological substrates of MS than conventional
Milan, Italy MRI. Indeed, correlative MR/pathological studies

© Springer Science+Business Media New York 2016 331


W. Van Hecke et al. (eds.), Diffusion Tensor Imaging, DOI 10.1007/978-1-4939-3118-7_16
332 M. Filippi et al.

have shown that the main pathological correlates full brain coverage) [5, 6]. The duration of such
of diffusivity abnormalities in MS are demyelin- an acquisition is about 10 min, during which the
ation and axonal loss [1, 2], which show a stron- patient is requested to lie still. Sequences with
ger correlation with anisotropy than with longer acquisition times, which allow a more
diffusivity indices [1, 2]. sophisticated analysis, can also be used. However,
DTI has been widely applied to measure and they are not advisable for all patients. Since the
grade MS burden not only within T2 white matter effect of demyelinating diseases on the myelin-
(WM) lesions but also in the normal-appearing ated portions of the CNS is usually diffuse and
WM (NAWM), gray matter (GM), optic nerve, unevenly distributed, imaging of the cervical
and spinal cord of patients at different stages of cord and optic nerves using DTI is of interest for
the disease [3, 4]. Such an extensive application the clinician. Nevertheless, obtaining reliable
has markedly contributed to improving our DTI estimates from these structures is more chal-
understanding of the different pathophysiological lenging than for the brain. Both these structures
substrates of the disease and the complexity of its are small and have a larger surface area to volume
clinical manifestations and evolution. ratio than the brain. This leads to a decreased
DTI has also been used to quantify CNS image quality from partial volume contamination
involvement in other demyelinating conditions, from the cerebrospinal fluid (CSF). Moreover,
which may mimic MS, such as neuromyelitis DTI is prone to movement artifacts and this is a
optica (NMO) and acute disseminated encepha- major issue especially in the study of the optic
lomyelitis (ADEM). After discussing the chal- nerves which are mobile structures, and of the
lenges specific to the use of DTI in MS and other spinal cord, where image quality may be affected
demyelinating diseases, this chapter summarizes by cardiac pulsation and swallowing. Since an in-
the main findings derived from the application of plane resolution of at least one square mm is rec-
DTI in these conditions and future possible ommended, the available single-shot echo planar
developments of the technique. imaging (EPI) technique may not be adequate. It
would be better to combine this acquisition
scheme with methods for reducing the field of
view [7, 8], which allow an improved resolution
Special Challenges of DTI with the same matrix and acquisition time, but
in the Assessment of Demyelinating avoiding problems of fold over. A multi-shot
Diseases acquisition scheme can be considered an alterna-
tive strategy to achieve a better image resolution
The application of DTI for the study of patients with less geometric distortions. However, these
with demyelinating conditions presents several techniques require longer acquisition times and
challenges related to: (1) the definition of acqui- are prone to image ghosting, if not adequately
sition protocols, both in terms of sequence setting corrected. To this end, other methods have been
and duration; (2) the selection of the most appro- developed, which are not based on a Cartesian
priate method for the analysis of the data; and (3) reading of the k-space and consider other trajec-
the interpretation of the results. tories. One of these methods is the so-called
PROPELLER technique [9] that acquires, at each
diffusion preparation, a central strip, rotated from
Setup of the Acquisition Protocol time to time at each excitation until the k-space is
entirely filled. The advantage of using
Usually, MS patients can tolerate acquisition pro- PROPELLER sequences is that each segment can
tocols with a relatively long duration, unless they be translated into the k-space to correct for varia-
are in an advanced and disabled stage of the dis- tion of the phase. These techniques are now more
ease. As a consequence, it is reasonable to use readily available on clinical scanners and should
optimized DTI sequences (i.e., at least 30 be considered as a feasible alternative to standard
diffusion-weighted directions, b factor ≈ 1000, single-shot methods.
16 The Role of DTI in Multiple Sclerosis and Other Demyelinating Conditions 333

Fig. 16.1 An example of diffusion tensor imaging (DTI) of the CC is shown at the bottom. In the MS patient, sev-
tractography analysis (diffusion toolkit http://trackvis.org/ eral periventricular lesions, causing a reduction of FA val-
dtk) of a healthy control (HC) (left side) and a patient with ues, are visible on the B0 image. Diffuse atrophy is also
secondary progressive multiple sclerosis (MS) (right evident (enlargement of the ventricles and thinning of the
side). The B0 image and fractional anisotropy (FA) map CC on the midsagittal slice). The reconstruction of the CC
are shown in the top part of the figure. In the middle part, shows the interruption of fiber propagation due to the
the global reconstruction of all fibers is shown. After the presence of low FA values in lesions. Parameters used for
definition of a region of interest (ROI) for tracking the tractography: FA threshold = 0.15, angle threshold = 35°,
corpus callosum (CC) on the FA map, the reconstruction propagation algorithm: FACT with spline filter applied

Analysis of DTI Data tiple crossing fibers in the voxel of interest or by


a lesion that damages the tissue. One possibility
The main problems associated with the analysis to overcome this problem is to decouple the use
of DTI data from patients with demyelinating of DTI to segment fiber tracts from its use to cal-
conditions, both for the brain and spinal cord, are culate measures of tissue damage. To this end,
related to the presence of focal WM lesions and tractography can be used to construct a probabil-
atrophy (Fig. 16.1). At present, one of the main ity map of a tract of interest from healthy volun-
obstacles to the development of DT tractography teer data and then apply such a map to patients
in vivo, and particularly in diseased states, is sec- scans to calculate DTI indexes of that tract (Fig.
ondary to the strategy used in tracking when a 16.2) [10]. For the construction of tract atlases
fiber tract enters a region of low fractional anisot- and their application to single subject data, spa-
ropy (FA). Low FA can be caused either by mul- tial normalization strategies need to be imple-
334 M. Filippi et al.

Fig. 16.2 White matter (WM) tract probability maps sented in neurologic convention. [Adapted from Preziosa
obtained from a reference group of 24 healthy controls. P, Rocca MA, et al. (2011). “Intrinsic damage to the major
Probability maps are superimposed on axial (top row), white matter tracts in patients with different clinical phe-
sagittal (middle row), and coronal (bottom row) sections notypes of multiple sclerosis: a voxelwise diffusion-tensor
of the fractional anisotropy (FA) atlas. A anterior, I infe- MR study.” Radiology 2011;260(2):541–50. With permis-
rior, L left, P posterior, R right, S superior. Images are pre- sion from The Radiological Society of North America]

mented, so that morphological and positioning allows matching of the shape, magnitude, and
differences between subjects are corrected. The orientation of local tensors between images bet-
need for nonlinear deformation is particularly ter than that of one channel of FA or one channel
important when compensating for differences of T2-weighted image.
caused by brain atrophy.
Likewise, voxel-based analysis depends
strongly on nonlinear registration methods and Interpretation of DTI Findings
on their capability to produce an adequate over-
lap between subjects. However, the application The interpretation of DTI findings is not straight-
of a nonlinear deformation introduces new forward. So far, the actual pathological features
issues related on how to use additional pieces of underlying diffusion abnormalities in MS are not
information on tract morphology in order to completely understood. Some pieces of evidence
drive the registration. This is the consequence of suggest that the various, and often concomitant,
an increased ability of this algorithm to com- pathological abnormalities occurring in MS (i.e.,
pensate for local morphological differences. It inflammation, demyelination, axonal loss,
has been shown that the accuracy of registration Wallerian degeneration) can affect diffusivity
is higher when the whole DTI acquisition set is and anisotropic characteristics of tissues in
used to drive the transformation. For instance, opposing ways, thereby complicating the inter-
the use of six channels of tensor components pretation of DTI findings [1, 2, 11].
16 The Role of DTI in Multiple Sclerosis and Other Demyelinating Conditions 335

DTI Features of MS earliest stages of the disease, also in patients


with clinically isolated syndrome (CIS) sugges-
Focal WM Lesions tive of MS [21, 22], and become more pro-
nounced with increasing disease duration and
DTI can contribute to grade the severity of dam- neurological impairment [3, 4]. NAWM dam-
age within focal MS lesions, which typically age in MS patients is distributed but tends to be
show increased mean diffusivity (MD) and more severe in perilesional areas [23, 24] and at
decreased fractional anisotropy (FA). However, sites where MS lesions are typically located
abnormalities of DTI indexes are highly hetero- [14, 25].
geneous among different T2 lesions [3, 4]. The NAWM damage is only partially correlated
more severe abnormalities are found in T1 with the extent of T2 lesions and the severity of
hypointense lesions (the so-called black holes) intrinsic lesion damage [3], which suggests that
[12–14], which represent areas of irreversible tis- diffusivity changes in NAWM are not entirely
sue disruption, gliosis, and axonal loss. dependent on retrograde degeneration of axons
Conversely, conflicting results have been reported transected in focal lesions, but they rather repre-
when comparing findings in gadolinium- sent diffuse abnormalities beyond the resolution
enhancing vs. nonenhancing lesions [12–15], of conventional MRI. A study [26] correlated dif-
although most of the studies found lower FA val- fusivity with perfusion findings in the corpus cal-
ues in enhancing lesions [14, 16]. These findings losum of patients with relapsing-remitting (RR)
suggest that DTI might grade intrinsic abnormal- MS. These results are more consistent with a pri-
ities of acute lesions, which in turn are likely to mary ischemia than a secondary hypoperfusion
reflect different substrates, some of which are due to Wallerian degeneration. These findings
transient (e.g., edema, demyelination, and remy- suggest a potentially reversible substrate of tissue
elination) and others, permanent (e.g., neurode- damage, related to vascular impairment, whereas
generation and axonal loss). Moreover, a hypometabolism from axonal degeneration
longitudinal study of enhancing MS lesions fol- would represent an advanced and irreversible
lowed up for 1–3 months [15] showed that MD condition.
values were increased in all lesions, but contin- DTI is also useful for assessing the evolution
ued to increase during follow-up only in a sub- of MS damage over time [21, 27]. Changes of
group of them. This finding highlights the notion DTI metrics seem to be independent of the con-
that contrast enhancement does not allow the dif- comitant accumulation of focal lesions and
ferentiation of acute MS lesions, which might be reduction in brain volume [27]. Thus, the appli-
characterized by varying degrees of tissue cation of DTI in monitoring the evolution of
disruption. MS-related tissue damage over time looks prom-
Compared to acute vascular neurological dis- ising for possible future trials of neuroprotective
eases such as stroke, a restricted diffusion has therapies.
been reported in only a few lesions in patients
with ADEM and MS in the acute phase of lesion
formation [17–19]. GM

DTI abnormalities have been demonstrated also


NAWM in the GM of MS patients [3, 4, 25, 28, 29]. They
tend to be more pronounced in patients with the
Using different methods of analysis, increased progressive forms of the disease [4, 29, 30], espe-
MD and decreased FA have been consistently cially in those with secondary progressive (SP)
found in the NAWM of MS patients [3, 4], even MS [29]. In patients with SPMS and primary pro-
before the formation of new focal lesions [20]. gressive (PP) MS [31, 32], as well as in those
Such abnormalities can be detected from the with RRMS [33] and CIS [34], such DTI abnor-
336 M. Filippi et al.

malities worsen over time. The severity of GM suppressed zonal oblique multisection echo pla-
damage has been correlated with the degree of nar imaging (ZOOM-EPI) sequence [7, 42, 43],
cognitive impairment in mildly disabled RRMS full DTI measurements from the optic nerve have
patients [35] and has been found to predict accu- been obtained.
mulation of disability over a 5-year period in It has been shown that in patients in the
patients with PPMS [32]. chronic phase following optic neuritis, MD of the
Despite the presence of conflicting results, diseased optic nerve is significantly higher than
higher MD values have been detected in deep in either the fellow eye or those from healthy
GM nuclei of MS patients, especially in the thal- individuals [42]. DTI abnormalities were found
amus [36]. Such abnormalities are more pro- to be correlated with abnormal visual evoked
nounced in SPMS than in RRMS patients [36]. In potentials (VEPs) latencies [42], loss of visual
another study, average NAWM MD and FA tha- acuity [42, 44], and retinal nerve fiber layer thin-
lamic changes over 1 year follow-up were predic- ning at optic coherence tomography, particularly
tors of clinical deterioration after 5 years in at high contrast and in nerves previously affected
PPMS patients [37]. by optic neuritis [44]. A multiparametric MRI
Possible explanations for the observed study showed that, 4 years after a unilateral optic
changes in GM diffusivity include a retrograde neuritis, decreased optic nerve FA and volume
degeneration of neurons caused by injury to WM are factors independently associated with visual
fiber tracks [38] and the presence of otherwise dysfunction [45].
undetected MS lesions in the GM. These hypoth-
eses are supported by the observation that GM
damage is only partially correlated with the Spinal Cord
extent of focal WM lesions and the severity of
their intrinsic damage [3, 4]. With the development of sophisticated MR
receiver coils and fast imaging techniques, it has
been possible to obtain more reliable imaging of
Cortical Lesions the spinal cord, which also allows the acquisition
of quantitative data, such as DTI. Clark et al. [46]
Pathologic and MRI studies have shown that cor- found higher MD values in spinal cord lesions of
tical lesions (CLs) are frequent in MS, especially MS patients than in spinal cord from healthy con-
in the progressive forms of the disease. Two DTI trols. Abnormal DTI quantities from the cervical
studies have demonstrated that, compared to cord have been shown in patients with established
patients’ GM, intracortical MS lesions have MS [47, 48] and NMO [49] but not in those with
increased FA values [39, 40], which might reflect CIS [50]. Cervical cord damage outside focal
an intralesional loss of dendrites, neuronal dam- lesions is diffuse in SPMS, while it is more lim-
age, and activation of microglial cells. One of ited in BMS patients [51]. Several studies high-
these studies [40] also found that quantifying CL lighted that brain and cord DTI metrics are
damage using DTI can help to distinguish the dif- independently associated with MS disability [27,
ferent MS clinical phenotypes, particularly 48, 49], thus calling for an aggregate use of these
benign (B) MS from SPMS patients (Fig. 16.3). measures to improve the understanding of dis-
ease progression.
One longitudinal study which obtained
Optic Nerve DTI from relapse-onset MS patients at base-
line and after a mean period of 2.4 years
Early DTI studies of the optic nerve measured showed that baseline cord cross-sectional area
diffusivity in a few directions [41]. With the and FA correlate with increased disability at
introduction of high-resolution fat- and CSF- follow-up [27 ].
16 The Role of DTI in Multiple Sclerosis and Other Demyelinating Conditions 337

0.40 FA 1.20 MD

1.00

0.30

0.80

0.20 0.60

0.40

0.10

0.20

0.00 0.00
Cortex CLs NAWM T2 Lesions Cortex CLs NAWM T2 Lesions

HC (41) RRMS (35) SPMS (23) BMS (12)

Fig. 16.3 Diffusion tensor imaging (DTI) values from ments was similar in SPMS and benign (B) MS patients.
cortical lesions (CLs), normal-appearing white matter Conversely, compared to SPMS, BMS patients had a lower
(NAWM), and T2 lesions in multiple sclerosis (MS) fractional anisotropy (FA) and mean diffusivity (MD) of
patients with different disease clinical phenotypes and a CLs. No difference was detected between RRMS and
group of healthy controls (HC). Compared to relapsing- BMS patients. MD is expressed in units of mm2/s × 10−3,
remitting (RR) MS patients, those with secondary progres- FA is dimensionless index. [Adapted from Filippi M,
sive (SP) MS had a higher T2 lesion volume and a more Preziosa P, et al. Microstructural MR imaging of cortical
severe damage to the cortex, NAWM, T2 lesions, and CLs. lesion in multiple sclerosis. Mult Scler. 2013;19(4):418–
With the exception of CLs, damage to all these compart- 26. With permission from Sage Publications]

Novel Strategies of Analysis in MS patients compared to healthy controls.


Moreover, tract-derived DTI metrics correlate
Tractography with several measures of locomotor disability
and cognitive impairment [10, 22, 53–62]. For
Thanks to the ability of DTI to depict anisotropic instance, MD and FA values of the CST corre-
tissues [52] and to detect their intrinsic structural lated with clinical measures of locomotor disabil-
abnormalities, several approaches have been ity or the pyramidal functional system score of
developed to investigate damage to selected WM the expanded disability status scale (EDSS) more
tracts, with the ultimate goal of improving the than T2 lesion volume and the overall extent of
correlation with clinical measures. DTI tractog- diffusivity changes of the brain [53–55]. CIS
raphy can be used to segment clinically eloquent patients with motor impairment had increased
WM pathways involved in different functions, MD and T2 lesion volume in the CST compared
such as the corticospinal tract (CST) [10, 53–55], to patients without pyramidal symptoms [10].
the corpus callosum (CC) [56], optic radiations Increased MD of the CC has been associated
(OR) [57], and many others [22, 58–60]. In line with MS cognitive dysfunction [54, 63].
with studies performed using histogram or Compared to cognitively unimpaired BMS
regions-of-interest analysis, these studies have patients, those with cognitive impairment had
detected higher MD and lower FA of WM tracts significantly higher NAWM MD of the CC [56].
338 M. Filippi et al.

By applying a random forest analysis to mea- NAWM, from the main clinical phenotypes of
sures derived from DTI tractography, damage to MS [22]. Compared to healthy controls, diffusiv-
“critical” WM tracts, such as the cingulum, was ity abnormalities were found in PPMS and CIS
shown to contribute significantly to the cognitive patients. The progressive MS forms showed the
impairment of MS patients [62]. most severe and distributed diffusivity abnormal-
Using DTI tractography of the ORs, one study ities, whereas BMS patients had only a limited
showed that patients with optic neuritis had WM damage. These findings support the notion
reduced connectivity values (as assessed by the that the assessment of regional damage using
number of reconstructed streamlines via tractog- DTI may be more rewarding than that of “global”
raphy) in both ORs compared to healthy controls, brain damage in order to gain insight into the
suggesting the occurrence of transsynaptic relation between clinical status and disease bur-
degeneration secondary to optic nerve damage den in MS.
[57]. In another study, OR DTI abnormalities Tract-based spatial statistics (TBSS) is a
correlated with retinal injury, assessed using opti- technique that allows voxel-wise analysis of
cal coherence tomography, and visual impair- multi-subject DTI data. Using such a tech-
ment [61]. nique, compared to healthy controls, MS
Advances in DTI and tractography have patients had reduced FA values of several WM
spurred the development of brain neuro- tracts, which were related to deficits of specific
connectivity techniques, which define and quan- cognitive domains [70, 71]. A voxel-based
tify anatomical links between remote brain method has been developed to obtain estimates
regions by axonal fiber pathways [64]. The use of of WM tract volumes using DTI [72]: an index
these approaches has revealed reduced network of volume change is derived from the differ-
efficiency in the WM structural networks of MS ences between an FA atlas based on the
patients [65], including those at the earliest stages morphological characteristics of a reference
of the disease [66]. population and an individual subject FA map.
This approach has been successfully applied to
assess the topographical distribution of age-
Voxel-Wise Approaches related WM volume changes in healthy sub-
jects [73] and may be helpful to improve our
Voxel-wise approaches to the analysis of quanti- understanding of MS abnormalities.
tative MRI data, such as voxel-based morphom-
etry [67], hold promise for improving our ability
to study the structural features of MS damage, Other Inflammatory Demyelinating
since they assess the topographical distribution of Diseases
brain damage at a voxel level. Despite voxel-
based MRI studies of MS patients having mainly NMO
focused on measures of atrophy [68], this
approach can also be used for the analysis of DTI NMO is an inflammatory demyelinating condi-
data. A voxel-based study [69] showed that tion clinically characterized by optic neuritis and
patients with RRMS and BMS differ in terms of transverse myelitis and by the presence of a
topographical distribution of WM damage, while serum autoantibody (NMO-IgG) found to spe-
no between-group differences were found when cifically target aquaporin-4 (AQP4). A DTI study
the overall extent of WM diffusivity abnormali- revealed more severe cervical cord damage in
ties was assessed. NMO than in MS patients [49]. The assessment
By combining tractography and voxel-based of brain NAWM and GM damage in NMO
analysis, another study evaluated damage to sev- patients gave conflicting results: some authors
eral WM tracts, in terms of focal lesions and found an isolated involvement of the GM [74],
16 The Role of DTI in Multiple Sclerosis and Other Demyelinating Conditions 339

while others described an involvement of several References


WM tracts [75], which was more severe in the
ORs and CSTs [76]. 1. Mottershead JP, Schmierer K, et al. High field MRI
correlates of myelin content and axonal density in
multiple sclerosis—a post-mortem study of the spinal
cord. J Neurol. 2003;250(11):1293–301.
ADEM 2. Schmierer K, Wheeler-Kingshott CA, et al. Diffusion
tensor imaging of post mortem multiple sclerosis
brain. Neuroimage. 2007;35(2):467–77.
ADEM is classically defined as a monophasic 3. Rovaris M, Gass A, et al. Diffusion MRI in multiple
dysimmune demyelinating disease, commonly sclerosis. Neurology. 2005;65(10):1526–32.
affecting children, generally preceded by infec- 4. Rovaris M, Agosta F, et al. Diffusion tensor MR imag-
tions or vaccinations. A large effort has been ing. Neuroimaging Clin N Am. 2009;19(1):37–43.
5. Jones DK, Horsfield MA, et al. Optimal strategies for
invested in trying to define MRI parameters that measuring diffusion in anisotropic systems by mag-
are able to differentiate MS from ADEM. Using netic resonance imaging. Magn Reson Med.
DTI, no abnormalities of the normal-appearing 1999;42(3):515–25.
brain tissue and spinal cord have been detected in 6. Jones DK. The effect of gradient sampling schemes
on measures derived from diffusion tensor MRI: a
ADEM patients after the acute phase of the dis- Monte Carlo study. Magn Reson Med.
ease [77], whereas mild DTI abnormalities of the 2004;51(4):807–15.
basal ganglia have been described [78]. 7. Wheeler-Kingshott CA, Parker GJ, et al. ADC map-
ping of the human optic nerve: increased resolution,
coverage, and reliability with CSF-suppressed
ZOOM-EPI. Magn Reson Med. 2002;47(1):24–31.
Conclusions 8. Saritas EU, Cunningham CH, et al. DWI of the spinal
cord with reduced FOV single-shot EPI. Magn Reson
Conventional MRI is limited by its lack of speci- Med. 2008;60(2):468–73.
9. Pipe JG. Motion correction with PROPELLER MRI:
ficity to the heterogeneous pathological substrates application to head motion and free-breathing car-
of inflammatory and demyelinating diseases of diac imaging. Magn Reson Med. 1999;42(5):
the CNS. DTI has proved to be able to quantify 963–9.
the amount of intrinsic tissue damage of focal 10. Pagani E, Filippi M, et al. A method for obtaining
tract-specific diffusion tensor MRI measurements in
lesions and to detect more subtle abnormalities the presence of disease: application to patients with
occurring in NAWM, GM, spinal cord, and optic clinically isolated syndromes suggestive of multiple
nerve. DTI is contributing to improving the under- sclerosis. Neuroimage. 2005;26(1):258–65.
standing of the mechanisms associated with the 11. Rosso C, Remy P, et al. Diffusion-weighted MR
imaging characteristics of an acute strokelike form of
development of locomotor and cognitive impair- multiple sclerosis. AJNR Am J Neuroradiol.
ment in patients with MS as well as the heteroge- 2006;27(5):1006–8.
neity of the disease clinical phenotypes. The role 12. Bammer R, Augustin M, et al. Magnetic resonance
of DTI in the diagnostic workup of patients with diffusion tensor imaging for characterizing diffuse
and focal white matter abnormalities in multiple scle-
different demyelinating conditions deserves fur- rosis. Magn Reson Med. 2000;44(4):583–91.
ther and more extensive investigation. 13. Filippi M, Iannucci G, et al. A quantitative study of
Several approaches have been applied to ana- water diffusion in multiple sclerosis lesions and
lyze DTI data. Each of them has demonstrated normal-appearing white matter using echo-planar
imaging. Arch Neurol. 2000;57(7):1017–21.
the potential to provide useful pieces of informa- 14. Filippi M, Cercignani M, et al. Diffusion tensor mag-
tion to improve our understanding of MS patho- netic resonance imaging in multiple sclerosis.
physiology. However, the best acquisition and Neurology. 2001;56(3):304–11.
postprocessing strategies for MS studies remain a 15. Castriota-Scanderbeg A, Sabatini U, et al. Diffusion
of water in large demyelinating lesions: a follow-up
matter of debate, and the contribution of newer study. Neuroradiology. 2002;44(9):764–7.
and more sophisticated DTI techniques in the 16. Werring DJ, Clark CA, et al. Diffusion tensor imaging
study of demyelinating conditions needs to be of lesions and normal-appearing white matter in mul-
evaluated further. tiple sclerosis. Neurology. 1999;52(8):1626–32.
340 M. Filippi et al.

17. Tievsky AL, Ptak T, et al. Investigation of apparent 31. Rovaris M, Gallo A, et al. Short-term accrual of gray
diffusion coefficient and diffusion tensor anisotropy matter pathology in patients with progressive multiple
in acute and chronic multiple sclerosis lesions. AJNR sclerosis: an in vivo study using diffusion tensor
Am J Neuroradiol. 1999;20(8):1491–9. MRI. Neuroimage. 2005;24(4):1139–46.
18. Bernarding J, Braun J, et al. Diffusion- and perfusion- 32. Rovaris M, Judica E, et al. Grey matter damage pre-
weighted MR imaging in a patient with acute demye- dicts the evolution of primary progressive multiple
linating encephalomyelitis (ADEM). J Magn Reson sclerosis at 5 years. Brain. 2006;129(Pt 10):2628–34.
Imaging. 2002;15(1):96–100. 33. Oreja-Guevara C, Rovaris M, et al. Progressive gray
19. Abou Zeid N, Pirko I, et al. Diffusion-weighted imag- matter damage in patients with relapsing-remitting
ing characteristics of biopsy-proven demyelinating multiple sclerosis: a longitudinal diffusion tensor
brain lesions. Neurology. 2012;78(21):1655–62. magnetic resonance imaging study. Arch Neurol.
20. Rocca MA, Cercignani M, et al. Weekly diffusion- 2005;62(4):578–84.
weighted imaging of normal-appearing white matter 34. Rovaris M, Judica E, et al. A 3-year diffusion tensor
in MS. Neurology. 2000;55(6):882–4. MRI study of grey matter damage progression during
21. Gallo A, Rovaris M, et al. Diffusion-tensor magnetic the earliest clinical stage of MS. J Neurol.
resonance imaging detects normal-appearing white 2008;255(8):1209–14.
matter damage unrelated to short-term disease activity 35. Rovaris M, Iannucci G, et al. Cognitive dysfunction in
in patients at the earliest clinical stage of multiple patients with mildly disabling relapsing-remitting mul-
sclerosis. Arch Neurol. 2005;62(5):803–8. tiple sclerosis: an exploratory study with diffusion ten-
22. Preziosa P, Rocca MA, et al. Intrinsic damage to the sor MR imaging. J Neurol Sci. 2002;195(2):103–9.
major white matter tracts in patients with different clini- 36. Fabiano AJ, Sharma J, et al. Thalamic involvement in
cal phenotypes of multiple sclerosis: a voxelwise diffu- multiple sclerosis: a diffusion-weighted magnetic
sion-tensor MR study. Radiology. 2011;260(2):541–50. resonance imaging study. J Neuroimaging. 2003;
23. Guo AC, Jewells VL, et al. Analysis of normal- 13(4):307–14.
appearing white matter in multiple sclerosis: compari- 37. Mesaros S, Rocca MA, et al. Thalamic damage pre-
son of diffusion tensor MR imaging and magnetization dicts the evolution of primary-progressive multiple
transfer imaging. AJNR Am J Neuroradiol. 2001; sclerosis at 5 years. AJNR Am J Neuroradiol.
22(10):1893–900. 2011;32(6):1016–20.
24. Hasan KM, Gupta RK, et al. Diffusion tensor frac- 38. Evangelou N, Konz D, et al. Regional axonal loss in
tional anisotropy of the normal-appearing seven seg- the corpus callosum correlates with cerebral white
ments of the corpus callosum in healthy adults and matter lesion volume and distribution in multiple scle-
relapsing-remitting multiple sclerosis patients. rosis. Brain. 2000;123(Pt 9):1845–9.
J Magn Reson Imaging. 2005;21(6):735–43. 39. Poonawalla AH, Hasan KM, et al. Diffusion-tensor
25. Cercignani M, Bozzali M, et al. Magnetisation trans- MR imaging of cortical lesions in multiple sclerosis:
fer ratio and mean diffusivity of normal appearing initial findings. Radiology. 2008;246(3):880–6.
white and grey matter from patients with multiple 40. Filippi M, Preziosa P, et al. Microstructural MR imag-
sclerosis. J Neurol Neurosurg Psychiatry. 2001;70(3): ing of cortical lesion in multiple sclerosis. Mult Scler.
311–7. 2013;19(4):418–26.
26. Saindane AM, Law M, et al. Correlation of diffusion 41. Iwasawa T, Matoba H, et al. Diffusion-weighted
tensor and dynamic perfusion MR imaging metrics in imaging of the human optic nerve: a new approach to
normal-appearing corpus callosum: support for pri- evaluate optic neuritis in multiple sclerosis. Magn
mary hypoperfusion in multiple sclerosis. AJNR Am Reson Med. 1997;38(3):484–91.
J Neuroradiol. 2007;28(4):767–72. 42. Hickman SJ, Wheeler-Kingshott CA, et al. Optic
27. Agosta F, Absinta M, et al. In vivo assessment of cer- nerve diffusion measurement from diffusion-weighted
vical cord damage in MS patients: a longitudinal dif- imaging in optic neuritis. AJNR Am J Neuroradiol.
fusion tensor MRI study. Brain. 2007;130(Pt 2005;26(4):951–6.
8):2211–9. 43. Trip SA, Wheeler-Kingshott C, et al. Optic nerve dif-
28. Bozzali M, Cercignani M, et al. Quantification of fusion tensor imaging in optic neuritis. Neuroimage.
brain gray matter damage in different MS phenotypes 2006;30(2):498–505.
by use of diffusion tensor MR imaging. AJNR Am 44. Smith SA, Williams ZR, et al. Diffusion tensor imag-
J Neuroradiol. 2002;23(6):985–8. ing of the optic nerve in multiple sclerosis: associa-
29. Rovaris M, Bozzali M, et al. Assessment of normal- tion with retinal damage and visual disability. AJNR
appearing white and gray matter in patients with pri- Am J Neuroradiol. 2011;32(9):1662–8.
mary progressive multiple sclerosis: a diffusion-tensor 45. Kolbe S, Chapman C, et al. Optic nerve diffusion
magnetic resonance imaging study. Arch Neurol. changes and atrophy jointly predict visual dysfunction
2002;59(9):1406–12. after optic neuritis. Neuroimage. 2009;45(3):679–86.
30. Pulizzi A, Rovaris M, et al. Determinants of disability 46. Clark CA, Werring DJ, et al. Diffusion imaging of the
in multiple sclerosis at various disease stages: a mul- spinal cord in vivo: estimation of the principal diffu-
tiparametric magnetic resonance study. Arch Neurol. sivities and application to multiple sclerosis. Magn
2007;64(8):1163–8. Reson Med. 2000;43(1):133–8.
16 The Role of DTI in Multiple Sclerosis and Other Demyelinating Conditions 341

47. Agosta F, Benedetti B, et al. Quantification of cervical 64. Guye M, Bettus G, et al. Graph theoretical analysis of
cord pathology in primary progressive MS using dif- structural and functional connectivity MRI in normal
fusion tensor MRI. Neurology. 2005;64(4):631–5. and pathological brain networks. MAGMA.
48. Valsasina P, Rocca MA, et al. Mean diffusivity and 2010;23(5–6):409–21.
fractional anisotropy histogram analysis of the cervical 65. Shu N, Liu Y, et al. Diffusion tensor tractography
cord in MS patients. Neuroimage. 2005;26(3):822–8. reveals disrupted topological efficiency in white mat-
49. Benedetti B, Valsasina P, et al. Grading cervical cord ter structural networks in multiple sclerosis. Cereb
damage in neuromyelitis optica and MS by diffusion Cortex. 2011;21:2565–77.
tensor MRI. Neurology. 2006;67(1):161–3. 66. Li Y, Jewells V, et al. Diffusion tensor imaging based
50. Agosta F, Filippi M. MRI of spinal cord in multiple network analysis detects alterations of neuroconnec-
sclerosis. J Neuroimaging. 2007;17 Suppl 1:46S–9S. tivity in patients with clinically early relapsing-
51. Benedetti B, Rocca MA, et al. A diffusion tensor MRI remitting multiple sclerosis. Hum Brain Mapp.
study of cervical cord damage in benign and second- 2013;34(12):3376–91.
ary progressive MS patients. J Neurol Neurosurg 67. Ashburner J, Friston KJ. Voxel-based morphome-
Psychiatry. 2010;81(1):26–30. try—the methods. Neuroimage. 2000;11(6 Pt 1):
52. Mori S, van Zijl PC. Fiber tracking: principles and 805–21.
strategies—a technical review. NMR Biomed. 68. Filippi M, Agosta F, et al. Regional assessment of
2002;15(7–8):468–80. brain atrophy: a novel approach to achieve a more
53. Wilson M, Tench CR, et al. Pyramidal tract mapping by complete picture of tissue damage associated with
diffusion tensor magnetic resonance imaging in multi- central nervous system disorders? AJNR Am
ple sclerosis: improving correlations with disability. J J Neuroradiol. 2007;28(2):260–1.
Neurol Neurosurg Psychiatry. 2003;74(2):203–7. 69. Ceccarelli A, Rocca MA, et al. The topographical dis-
54. Lin X, Tench CR, et al. ‘Importance sampling’ in MS: tribution of tissue injury in benign MS: a 3T multipa-
use of diffusion tensor tractography to quantify rametric MRI study. Neuroimage. 2008;39(4):
pathology related to specific impairment. J Neurol 1499–509.
Sci. 2005;237(1–2):13–9. 70. Dineen RA, Vilisaar J, et al. Disconnection as a mech-
55. Lin F, Yu C, et al. Diffusion tensor tractography-based anism for cognitive dysfunction in multiple sclerosis.
group mapping of the pyramidal tract in relapsing- Brain. 2009;132(Pt 1):239–49.
remitting multiple sclerosis patients. AJNR Am 71. Roosendaal SD, Geurts JJ, et al. Regional DTI differ-
J Neuroradiol. 2007;28(2):278–82. ences in multiple sclerosis patients. Neuroimage.
56. Mesaros S, Rocca MA, et al. Corpus callosum dam- 2009;44(4):1397–403.
age and cognitive dysfunction in benign MS. Hum 72. Pagani E, Horsfield MA, et al. Assessing atrophy of
Brain Mapp. 2009;30(8):2656–66. the major white matter fiber bundles of the brain from
57. Ciccarelli O, Toosy AT, et al. Optic radiation changes diffusion tensor MRI data. Magn Reson Med.
after optic neuritis detected by tractography-based 2007;58(3):527–34.
group mapping. Hum Brain Mapp. 2005;25(3): 73. Pagani E, Agosta F, et al. Voxel-based analysis derived
308–16. from fractional anisotropy images of white matter
58. Audoin B, Guye M, et al. Structure of WM bundles volume changes with aging. Neuroimage. 2008;41(3):
constituting the working memory system in early 657–67.
multiple sclerosis: a quantitative DTI tractography 74. Rocca MA, Agosta F, et al. Magnetization transfer
study. Neuroimage. 2007;36(4):1324–30. and diffusion tensor MRI show gray matter damage in
59. Rocca MA, Pagani E, et al. Altered functional and neuromyelitis optica. Neurology. 2004;62(3):476–8.
structural connectivities in patients with MS: a 3-T 75. Liu Y, Duan Y, et al. A tract-based diffusion study of
study. Neurology. 2007;69(23):2136–45. cerebral white matter in neuromyelitis optica reveals
60. Rocca MA, Valsasina P, et al. Structural and func- widespread pathological alterations. Mult Scler.
tional MRI correlates of Stroop control in benign 2012;18(7):1013–21.
MS. Hum Brain Mapp. 2009;30(1):276–90. 76. Zhao DD, Zhou HY, et al. Diffusion tensor imaging
61. Reich DS, Smith SA, et al. Damage to the optic radia- characterization of occult brain damage in relapsing
tion in multiple sclerosis is associated with retinal neuromyelitis optica using 3.0T magnetic resonance
injury and visual disability. Arch Neurol. 2009; imaging techniques. Neuroimage. 2012;59(4):
66(8):998–1006. 3173–7.
62. Mesaros S, Rocca MA, et al. Diffusion tensor MRI 77. Inglese M, Salvi F, et al. Magnetization transfer and
tractography and cognitive impairment in multiple diffusion tensor MR imaging of acute disseminated
sclerosis. Neurology. 2012;78(13):969–75. encephalomyelitis. AJNR Am J Neuroradiol.
63. Lin X, Tench CR, et al. Use of combined conventional 2002;23(2):267–72.
and quantitative MRI to quantify pathology related to 78. Holtmannspotter M, Inglese M, et al. A diffusion ten-
cognitive impairment in multiple sclerosis. J Neurol sor MRI study of basal ganglia from patients with
Neurosurg Psychiatry. 2008;79(4):437–41. ADEM. J Neurol Sci. 2003;206(1):27–30.
DTI in Dementing Conditions
17
Massimo Filippi, Federica Agosta,
and Edoardo Gioele Spinelli

• Using DTI, WM abnormalities can be identi-


Learning Points fied, which can be used to follow the progres-
sion from normal cognition to mild cognitive
• Diffusion-weighted imaging is useful in the impairment (MCI) and from MCI to AD.
early diagnosis of Creutzfeld–Jakob disease. • Patterns of DTI abnormalities are likely to dis-
• Diffusion tensor imaging (DTI) reveals brain criminate between AD and other dementing
microstructural abnormalities associated with conditions, such as frontotemporal lobar
the most common neurodegenerative dement- degeneration spectrum and dementia with
ing conditions. Lewy bodies.
• DTI studies in Alzheimer’s disease (AD) have
consistently found white matter (WM) micro-
structural damage in temporal and frontal
lobes, posterior cingulum, corpus callosum, Introduction
superior longitudinal fasciculus, and uncinate
fasciculus, with a posterior-to-anterior gradi- Although a detailed clinical assessment remains
ent of severity. the basis of the evaluation of a patient with sus-
pected dementia, current European [1, 2], UK
((NICE) March [3]), and US [4] guidelines rec-
ommend that structural imaging should be used
M. Filippi, MD (*) • E.G. Spinelli, MD in the assessment of people with suspected
Neuroimaging Research Unit and Department of dementia to exclude other cerebral pathologies
Neurology, Division of Neuroscience, Institute of
and to help establish subtype diagnosis. In gen-
Experimental Neurology, San Raffaele Scientific
Institute, Vita-Salute San Raffaele University, eral, the tendency is to move away from simply
Via Olgettina 60, 20132 Milan, Italy excluding other (brain) diseases, towards finding
e-mail: filippi.massimo@hsr.it specific pointers to a diagnosis [5]. This approach
F. Agosta, MD, PhD is exemplified by the formal incorporation of bio-
Neuroimaging Research Unit, Division of markers, including those from neuroimaging, in
Neuroscience, Institute of Experimental Neurology,
the most recent revisions of the diagnostic crite-
San Raffaele Scientific Institute, Vita-Salute San
Raffaele University, Via Olgettina 60, 20132 ria for Alzheimer’s disease (AD) [6, 7] and other
Milan, Italy neurodegenerative forms of dementia [8–10].

© Springer Science+Business Media New York 2016 343


W. Van Hecke et al. (eds.), Diffusion Tensor Imaging, DOI 10.1007/978-1-4939-3118-7_17
344 M. Filippi et al.

Recent years have also witnessed an impres- of the brain. Variant CJD is believed to have
sive advancement in the development of novel occurred as a consequence of the epidemic of
imaging approaches, which, with varying degrees bovine spongiform encephalopathy in the UK.
of success, have improved our understanding of Currently, DWI is more useful for the diagno-
the pathophysiology of dementing conditions. sis of prion disease than for any other dementia
These new techniques, which include diffusion- [14, 15]. DWI can show focal changes in CJD not
weighted (DWI) and diffusion tensor (DTI) yet apparent on fluid-attenuated inversion recov-
imaging, are likely able to fill voids and improve ery (FLAIR) images (up to 20 % of cases) [14,
our ability to diagnose, monitor and understand 15]. The better performance of DW MRI over
the pathophysiology of these diseases. DWI is FLAIR in the identification of hyperintensities
commonly used in the diagnostic work up of rap- suggests that diffusion restriction is a crucial fea-
idly progressive dementia cases, particularly ture of CJD pathology, probably due to a process
when Creutzfeld–Jakob disease (CJD) is sus- of vacuolation [16]. The combined presence of
pected [1]. Although, at present, DTI does not DWI and/or FLAIR hyperintensities has the
have a role in the diagnosis, routine assessment, highest sensitivity and specificity for the diagno-
and monitoring of neurodegenerative dementia sis of sCJD (83–96 % sensitivity, 83–95 % speci-
[1], significant efforts are underway in order to ficity) [14, 15, 17–19]. The recently updated
achieve harmonization of both acquisition and diagnostic criteria of sCJD include high signal
post-processing procedures [11], which are likely abnormalities on DWI or FLAIR in the caudate
to contribute to a dramatic change of the clinical nucleus and putamen or in at least two cortical
scenario. This chapter provides an overview of regions as one of the supportive markers for a
the findings obtained using DWI and DTI in diagnosis of probable CJD, together with peri-
patients with neurodegenerative dementing con- odic sharp wave complexes on the electroenceph-
ditions, which support a more extensive use of alogram and 14-3-3 protein detection in the
these techniques to study disease evolution and to cerebrospinal fluid [19].
monitor drug efficacy in clinical trials. In sCJD, the most common patterns of DW
MRI hyperintensities are neocortical, limbic
and subcortical (54–68 % of cases), and neo-
Creutzfeldt–Jakob Disease cortical and limbic (24–27 % of cases) [14, 15].
No neocortical involvement is detected in 5–11
CJD is a rapidly progressive dementing syn- % of patients with sCJD [14, 15]. In sCJD, cor-
drome, eventually leading to death in a few tical areas most commonly involved are the cin-
months after disease onset [12]. Typically, the gulate gyrus, superior and middle frontal gyrus,
clinical presentation of the disease includes neu- insula, precuneus, angular gyrus, and parahip-
rological signs, such as ataxia and myoclonus. pocampal gyrus, with a relative sparing of the
Most CJD cases are sporadic (sCJD), but some precentral gyrus [15] (Fig. 17.1). In both sCJD
are genetically determined (familial CJD—fCJD) and fCJD, striatal hyperintensity almost always
or transmitted through contaminated biological shows an anterior-to-posterior gradient, with
material. The pathogenesis of sCJD remains prevalent involvement of the caudate and rela-
partly unknown, but it is believed that it is likely tive sparing of the posterior putamen [15].
due to the conversion of normal prion protein— Thalamic alterations on DWI are usually bilat-
whose physiologic function remains undeter- eral, involving the dorsomedial and posterior
mined—to proteinaceous infectious scrapie (pulvinar) regions [15]. The DWI hyperinten-
particles (PrPSc) that accumulate in and around sity in subcortical regions almost invariably
neurons, leading to widespread neuronal degen- corresponds to a hypointensity on the apparent
eration [12, 13]. This pathologic process is mac- diffusion coefficient map, a finding which con-
roscopically described as spongiform degeneration firms a pattern of restricted diffusion (Fig. 17.1)
17 DTI in Dementing Conditions 345

Fig. 17.1 Three common variations of sporadic more than the corresponding FLAIR sequences, and that
Creutzfeldt–Jakob disease presentation on MRI. (a) DWI hyperintensities often have corresponding apparent
Neocortical (solid arrow), limbic (dashed arrow), and sub- diffusion coefficient (ADC) hypointensity. ADC abnormal-
cortical gray matter hyperintensities (dotted arrow) on dif- ities are most easily identified in the basal ganglia.
fusion weighted imaging (DWI) and fluid attenuated [Reprinted from Vitali, P., E. Maccagnano, et al. (2011).
inversion recovery (FLAIR) scans. (b) Neocortical and lim- “Diffusion-weighted MRI hyperintensity patterns differen-
bic cortex involvement. (c) Limbic and subcortical involve- tiate CJD from other rapid dementias.” Neurology 76(20):
ment. Note that the DWI shows the hyperintensities much 1711–9. With permission from Wolters Kluwer Health.]

[15]. In variant CJD, there is a selective involve- these patients [22, 23]. One study, which assessed
ment of the medial and dorsal (pulvinar) tha- patients with the E200K familial variant of CJD
lamic nuclei, leading to the so-called hockey [22], found a significant decrease of fractional
stick sign [20]. anisotropy (FA) of the corticospinal tract, inter-
To our knowledge, only one study evaluated nal capsule, external capsule, fornix, and poste-
the relationship between DWI alterations and the rior thalamic radiation, which was correlated
severity of the clinical course in nine patients with with disease duration. FA alterations of fCJD
sCJD [21], demonstrating that patients harboring were mainly due to an increase of radial diffusiv-
hyperintensities in both the cortex and basal gan- ity [22], suggesting axonal damage presumably
glia experience a significantly shorter interval secondary to PrPSc propagation along WM path-
from disease onset to akinetic mutism than those ways. A second DTI study in sCJD patients did
with only cortical ribbon hyperintensity. not find WM abnormalities in the corpus callo-
Cortical and basal ganglia degeneration is an sum and posterior limb of the internal capsule,
important feature of CJD pathology. On the con- but showed a significant mean diffusivity (MD)
trary, only a very few studies investigated white decrease in the caudate and pulvinar regions rela-
matter (WM) involvement using DT MRI in tive to healthy controls [23].
346 M. Filippi et al.

Alzheimer’s Disease and Mild also showed that decreased FA in the frontal lobe
Cognitive Impairment correlates with poor performance at neuropsy-
chological assessment of executive functions of
The earliest pathologic brain abnormalities asso- AD patients [35, 47–49], as well as with the
ciated with AD develop years, if not decades, severity of neuropsychiatric symptoms [50, 51].
before the onset of the first memory symptoms DTI studies have attempted to elucidate the
[24, 25]. Such alterations include misfolded pro- earliest point at which diffusivity abnormalities
teins aggregating into extracellular amyloid β can be detected by focusing on patients with
plaques and intracellular neurofibrillary tangles, amnestic MCI [28, 33, 35, 43, 46, 52–63]. Two
followed by inflammatory damage, oxidation, recent meta-analyses showed that differences
excitotoxicity, and cell death in the central ner- between amnestic MCI and controls parallel
vous system. The prospect of experimental those between AD and controls, but fewer regions
treatment with the potential to slow or prevent reached statistical significance [26, 27], possibly
AD progression has prompted an increased inter- because MCI consists of an heterogeneous group
est in the identification of individuals with AD of individuals and there are no universally recog-
early in the course of the disease, even at the mild nized criteria to define this condition. Corpus cal-
cognitive impairment (MCI) stage [6]. losum, cingulum, parahippocampal and frontal
tracts were found to be affected in the prodromal
stages of AD [28, 43, 54, 55, 59, 60, 63].
DTI in AD and MCI Patients Interestingly, in patients with amnestic MCI,
damage to the majority of WM tracts was not
The loss of cortical neurons in AD is invariably correlated with cortical atrophy [28], thus sug-
accompanied by axonal degeneration along WM gesting that, in the early phases of the disease,
pathways. DTI studies of AD have found consis- WM abnormalities may reflect a primary WM
tently increased MD and decreased FA compared tract damage over and above cortical pathology.
with controls in several brain regions (Fig. 17.2), Similar findings have been reported in cogni-
most notably in temporal and frontal lobes, pos- tively normal individuals who were later diag-
terior cingulum, corpus callosum, superior longi- nosed with amnestic MCI at 2-year follow-up
tudinal fasciculus (SLF), and uncinate fasciculus [64]. WM alterations at baseline in these subjects
[26, 27]. A posterior-to-anterior gradient in the were predictive of subsequent cognitive decline,
severity of WM abnormalities has been observed, in the absence of significant correlation with cor-
with posterior regions being affected more tical atrophy [64]. In addition, a number of stud-
severely [26]. WM changes in AD generally fol- ies [65–72] showed a similar pattern of WM
low the anatomical pattern of cortical atrophy damage in cognitively normal individuals with
[26, 28], supporting the theory that Wallerian high risk for the development of AD (i.e.,
degeneration may account for WM involvement Apolipoprotein E4 genotype and/or positive fam-
in this disease. ily history). Preclinical subjects with familial
The clinical relevance of DTI alterations in autosomal dominantly inherited AD also showed
AD patients is reflected by the association decreased WM integrity of the fornix and orbito-
between decreased global cognition, as measured frontal lobe [73]. Taking together, these studies
using the Mini-Mental State Examination suggest that microstructural WM changes may
(MMSE), and reduced FA [29–41] or increased serve as a potential imaging marker of early
MD [30, 32, 33] values in at least one brain AD-related brain damage.
region. Significant correlations between altered DTI tractography can improve the sensitivity
DTI metrics of temporo-parietal WM structures and specificity of diffusion measurements by
and neuropsychological measures of memory localizing WM changes in specific neuronal
performance have also been reported by several pathways of MCI/AD patients. Diffusivity
studies [42–46]. In addition, a number of studies abnormalities have been shown in the splenium
17 DTI in Dementing Conditions 347

Fig. 17.2 Diffusion tensor imaging (DTI) abnormalities respectively. The results for mean, axial, and radial diffu-
in patients with Alzheimer’s disease (AD) compared with sivity are shown at p < 0.05 corrected for multiple com-
healthy controls. Tract-based spatial statistics color maps parisons (family-wise error). Results from FA are shown
showing voxel-wise differences between patients and at an uncorrected statistical threshold (p < 0.05) [Reprinted
healthy controls are overlaid on a mean fractional anisot- from Agosta F, Pievani M, et al. “White matter damage in
ropy (FA) skeleton. Voxels of increased mean diffusivity Alzheimer disease and its relationship to gray matter atro-
and decreased FA are shown in red; voxels of increased phy.” Radiology 2011;258(3): 853–63. With permission
axial and radial diffusivity are shown in yellow and blue, from The Radiological Society of North America.]

of the corpus callosum, posterior cingulum and assess abnormalities of structural brain connec-
uncinate fasciculi [74–78] of AD patients. Only tivity. ACM can be easily obtained by initiating
a few tractography studies have been carried out diffusion tractography streamlines from all
in amnestic MCI patients [79–82], and diffusion parenchymal voxels, and then counting the
abnormalities of the posterior cingulum were number of streamlines passing through each
found in these patients relatively to controls, but voxel of the brain. An exploratory study employ-
not to early AD patients [79]. In amnestic MCI, ing such an approach in patients with AD and
WM damage was found not only in the limbic amnestic MCI revealed that ACM provides
pathways (i.e., fornix [82] and cingulum [81]), information that is complementary to that
but also in the major cortico-cortical WM tracts offered by FA with increased sensitivity [83].
subserving association cortices (i.e., the unci- Additionally, an unexpected increase of ACM
nate fasciculus, the inferior fronto-occipital has been found in the putamen of AD (the only
fasciculus, the inferior longitudinal fasciculus, group under treatment with cholinesterase
the superior longitudinal fasciculus and the cor- inhibitors) but not of MCI patients [83]. An
pus callosum) [80]. Recently, a novel method to intriguing explanation is that such an increase of
analyze DTI data, called anatomical connectiv- ACM in AD patients might reflect brain plastic-
ity mapping (ACM), has been proposed to ity driven by cholinesterase inhibitors.
348 M. Filippi et al.

Diagnostic Accuracy of DTI in AD anisotropy (FA) decline predominantly in the


and MCI Patients anterior corpus callosum after 13–16 months
from a baseline scan [84]. More recently, longitu-
For DTI to be useful in the clinical setting, one dinal changes of WM microstructural alterations
must be able to make inferences at the level of the in AD patients with and without treatment with
individual rather than the group. Adding DTI galantamine, a cholinesterase inhibitor, were
measurements of the posterior cingulum to hip- evaluated over a 12-month follow up [85].
pocampal volume significantly improves the Galantamine slowed the FA decrease of the pos-
accuracy in separating AD and amnestic MCI terior body of the corpus callosum over a 6-month
subjects from healthy controls (from 63 to 74 % period compared to placebo, but this effect was
in amnestic MCI, and from 78 to 91 % in AD) not seen anymore after a 6-month open-label
[55]. Diffusivity abnormalities of the left poste- treatment of all AD patients [85]. A large-scale
rior cingulum were able to distinguish subjects application of DTI in multicentre studies may
with amnestic MCI from those with non-amnestic help in the identification of markers that are
MCI with an accuracy of 85 % [56]. In addition, likely to provide sensitive outcome measures in
hippocampal diffusivity measurements were clinical trials of patients with AD and MCI.
found to be more sensitive than hippocampal vol-
ume in predicting conversion to AD in patients
with amnestic MCI [33, 52, 53]. The severity of Novel Strategies of Analysis
microstructural damage beyond the medial tem-
poral lobe was also associated with an increased A correlation between the site of amyloid-β
short-term risk to develop AD in amnestic MCI deposition in AD patients and the location of
patients [58]. A reduction of FA of the fornix has major brain hubs as defined by graph theoretical
been recently proposed as an imaging sign that analysis of functional connectivity in healthy
may be helpful in order to differentiate AD adults has been demonstrated [86]. These regions
patients from normal controls with an accuracy include the posterior cingulate cortex/precuneus,
of 75 %, as well as to predict conversion from the inferior parietal lobule, and the medial frontal
normal cognitive status to amnestic MCI and cortex, implying that the hubs are preferentially
from amnestic MCI to AD (with an accuracy of affected in the progression of AD. Using struc-
96 % and 92 %, respectively) [41]. An individual tural and DTI, abnormal topological properties
classification of MCI cases using support vector were described also in the structural brain net-
machine analysis of DTI data allowed for an indi- works of patients with AD. In a study that used
vidual classification with an accuracy up to 91 % between-subject covariation in regional measures
(healthy controls vs. MCI) and 98 % (stable vs. of cortical thickness to infer anatomical networks
progressive MCI at 1 year) [57]. Furthermore, from a large structural MRI data set, global clus-
such an approach resulted in a highly accurate tering and path length were increased in patients
individual classification of stable vs. progressive with AD relative to controls [87]. This study also
MCI at 1 year, regardless of the MCI subtype, found a decreased centrality of the classical hubs,
indicating that it might become a tool for early such as the temporal and parietal heteromodal
detection of MCI subjects evolving to overt cortices, and an increased centrality of unimodal
dementia [57]. association cortex, such as the lingual gyrus and
lateral occipitotemporal gyrus, as well as paralim-
bic regions [87]. The global clustering coefficient
Longitudinal DTI Studies and path length of MCI structural networks were
found to be intermediate between the AD group
Longitudinal studies of fiber tract injury in neu- and normal elderly people [88]. In addition, com-
rodegenerative diseases are still scanty. Amnestic pared with controls, AD and MCI patients
MCI subjects experience a significant fractional retained their hub regions in the frontal lobe but
17 DTI in Dementing Conditions 349

showed a loss of such regions in the temporal and motor speech abilities. Clinicopathological
lobe [88]. Similar to functional MRI data, this series suggest that the majority of nonfluent
study also revealed increased short-range inter- patients exhibit FTLD-tau or, less frequently,
regional correlations and disrupted long distance FTLD-TDP pathology, while most semantic
interregional correlations in MCI and AD [88]. cases have a TDP-43 proteinopathy [90]. AD is
An abnormal topological organization of large- the most likely underlying pathology of the logo-
scale WM networks was found in AD patients penic variant [92].
using DTI, with increased path length and
decreased global efficiency compared with con-
trols [89]. More importantly, WM connectivity DTI vs. Pathology and Genetic
patterns were associated with cognitive deficits Background in FTLD
[89], implying that a disturbed communication
between different brain regions is likely to be In a single, pathology-proven bvFTD case, DTI
important in the cognitive decline typical of this detected decreased FA values in WM frontal
condition. regions, where histopathology revealed a typical
frontal lobe degeneration of non-AD type [93].
Neuropathological [94, 95] and structural MRI
Frontotemporal Lobar [96] observations suggested that FTLD-tau may
Degeneration have a more severe involvement of WM than
FTLD-TDP cases. In view of this, DTI metrics
Frontotemporal lobar degeneration (FTLD) rep- were recently evaluated in autopsy- or genetic-
resents the second most common early-onset proven FTLD patients and showed a 96 % sensi-
neurodegenerative dementia after AD. FTLD is a tivity and 100 % specificity in distinguishing
clinically and pathologically heterogeneous between the two pathological variants [97]. This
spectrum of disorders, which encompasses dis- could prove to be clinically relevant, as potential
tinct clinical syndromes: the behavioral variant of disease-modifying treatments emerge that target
frontotemporal dementia (bvFTD) [10], and the tau or TDP-43.
language variant [9]. BvFTD presents with Microtubule-associated protein tau (MAPT),
marked changes in personality and behavior [10], progranulin (GRN), and C9orf72 mutations are
and, pathologically, is associated with all the the major genetic causes of autosomal dominant
three major FTLD pathologies, characterized by FTLD [98, 99]. Presymptomatic individuals with
abnormal cellular inclusions containing either these mutations represent the ideal population to
tau, TAR DNA-binding protein 43 (TDP-43), or assess the initial alterations of FTLD. A DTI
fused-in-sarcoma protein [90]. In the language study performed in a small sample of subjects
variant, known as primary progressive aphasia with GRN mutation revealed decreased FA of the
(PPA), a prominent, isolated language deficit is left uncinate and left inferior occipitofrontal fas-
the dominant feature during the initial phase of ciculi, with respect to non-carrier controls [100].
the disease [91]. Distinct profiles of language More recently, a larger sample of MAPT or GRN
impairment define the three clinical phenotypes mutation carriers was found to have distributed
of PPA [9]: the nonfluent/agrammatic (for conve- pattern of reduced FA in frontotemporal WM
nience hereafter called nonfluent), characterized tracts, in comparison with non-carriers [101].
by agrammatism in language production and
effortful speech with motor speech deficits; the
semantic, characterized by progressive loss of DTI in bvFTD Patients
knowledge about words and objects in the con-
text of relatively preserved fluency of speech; and Using a regions of interest (ROI)-based approach, a
the logopenic, characterized by impaired naming study of bvFTD patients found DTI abnormalities
and repetition in the context of spared syntactic of WM tracts passing through the frontal—anterior
350 M. Filippi et al.

cingulum, genu of the corpus callosum, anterior ciculi) were relatively spared [102, 109, 111].
SLF—and temporal lobes—uncinate fasciculus Studies using a voxel-wise approach showed that
and inferior longitudinal fasciculus (ILF) [102]. nonfluent patients have not only a damage to the
Voxel-based DTI studies in bvFTD patients fronto-parieto-temporal connections but also an
found FA reduction in frontoparietal regions, involvement of the corpus callosum, cingulum
which are likely to correspond to the SLF [103, bundle, external capsule, several regions of the
104], and in frontal and temporal WM regions, prefrontal and orbitofrontal and parietotemporal
including the anterior corpus callosum, anterior WM, mainly in the left hemisphere [104, 112,
cingulum, and uncinate, bilaterally (Fig. 17.3) 113]. A recent tract-based spatial statistics
[75, 104]. Such findings were confirmed by a (TBSS) study found a damage to portions of the
DTI tractography study, which disclosed a sig- ILF in nonfluent patients [112].
nificant FA decrease in all major association WM In the semantic variant, abnormalities of WM
tracts (ILF, uncinate, and SLF) and genu of the tract diffusivity were identified in the major
corpus callosum, as well as a sparing of cortico- inferior and superior temporal connections of
spinal tracts and splenium of the corpus callosum the left hemisphere, thus mirroring the severe
[105]. The majority of studies also detected dif- atrophy affecting the same regions: the ILF,
fusivity abnormalities in posterior WM regions, inferior-fronto-occipital fasciculus, and the
such as the posterior SLF and the posterior cingu- uncinate fasciculus within the ventral stream,
lum [75, 102, 104]. Although bvFTD is typically and the arcuate and the temporo-parietal com-
associated with frontal and anterior temporal ponent of the SLF within the dorsal stream [102,
atrophy [106], patterns of atrophy are known to 104, 109, 112, 114, 115] (Fig. 17.4). Although
be heterogeneous in this condition [107]. In addi- tractography studies have suggested that the
tion, even in the classic frontotemporal cases, the fronto-parietal connections are relatively spared
lateral and medial parietal lobes usually become in the semantic variant [109, 114], a finding
affected later in the disease course [108]. It is which couples with the absence of syntactic
therefore tempting to speculate that DTI metrics deficits in these patients, a few TBSS reports
may be viewed as early markers of WM injury in have shown an involvement of the left prefrontal
bvFTD patients, which may result at a later stage and parieto-frontal WM contributing to the SLF
in detectable volumetric abnormalities. and corona radiata [104, 112].
DTI measures of the anterior corpus callo-
sum and left SLF differentiated bvFTD from
DTI in PPA Patients nonfluent cases, while the best predictors of
semantic PPA compared with both bvFTD and
To date, only a few studies to date have investi- nonfluent cases were diffusivity abnormalities
gated the patterns of WM abnormalities in small of the left uncinate and inferior longitudinal fas-
samples of PPA patients using DTI. Most of ciculus [104].
these studies were performed in patients with Two studies so far applied DTI to patients
the two major variants (i.e., nonfluent and with the logopenic variant of PPA [104, 109],
semantic). showing the most consistent abnormalities in the
Using ROI-based or tractography DTI to left SLF temporoparietal component; abnormali-
investigate the WM language tracts of these ties were also detected in the left arcuate fascicu-
patients, it has been showed that the nonfluent lus, other components of the left SLF and right
variant is characterized by an involvement of all temporoparietal SLF (Fig. 17.4). These findings
the left SLF components [102, 109–111] (Fig. suggest that the logopenic variant is associated
17.4). In contrast, the ventral tracts connecting with damage to the WM tracts connecting regions
the temporal lobe with the occipital and the orbi- important for sentence repetition and phonologi-
tofrontal cortices (i.e., the ILF and uncinate fas- cal short-term memory [116].
17 DTI in Dementing Conditions 351

Fig. 17.3 Rendered


displays of diffusion tensor
imaging (DTI)
abnormalities in patients
with the behavioral variant
of frontotemporal dementia
(bvFTD) or Alzheimer’s
disease (AD) compared
with cognitively normal
subjects, as well as direct
comparisons between
bvFTD and AD: (a)
reduced fractional
anisotropy (warm colors);
(b) increased radial
diffusivity (cool colors);
(c) increased axial
diffusivity (cool colors)
[Reprinted from Zhang, Y.,
N. Schuff, et al. “White
matter damage in
frontotemporal dementia
and Alzheimer’s disease
measured by diffusion
MRI.” Brain 2009;132(Pt
9): 2579–92. With
permission from Oxford
University Press.]

Differential Diagnosis: FTLD vs. AD injury is more prominent in bvFTD than in


AD. One recent study combined cortical thick-
DTI is increasingly being used to improve the ness measurement and DTI to compare FTLD
diagnostic differentiation between FTLD and (both bvFTD and PPA) and AD patients, with
AD. When compared with AD, bvFTD is associ- autopsy- or cerebrospinal fluid-confirmed disease
ated with greater reductions of FA in frontal [117]. The direct comparison of the two groups
regions (Fig. 17.3) [75], which suggests that WM showed a significantly greater atrophy in inferior
352 M. Filippi et al.

Fig. 17.4 Mean diffusivity (MD) values of healthy con- denote significant difference relative to normal controls at
trols, nonfluent, semantic, and logopenic variants of primary p < 0.05. The chromatic scale represents average MD values
progressive aphasia are shown on the probability maps for ranging from lower (violet-blue) to higher values (yellow-
left superior longitudinal fasciculus (SLF), inferior longitu- red). MD is measured in mm2/s × 10−3 [Reprinted from
dinal fasciculus (ILF), uncinate fasciculus (UNC), overlaid Galantucci, S., M. C. Tartaglia, et al. White matter damage
on a standard Montreal Neurological Institute brain. Only in primary progressive aphasias: a diffusion tensor tractog-
voxels that are in common in at least 20 % of the subjects in raphy study. Brain 2011;134(Pt 10): 3011–29. With permis-
each group were included in the probability maps. Asterisks sion from Oxford University Press.]

frontal, medial frontal and prefrontal cortical patterns of cortical atrophy between FTLD and
regions, and a reduced FA of the genu of the cor- AD patients, identifying a significant area of
pus callosum, left inferior fronto-occipital fascic- reduced FA in the anterior corpus callosum of
ulus, cingulum, uncinate and bilateral corona FTLD patients, as well as a significantly more
radiata in FTLD cases relative to AD [117]. severe atrophy of the precuneus and posterior cin-
Conversely, AD patients showed no areas of sig- gulate cortex in AD patients [118]. A combina-
nificant reduction in cortical thickness or WM tion of WM and cortical data provided a highly
integrity relative to FTLD patients [117]. Another accurate classification of these two conditions,
recent study compared DT MRI alterations and with 87 % sensitivity and 83 % specificity [118].
17 DTI in Dementing Conditions 353

Dementia with Lewy Bodies fusivity and a reduced cortical volume in the
medial temporal lobe, posterior cingulate cor-
Dementia with Lewy bodies (DLB) is the second tex, precuneus, and temporoparietal association
most common form of neurodegenerative demen- cortex compared to both healthy controls and
tia in elderly subjects [8]. Memory impairment is patients with DLB [124]. The addition of diffu-
generally less severe in patients with DLB than in sivity values of the hippocampus and parahip-
those with AD, whereas deficits on tests of atten- pocampal gyrus to those of cortical volumes
tion, executive function, and visuospatial ability improves further the ability to distinguish AD
can be prominent. In pathological studies, a dis- patients from those with DLB [124].
tributed pattern of Lewy bodies has been observed A reduction of FA in the uncinate fasciculus
in the neocortex, limbic structures, subcortical has been shown both in AD and DLB patients
nuclei and brainstem of these patients [119, 120]. [76, 126], with additional more posterior dam-
age in patients with DLB [76]. A recent study
[127] showed an involvement of the parieto-
DTI in DLB Patients occipital and temporal WM tracts both in DLB
and AD, but a much greater ratio of posterior-to-
Only a few studies investigated diffusivity altera- anterior suprathreshold voxels was found in
tions in DLB patients [76, 121–127]. The com- DLB—6.5—when compared to AD—1.1 (Fig.
parison between DLB patients and controls using 17.5) [127]. Similarly, the few other studies
DTI resulted in controversial findings, ranging investigating WM damage in DLB showed a
from isolated damage to the ILF [123, 124], unci- more posterior pattern of abnormalities in com-
nate fasciculus [126] or parietal lobe [122] to a parison to AD [122, 123], with a relative sparing
distributed pattern of FA reduction [121, 125]. of the structures of the temporal and frontal
The most consistent finding among studies was a lobes, reflecting the structural preservation of
decreased FA in the ILF [121, 123, 124], as well these areas in DLB [128].
as reduced FA or increased MD in tracts of the
parieto-occipital lobes [121, 122, 124, 125, 127],
particularly the posterior cingulum and precu- Conclusions
neus (Fig. 17.5a). Damage to these regions,
which are important for visual information pro- DTI is a sensitive tool to detect WM abnormali-
cessing, may reflect the prevalent impairment of ties in subjects with neurodegenerative dement-
visuospatial abilities, as well as the presence of ing conditions. Diffusivity seems to be impaired
visual hallucinations in these patients. One study in several brain regions early in the disease pro-
[124] reported significantly higher diffusivity of cess; then, WM damage increases with disease
the ILF from patients with DLB experiencing severity. Since new disease-modifying therapies
visual hallucinations, in comparison with DLB in AD and other neurodegenerative dementia will
patients who did not. likely be most beneficial before substantial neu-
ronal loss and clinical impairment have occurred,
DTI holds promise as valuable tool for selecting
Differential Diagnosis: DLB vs. AD candidates for clinical trials and as predictive
markers of dementia progression in defined risk
Diffusivity abnormalities in the cortex of groups of patients. The reliability and reproduc-
patients with AD are more distributed and ibility of DTI in a large-scale, multicenter setting
severe than in those with DLB [122, 124]. in patients with these diseases warrant further
Patients with AD experience an increased dif- investigation.
354 M. Filippi et al.

Fig. 17.5 Fractional anisotropy (FA) reduction in patients included the optic radiation (p < 0.05, corrected for multi-
with dementia with Lewy bodies (DLB) and Alzheimer’s ple comparisons). (b) AD vs. controls: the pattern of
disease (AD). Tract-based spatial statistics color maps abnormalities was more distributed than in DLB and
showing voxel-wise differences in FA (blue) between included clusters in the temporal, parieto-occipital, and
these groups are overlaid on a mean FA skeleton (green). frontal lobes (p < 0.05, corrected) [Reprinted from Watson,
(a) DLB vs. controls: abnormalities were identified R., A. M. Blamire, et al. “Characterizing dementia with
mainly in the parieto-occipital areas (precuneus and cin- Lewy bodies by means of diffusion tensor imaging.”
gulate gyrus). Reduced FA in the temporal lobes was Neurology 2012;79(9): 906–14. With permission from
detected in a region of the posterior thalamic radiation that Wolters Kluwer Health.]

Report of the Quality Standards Subcommittee of the


References American Academy of Neurology. Neurology.
2001;56(9):1143–53.
1. Filippi M, Agosta F, et al. EFNS task force: the use 5. Barkhof F, Fox NC, et al. Neuroimaging in demen-
of neuroimaging in the diagnosis of dementia. Eur tia. New York, NY: Springer; 2011.
J Neurol. 2012;19(12):e131–40. 1487-501. 6. Dubois B, Feldman HH, et al. Revising the definition
2. Sorbi S, Hort J, et al. EFNS-ENS Guidelines on the of Alzheimer’s disease: a new lexicon. Lancet
diagnosis and management of disorders associated Neurol. 2010;9(11):1118–27.
with dementia. Eur J Neurol. 2012;19(9): 7. McKhann GM, Knopman DS, et al. The diagnosis of
1159–79. dementia due to Alzheimer’s disease:
3. (NICE), N. I. f. H. a. C. E. (March 2011). CG42 recommendations from the National Institute on
Dementia: NICE guideline. Aging-Alzheimer’s Association workgroups on
4. Knopman DS, DeKosky ST, et al. Practice parameter: diagnostic guidelines for Alzheimer’s disease.
diagnosis of dementia (an evidence-based review). Alzheimers Dement. 2011;7(3):263–9.
17 DTI in Dementing Conditions 355

8. McKeith IG, Dickson DW, et al. Diagnosis and man- Creutzfeldt-Jakob disease. J Neurol. 2013;260(2):
agement of dementia with Lewy bodies: third report 498–506.
of the DLB Consortium. Neurology. 2005;65(12): 24. Jack Jr CR, Lowe VJ, et al. Serial PIB and MRI in
1863–72. normal, mild cognitive impairment and Alzheimer’s
9. Gorno-Tempini ML, Hillis AE, et al. Classification disease: implications for sequence of pathological
of primary progressive aphasia and its variants. events in Alzheimer’s disease. Brain. 2009;132(Pt
Neurology. 2011;76(11):1006–14. 5):1355–65.
10. Rascovsky K, Hodges JR, et al. Sensitivity of revised 25. Bateman RJ, Xiong C, et al. Clinical and biomarker
diagnostic criteria for the behavioural variant of changes in dominantly inherited Alzheimer’s dis-
frontotemporal dementia. Brain. 2011;134(Pt 9): ease. N Engl J Med. 2012;367(9):795–804.
2456–77. 26. Sexton CE, Kalu UG, et al. A meta-analysis of diffu-
11. Teipel SJ, Wegrzyn M, et al. Anatomical MRI and sion tensor imaging in mild cognitive impairment
DTI in the diagnosis of Alzheimer’s disease: a and Alzheimer’s disease. Neurobiol Aging. 2011;
European multicenter study. J Alzheimers Dis. 32(12):2322.e5–18.
2012;31 Suppl 3:S33–47. 27. Clerx L, Visser PJ, et al. New MRI markers for
12. Geschwind MD, Shu H, et al. Rapidly progressive Alzheimer’s disease: a meta-analysis of diffusion
dementia. Ann Neurol. 2008;64(1):97–108. tensor imaging and a comparison with medial tem-
13. Prusiner SB. Shattuck lecture--neurodegenerative poral lobe measurements. J Alzheimers Dis. 2012;
diseases and prions. N Engl J Med. 2001;344(20): 29(2):405–29.
1516–26. 28. Agosta F, Pievani M, et al. White matter damage in
14. Young GS, Geschwind MD, et al. Diffusion- Alzheimer disease and its relationship to gray matter
weighted and fluid-attenuated inversion recovery atrophy. Radiology. 2011;258(3):853–63.
imaging in Creutzfeldt-Jakob disease: high sensitiv- 29. Rose SE, Chen F, et al. Loss of connectivity in
ity and specificity for diagnosis. AJNR Am Alzheimer’s disease: an evaluation of white matter
J Neuroradiol. 2005;26(6):1551–62. tract integrity with colour coded MR diffusion tensor
15. Vitali P, Maccagnano E, et al. Diffusion-weighted imaging. J Neurol Neurosurg Psychiatry. 2000;69(4):
MRI hyperintensity patterns differentiate CJD from 528–30.
other rapid dementias. Neurology. 2011;76(20): 30. Bozzali M, Falini A, et al. White matter damage in
1711–9. Alzheimer’s disease assessed in vivo using diffusion
16. Geschwind MD, Potter CA, et al. Correlating DWI tensor magnetic resonance imaging. J Neurol
MRI with pathologic and other features of Jakob- Neurosurg Psychiatry. 2002;72(6):742–6.
Creutzfeldt disease. Alzheimer Dis Assoc Disord. 31. Xie S, Xiao JX, et al. Evaluation of bilateral cingu-
2009;23(1):82–7. lum with tractography in patients with Alzheimer’s
17. Murata T, Shiga Y, et al. Conspicuity and evolution disease. Neuroreport. 2005;16(12):1275–8.
of lesions in Creutzfeldt-Jakob disease at diffusion- 32. Duan JH, Wang HQ, et al. White matter damage of
weighted imaging. AJNR Am J Neuroradiol. patients with Alzheimer’s disease correlated with the
2002;23(7):1164–72. decreased cognitive function. Surg Radiol Anat.
18. Shiga Y, Miyazawa K, et al. Diffusion-weighted 2006;28(2):150–6.
MRI abnormalities as an early diagnostic marker for 33. Muller MJ, Greverus D, et al. Diagnostic utility of
Creutzfeldt-Jakob disease. Neurology. 2004;63(3): hippocampal size and mean diffusivity in amnestic
443–9. MCI. Neurobiol Aging. 2007;28(3):398–403.
19. Zerr I, Kallenberg K, et al. Updated clinical diagnos- 34. Ukmar M, Makuc E, et al. Evaluation of white mat-
tic criteria for sporadic Creutzfeldt-Jakob disease. ter damage in patients with Alzheimer’s disease and
Brain. 2009;132(Pt 10):2659–68. in patients with mild cognitive impairment by using
20. Collie DA, Summers DM, et al. Diagnosing variant diffusion tensor imaging. Radiol Med. 2008;113(6):
Creutzfeldt-Jakob disease with the pulvinar sign: 915–22.
MR imaging findings in 86 neuropathologically con- 35. Bai F, Zhang Z, et al. Abnormal integrity of associa-
firmed cases. AJNR Am J Neuroradiol. 2003;24(8): tion fiber tracts in amnestic mild cognitive impair-
1560–9. ment. J Neurol Sci. 2009;278(1-2):102–6.
21. Yi SH, Park KC, et al. Relationship between clinical 36. Mielke MM, Kozauer NA, et al. Regionally-specific
course and Diffusion-weighted MRI findings in spo- diffusion tensor imaging in mild cognitive impair-
radic Creutzfeldt-Jakob Disease. Neurol Sci. ment and Alzheimer’s disease. Neuroimage.
2008;29(4):251–5. 2009;46(1):47–55.
22. Lee H, Cohen OS, et al. Cerebral white matter dis- 37. Catheline G, Periot O, et al. Distinctive alterations
ruption in Creutzfeldt-Jakob disease. AJNR Am of the cingulum bundle during aging and
J Neuroradiol. 2012;33(10):1945–50. Alzheimer’s disease. Neurobiol Aging. 2010;31(9):
23. Wang LH, Bucelli RC, et al. Role of magnetic reso- 1582–92.
nance imaging, cerebrospinal fluid, and electroen- 38. Morikawa M, Kiuchi K, et al. Uncinate fasciculus-
cephalogram in diagnosis of sporadic correlated cognition in Alzheimer’s disease: a diffusion
356 M. Filippi et al.

tensor imaging study by tractography. 52. Kantarci K, Petersen RC, et al. DWI predicts future
Psychogeriatrics. 2010;10(1):15–20. progression to Alzheimer disease in amnestic mild
39. Zarei M, Patenaude B, et al. Combining shape and cognitive impairment. Neurology. 2005;64(5):
connectivity analysis: an MRI study of thalamic 902–4.
degeneration in Alzheimer’s disease. Neuroimage. 53. Fellgiebel A, Dellani PR, et al. Predicting conver-
2010;49(1):1–8. sion to dementia in mild cognitive impairment by
40. Zimny A, Szewczyk P, et al. Multimodal imaging in volumetric and diffusivity measurements of the hip-
diagnosis of Alzheimer’s disease and amnestic mild pocampus. Psychiatry Res. 2006;146(3):283–7.
cognitive impairment: value of magnetic resonance 54. Stahl R, Dietrich O, et al. White matter damage in
spectroscopy, perfusion, and diffusion tensor imag- Alzheimer disease and mild cognitive impairment:
ing of the posterior cingulate region. J Alzheimers assessment with diffusion-tensor MR imaging and
Dis. 2011;27(3):591–601. parallel imaging techniques. Radiology. 2007;
41. Oishi K, Mielke MM, et al. The fornix sign: a poten- 243(2):483–92.
tial sign for Alzheimer’s disease based on diffusion 55. Zhang Y, Schuff N, et al. Diffusion tensor imaging of
tensor imaging. J Neuroimaging. 2012;22(4):365–74. cingulum fibers in mild cognitive impairment and
42. Fellgiebel A, Muller MJ, et al. Color-coded Alzheimer disease. Neurology. 2007;68(1):13–9.
diffusion-tensor-imaging of posterior cingulate fiber 56. Chua TC, Wen W, et al. Diffusion tensor imaging of
tracts in mild cognitive impairment. Neurobiol the posterior cingulate is a useful biomarker of mild
Aging. 2005;26(8):1193–8. cognitive impairment. Am J Geriatr Psychiatry.
43. Rose SE, McMahon KL, et al. Diffusion indices on 2009;17(7):602–13.
magnetic resonance imaging and neuropsychologi- 57. Haller S, Nguyen D, et al. Individual prediction of
cal performance in amnestic mild cognitive impair- cognitive decline in mild cognitive impairment using
ment. J Neurol Neurosurg Psychiatry. 2006;77(10): support vector machine-based analysis of diffusion
1122–8. tensor imaging data. J Alzheimers Dis. 2010;
44. Fellgiebel A, Schermuly I, et al. Functional relevant 22(1):315–27.
loss of long association fibre tracts integrity in early 58. Scola E, Bozzali M, et al. A diffusion tensor MRI
Alzheimer’s disease. Neuropsychologia. 2008;46(6): study of patients with MCI and AD with a 2-year
1698–706. clinical follow-up. J Neurol Neurosurg Psychiatry.
45. Walhovd KB, Fjell AM, et al. Multimodal imaging 2010;81(7):798–805.
in mild cognitive impairment: metabolism, mor- 59. Shu N, Wang Z, et al. Multiple diffusion indices
phometry and diffusion of the temporal-parietal reveals white matter degeneration in Alzheimer’s
memory network. Neuroimage. 2009;45(1):215–23. disease and mild cognitive impairment: a tract-based
46. Bosch B, Arenaza-Urquijo EM, et al. Multiple DTI spatial statistics study. J Alzheimers Dis. 2011;26
index analysis in normal aging, amnestic MCI and Suppl 3:275–85.
AD. Relationship with neuropsychological perfor- 60. Alves GS, O’Dwyer L, et al. Different patterns of
mance. Neurobiol Aging. 2012;33(1):61–74. white matter degeneration using multiple diffusion
47. Huang J, Friedland RP, et al. Diffusion tensor imag- indices and volumetric data in mild cognitive impair-
ing of normal-appearing white matter in mild cogni- ment and Alzheimer patients. PLoS One. 2012;
tive impairment and early Alzheimer disease: 7(12):e52859.
preliminary evidence of axonal degeneration in the 61. Nowrangi MA, Lyketsos CG, et al. Longitudinal,
temporal lobe. AJNR Am J Neuroradiol. 2007; region-specific course of diffusion tensor imaging
28(10):1943–8. measures in mild cognitive impairment and
48. Chen TF, Chen YF, et al. Executive dysfunction and Alzheimer’s disease. Alzheimers Dement. 2013;9(5):
periventricular diffusion tensor changes in amnesic 519–28.
mild cognitive impairment and early Alzheimer’s 62. van Bruggen T, Stieltjes B, et al. Do Alzheimer-
disease. Hum Brain Mapp. 2009;30(11):3826–36. specific microstructural changes in mild cognitive
49. Sjobeck M, Elfgren C, et al. Alzheimer’s disease impairment predict conversion? Psychiatry Res.
(AD) and executive dysfunction. A case-control 2012;203(2-3):184–93.
study on the significance of frontal white matter 63. Amlien IK, Fjell AM, et al. Mild cognitive impair-
changes detected by diffusion tensor imaging (DTI). ment: cerebrospinal fluid tau biomarker pathologic
Arch Gerontol Geriatr. 2010;50(3):260–6. levels and longitudinal changes in white matter
50. Ota M, Sato N, et al. Relationship between apathy integrity. Radiology. 2013;266(1):295–303.
and diffusion tensor imaging metrics of the brain in 64. Zhuang L, Sachdev PS, et al. Microstructural white
Alzheimer’s disease. Int J Geriatr Psychiatry. matter changes in cognitively normal individuals at
2011;27(7):722–6. risk of amnestic MCI. Neurology. 2012;79(8):
51. Tighe SK, Oishi K, et al. Diffusion tensor imaging of 748–54.
neuropsychiatric symptoms in mild cognitive 65. Nierenberg J, Pomara N, et al. Abnormal white mat-
impairment and Alzheimer’s dementia. J ter integrity in healthy apolipoprotein E epsilon4
Neuropsychiatry Clin Neurosci. 2012;24(4):484–8. carriers. Neuroreport. 2005;16(12):1369–72.
17 DTI in Dementing Conditions 357

66. Persson J, Lind J, et al. Altered brain white matter 82. Zhuang L, Wen W, et al. Abnormalities of the fornix
integrity in healthy carriers of the APOE epsilon4 in mild cognitive impairment are related to episodic
allele: a risk for AD? Neurology. 2006;66(7):1029–33. memory loss. J Alzheimers Dis. 2012;29(3):
67. Honea RA, Vidoni E, et al. Impact of APOE on the 629–39.
healthy aging brain: a voxel-based MRI and DTI 83. Bozzali M, Parker GJ, et al. Anatomical connectivity
study. J Alzheimers Dis. 2009;18(3):553–64. mapping: a new tool to assess brain disconnection in
68. Bendlin BB, Ries ML, et al. White matter is altered Alzheimer’s disease. Neuroimage. 2011;
with parental family history of Alzheimer’s disease. 54(3):2045–51.
Alzheimers Dement. 2010;6(5):394–403. 84. Teipel SJ, Meindl T, et al. Longitudinal changes in
69. Gold BT, Powell DK, et al. Alterations in multiple fiber tract integrity in healthy aging and mild cogni-
measures of white matter integrity in normal women tive impairment: a DTI follow-up study. J Alzheimers
at high risk for Alzheimer’s disease. Neuroimage. Dis. 2011;22(2):507–22.
2010;52(4):1487–94. 85. Likitjaroen Y, Meindl T, et al. Longitudinal changes
70. Smith CD, Chebrolu H, et al. White matter diffusion of fractional anisotropy in Alzheimer’s disease
alterations in normal women at risk of Alzheimer’s patients treated with galantamine: a 12-month ran-
disease. Neurobiol Aging. 2010;31(7):1122–31. domized, placebo-controlled, double-blinded study.
71. Heise V, Filippini N, et al. The APOE varepsilon4 Eur Arch Psychiatry Clin Neurosci. 2012;
allele modulates brain white matter integrity in 262(4):341–50.
healthy adults. Mol Psychiatry. 2011;16(9):908–16. 86. Buckner RL, Sepulcre J, et al. Cortical hubs revealed
72. Westlye LT, Reinvang I, et al. Effects of APOE on by intrinsic functional connectivity: mapping,
brain white matter microstructure in healthy adults. assessment of stability, and relation to Alzheimer’s
Neurology. 2012;79(19):1961–9. disease. J Neurosci. 2009;29(6):1860–73.
73. Ringman JM, O’Neill J, et al. Diffusion tensor imag- 87. He Y, Chen Z, et al. Structural insights into aberrant
ing in preclinical and presymptomatic carriers of topological patterns of large-scale cortical networks
familial Alzheimer’s disease mutations. Brain. in Alzheimer’s disease. J Neurosci. 2008;
2007;130(Pt 7):1767–76. 28(18):4756–66.
74. Taoka T, Iwasaki S, et al. Diffusion anisotropy and 88. Yao Z, Zhang Y, et al. Abnormal cortical networks in
diffusivity of white matter tracts within the temporal mild cognitive impairment and Alzheimer’s disease.
stem in Alzheimer disease: evaluation of the “tract of PLoS Comput Biol. 2010;6(11):e1001006.
interest” by diffusion tensor tractography. AJNR Am 89. Lo CY, Wang PN, et al. Diffusion tensor tractogra-
J Neuroradiol. 2006;27(5):1040–5. phy reveals abnormal topological organization in
75. Zhang Y, Schuff N, et al. White matter damage in structural cortical networks in Alzheimer’s disease.
frontotemporal dementia and Alzheimer’s disease J Neurosci. 2010;30(50):16876–85.
measured by diffusion MRI. Brain. 2009;132(Pt 90. Mackenzie IR, Neumann M, et al. Nomenclature and
9):2579–92. nosology for neuropathologic subtypes of fronto-
76. Kiuchi K, Morikawa M, et al. White matter changes temporal lobar degeneration: an update. Acta
in dementia with Lewy bodies and Alzheimer’s dis- Neuropathol. 2010;119(1):1–4.
ease: a tractography-based study. J Psychiatr Res. 91. Mesulam MM. Primary progressive aphasia. Ann
2011;45(8):1095–100. Neurol. 2001;49(4):425–32.
77. Fischer FU, Scheurich A, et al. Automated tractogra- 92. Mesulam M, Wicklund A, et al. Alzheimer and fron-
phy of the cingulate bundle in Alzheimer’s disease: a totemporal pathology in subsets of primary progres-
multicenter DTI study. J Magn Reson Imaging. sive aphasia. Ann Neurol. 2008;63(6):709–19.
2012;36(1):84–91. 93. Larsson EM, Englund E, et al. MRI with diffusion
78. Preti MG, Baglio F, et al. Assessing corpus callosum tensor imaging post-mortem at 3.0 T in a patient
changes in Alzheimer’s disease: comparison with frontotemporal dementia. Dement Geriatr Cogn
between tract-based spatial statistics and atlas-based Disord. 2004;17(4):316–9.
tractography. PLoS One. 2012;7(4):e35856. 94. Forman MS, Zhukareva V, et al. Signature tau neuro-
79. Kiuchi K, Morikawa M, et al. Abnormalities of the pathology in gray and white matter of corticobasal
uncinate fasciculus and posterior cingulate fascicu- degeneration. Am J Pathol. 2002;160(6):2045–53.
lus in mild cognitive impairment and early 95. Geser F, Martinez-Lage M, et al. Clinical and patho-
Alzheimer’s disease: a diffusion tensor tractography logical continuum of multisystem TDP-43 pro-
study. Brain Res. 2009;1287:184–91. teinopathies. Arch Neurol. 2009;66(2):180–9.
80. Pievani M, Agosta F, et al. Assessment of white mat- 96. Kim EJ, Rabinovici GD, et al. Patterns of MRI atro-
ter tract damage in mild cognitive impairment and phy in tau positive and ubiquitin positive frontotem-
Alzheimer’s disease. Hum Brain Mapp. 2010;31(12): poral lobar degeneration. J Neurol Neurosurg
1862–75. Psychiatry. 2007;78(12):1375–8.
81. Bozzali M, Giulietti G, et al. Damage to the cingu- 97. McMillan CT, Irwin DJ, et al. White matter imaging
lum contributes to Alzheimer’s disease pathophysi- helps dissociate tau from TDP-43 in frontotemporal
ology by deafferentation mechanism. Hum Brain lobar degeneration. J Neurol Neurosurg Psychiatry.
Mapp. 2012;33(6):1295–308. 2013;84:949.
358 M. Filippi et al.

98. Seelaar H, Kamphorst W, et al. Distinct genetic 114. Agosta F, Henry RG, et al. Language networks in
forms of frontotemporal dementia. Neurology. 2008; semantic dementia. Brain. 2010;133(Pt 1):286–99.
71(16):1220–6. 115. Acosta-Cabronero J, Patterson K, et al. Atrophy,
99. DeJesus-Hernandez M, Mackenzie IR, et al. Expanded hypometabolism and white matter abnormalities in
GGGGCC hexanucleotide repeat in noncoding region semantic dementia tell a coherent story. Brain.
of C9ORF72 causes chromosome 9p-linked FTD and 2011;134(Pt 7):2025–35.
ALS. Neuron. 2011;72(2):245–56. 116. Gorno-Tempini ML, Brambati SM, et al. The logo-
100. Borroni B, Alberici A, et al. Brain magnetic reso- penic/phonological variant of primary progressive
nance imaging structural changes in a pedigree of aphasia. Neurology. 2008;71(16):1227–34.
asymptomatic progranulin mutation carriers. 117. Avants BB, Cook PA, et al. Dementia induces cor-
Rejuvenation Res. 2008;11(3):585–95. related reductions in white matter integrity and corti-
101. Dopper EG, Rombouts SA, et al. Structural and cal thickness: a multivariate neuroimaging study
functional brain connectivity in presymptomatic with sparse canonical correlation analysis.
familial frontotemporal dementia. Neurology. 2013; Neuroimage. 2010;50(3):1004–16.
80(9):814–23. 118. McMillan CT, Brun C, et al. White matter imaging
102. Whitwell JL, Avula R, et al. Gray and white matter contributes to the multimodal diagnosis of fronto-
water diffusion in the syndromic variants of temporal lobar degeneration. Neurology. 2012;
frontotemporal dementia. Neurology. 2011;74(16): 78(22):1761–8.
1279–87. 119. McKeith IG, Galasko D, et al. Consensus guidelines
103. Borroni B, Brambati SM, et al. Evidence of white for the clinical and pathologic diagnosis of dementia
matter changes on diffusion tensor imaging in fron- with Lewy bodies (DLB): report of the consortium
totemporal dementia. Arch Neurol. 2007;64(2): on DLB international workshop. Neurology.
246–51. 1996;47(5):1113–24.
104. Agosta F, Scola E, et al. White matter damage in 120. Spillantini MG, Crowther RA, et al. alpha-Synu-
frontotemporal lobar degeneration spectrum. Cereb clein in filamentous inclusions of Lewy bodies from
Cortex. 2012;22(12):2705–14. Parkinson’s disease and dementia with Lewy bod-
105. Matsuo K, Mizuno T, et al. Cerebral white matter ies. Proc Natl Acad Sci U S A. 1998;95(11):
damage in frontotemporal dementia assessed by dif- 6469–73.
fusion tensor tractography. Neuroradiology. 121. Bozzali M, Falini A, et al. Brain tissue damage in
2008;50(7):605–11. dementia with Lewy bodies: an in vivo diffusion ten-
106. Rosen HJ, Gorno-Tempini ML, et al. Patterns of sor MRI study. Brain. 2005;128(Pt 7):1595–604.
brain atrophy in frontotemporal dementia and seman- 122. Firbank MJ, Blamire AM, et al. Diffusion tensor
tic dementia. Neurology. 2002;58(2):198–208. imaging in dementia with Lewy bodies and
107. Whitwell JL, Przybelski SA, et al. Distinct anatomi- Alzheimer’s disease. Psychiatry Res. 2007;155(2):
cal subtypes of the behavioural variant of frontotem- 135–45.
poral dementia: a cluster analysis study. Brain. 123. Ota M, Sato N, et al. Degeneration of dementia with
2009;132(Pt 11):2932–46. Lewy bodies measured by diffusion tensor imaging.
108. Brambati SM, Renda NC, et al. A tensor based mor- NMR Biomed. 2009;22(3):280–4.
phometry study of longitudinal gray matter contrac- 124. Kantarci K, Avula R, et al. Dementia with Lewy
tion in FTD. Neuroimage. 2007;35(3):998–1003. bodies and Alzheimer disease: neurodegenerative
109. Galantucci S, Tartaglia MC, et al. White matter dam- patterns characterized by DTI. Neurology.
age in primary progressive aphasias: a diffusion tensor 2010;74(22):1814–21.
tractography study. Brain. 2011;134(Pt 10):3011–29. 125. Lee JE, Park HJ, et al. A comparative analysis of
110. Grossman M, Powers J, et al. Disruption of large- cognitive profiles and white-matter alterations using
scale neural networks in non-fluent/agrammatic voxel-based diffusion tensor imaging between
variant primary progressive aphasia associated with patients with Parkinson’s disease dementia and
frontotemporal degeneration pathology. Brain Lang. dementia with Lewy bodies. J Neurol Neurosurg
2012;127:106–20. Psychiatry. 2010;81(3):320–6.
111. Zhang Y, Tartaglia MC, et al. MRI signatures of 126. Serra L, Cercignani M, et al. White matter damage
brain macrostructural atrophy and microstructural along the uncinate fasciculus contributes to cogni-
degradation in frontotemporal lobar degeneration tive decline in AD and DLB. Curr Alzheimer Res.
subtypes. J Alzheimers Dis. 2013;33(2):431–44. 2012;9(3):326–33.
112. Schwindt GC, Graham NL, et al. Whole-brain white 127. Watson R, Blamire AM, et al. Characterizing demen-
matter disruption in semantic and nonfluent variants tia with Lewy bodies by means of diffusion tensor
of primary progressive aphasia. Hum Brain Mapp. imaging. Neurology. 2012;79(9):906–14.
2013;34:973. 128. Whitwell JL, Weigand SD, et al. Focal atrophy in
113. Mahoney CJ, Malone IB, et al. White matter tract dementia with Lewy bodies on MRI: a distinct
signatures of the progressive aphasias. Neurobiol pattern from Alzheimer’s disease. Brain. 2007;
Aging. 2013;34(6):1687–99. 130(Pt 3):708–19.
DTI in Psychiatry
18
Josselin Houenou and Louise Emsell

neurobiological models of psychiatric illness,


Learning Points such as those based on “connectivity.” DTI
metrics can be used in combination with other
• DTI is not used routinely in clinical practice neuroimaging data as potential biomarkers
owing to special challenges inherent to defin- that may aid patient stratification and improve
ing psychopathology, the practical issues treatment.
associated with scanning patients and the lack • DTI studies in psychiatry face a number of
of sensitivity and specificity of DTI measures. issues. Specifically, the categorical classifica-
In the future, it could be used to inform inva- tion of mental disorders is subjective and defi-
sive neurosurgical treatments of psychiatric nitions are continually evolving. Clinical
illness, such as deep brain stimulation. samples are therefore highly heterogeneous
• DTI is increasingly used as clinical research with regard to clinical history, psychiatric and
tool in psychiatry. It can be used to inform medical comorbidity, active symptoms, and
medication. Alcohol misuse may represent a
significant confound in studies of psychiatric
populations.
• Scanning psychiatric patients presents some
practical challenges, including obtaining
J. Houenou, MD, PhD
INSERM U955 Team 15 “Translation Psychiatry”, informed consent, reduced compliance with
Fondation Fondamental, APHP, Hôpitaux procedures owing to anxiety or hyperactivity,
Universitaires Mondor, Créteil, France and increased movement within the scanner
NeuroSpin Neuroimaging Center, UNIACT Lab, compared to healthy subjects.
Psychiatry Team, CEA Saclay, Gif-Sur-Yvette, • DTI has been used to investigate a number of
France
psychiatric disorders, including, but not lim-
L. Emsell, PhD (*) ited to schizophrenia, mood, anxiety, person-
Translational MRI, Department of Imaging and
ality and neurodevelopmental disorders.
Pathology, KU Leuven, and Radiology, University
Hospitals, Leuven, Belgium Findings are largely nonspecific and suggest
varying degrees of white matter microstruc-
Universitair Psychiatrisch Centrum (UPC),
KU Leuven, Leuven, Belgium tural abnormality in cortical and subcortical
e-mail: louise.emsell@med.kuleuven.be cognitive and limbic networks.

© Springer Science+Business Media New York 2016 359


W. Van Hecke et al. (eds.), Diffusion Tensor Imaging, DOI 10.1007/978-1-4939-3118-7_18
360 J. Houenou and L. Emsell

Role of DTI in Psychiatry Strikingly, T1, T2, FLAIR, and fMRI studies
point to crucial abnormalities of white matter in
Clinical Research major psychiatric disorders. On T1 scans, total
white matter volume has been found to be reduced
Presently, DTI is not used routinely in clinical psy- in schizophrenia, whilst regional volumetric
chiatric practice. There are a number of reasons for reduction (e.g., corpus callosum) has also been
this, which broadly relate to both the complex reported in schizophrenia and in other conditions
nature of defining psychopathology and the practi- such as bipolar disorder [7]. In mood disorders,
cal challenges associated with scanning patients white matter hyperintensities observed on T2 and
with mental illness. These issues are discussed in FLAIR are the most commonly reported abnor-
more detail in the body of this chapter. Nevertheless, malities, especially in bipolar disorder and late-
DTI does have an increasingly significant role in life depression [1]. Altered functional connectivity
psychiatry, and that is in the field of clinical research. between brain areas, as measured by inter-
Early clinical neuroimaging studies in psychia- regional BOLD levels correlations, has been
try used computed tomography (CT) and subse- reported in schizophrenia, bipolar disorder, and
quently classical structural magnetic resonance anxiety disorders, both during the completion of
imaging (MRI) (using T1, T2, and fluid attenua- specific tasks and at rest [8]. Some authors believe
tion inversion recovery—FLAIR—sequences) that schizophrenia and even bipolar disorder can
[1]. These studies have allowed us to better under- be conceptualized as “connectivity disorders.”
stand the volumetric changes present in psychiat- Schizophrenia, for example, is characterized by a
ric disorders such as schizophrenia, mood global alteration in brain connectivity [9]. This
disorders (bipolar and unipolar disorders), anxiety could explain the widespread cognitive deficits
disorders, addiction, personality disorders, autism, characteristic of the disorder. Neurobiological
and attention deficit hyperactivity disorder models of mood disorders assume compromised
(ADHD). As an example, in schizophrenia, we functional regulation of prefrontal-limbic con-
now know from these neuroimaging studies that nectivity. As functional connectivity is obviously
global brain volumes are decreased in patients linked to structural connectivity, there is a need to
compared to controls, even before the first clinical precisely explore and characterize white matter
episode [2, 3]. Regional volumes are also in the context of psychiatric illness [10].
decreased, especially in the prefrontal cortex [4]. This is why DTI has steadily gained impor-
High-risk subjects are also a population of interest tance as an investigative tool in psychiatric disor-
in these pathologies and are generally defined as ders. Its unique ability to examine WM
healthy relatives of patients. They thus share some microstructure in vivo provides a means to build
common genetic risk with the patients, but without upon findings from previous classical MR stud-
the expression of the disease per se and without ies. When integrated with findings from func-
some confounding factors such as medication and tional neuroimaging studies and molecular
number of episodes. Usually, these high-risk sub- biology, it can be used to refine neurobiological
jects share most of the same features regarding models of psychiatric disorders. A brief review of
brain volumes, though at a lower amplitude than DTI findings in selected psychiatric conditions is
patients [5]. However, although such computa- provided at the end of this chapter.
tional morphometry based studies are useful, they
are unable to provide information beyond total and
regional white matter volume, density, and shape. The Development of Imaging
Functional MRI has also provided insight into Biomarkers
the mechanisms of psychiatric disorders, via the
identification of over- or under-active areas dur- The assessment of psychiatric disorders is cur-
ing the completion of specific tasks in groups of rently based entirely on clinical evaluation, with-
patients [6]. out any possibility of laboratory tests. Diagnosis,
18 DTI in Psychiatry 361

prediction of the transition to illness, course, and neuropsychiatric disorders in recent studies.
outcome of major psychiatric illnesses thus con- Koutsouleris et al. used multivariate machine
tinue to be very challenging and remain difficult learning algorithms to predict disease transition
to predict using classical clinical instruments. in schizophrenia: using T1 MRI scans from at-
The absence of an objective biomarker to assess risk subjects, they were able to predict transition
the evolution and severity of the illnesses leads to psychosis 4 years later, with an accuracy of 82
to mismanagement and increased burden [11]. % [16]. They performed this study with only 15
There is therefore a strong need to develop bio- subjects having a transition to psychosis and 18
markers of outcome to perform more personal- without such a transition.
ized healthcare plans. Recent studies have raised In mood disorders, a recent study has high-
hopes of identifying possible biomarkers that are lighted the utility of such approaches to predict
usable at an individual level [12]. The most relapses. Farb et al. [19] recruited 16 remitted
promising predictive biomarkers include neuro- unipolar depressed patients who underwent fMRI
imaging features such as white matter abnormal- while viewing sad and neutral film clips. They
ities. The development and use of such used a receiver operating characteristic analysis
biomarkers of prognosis may help to identify to determine signal cutoffs for predicting relapse.
patients that should receive specific targeted Within the depressed group, relapse was pre-
interventions [13]. dicted by medial prefrontal cortical activity and
One technique to achieve the development of contraindicated by visual cortical activity with
individual neuroimaging biomarkers usable at sensitivity and specificity scores all above 80 %.
the bedside is “Machine learning” [14]. This study clearly demonstrates the feasibility of
Techniques such as support vector machines have discovering neuroimaging-based predictors of
been developed in recent years and have already clinical outcome in mood disorders. It must be
shown potential to classify patients with psychi- noted however that the sample size of this study
atric disorders using neuroimaging data [15–17]. was quite small.
In such machine learning multivariate algorithms, A few studies have used DTI data as an entry
the computer applies a specific mathematical point for such machine learning algorithms in
method (e.g., support vector machine algorithms) psychiatry [20]. Such studies have achieved very
to find specific patterns in a “learning dataset” high rates of accuracy, sensitivity, and specificity
(group information supplied to the computer) [21–23] and are a promising application of DTI
that form the basis of rules for distinguishing the in future psychiatric research.
MRI scans of different groups (e.g., patients from
those of healthy controls). The computer then
applies these rules to new datasets (e.g., for the Planning Psychosurgical Procedures
automatic classification of patients and healthy
subjects within the sample). Therefore, a bio- Neurosurgical treatments of severe, intractable
marker is constructed, with measurable metrics psychiatric disorders using procedures that
such as specificity, sensitivity, positive and nega- destroy or disconnect brain tissue have a contro-
tive predictive values, and accuracy. versial history and despite their reported efficacy
Proof-of-concept of such approaches in psy- are not widely used. A major criticism of such
chiatry has already been demonstrated in schizo- procedures is that the pathways involved in
phrenia and autism. In 2005, Davatzikos and psychiatric illness are ill defined and therefore
colleagues [18] applied such an automated classifi- reliable surgical targets are lacking, resulting
cation technique to T1 MRI scans from 69 patients in widely variable postsurgical outcomes.
with schizophrenia and 79 healthy controls. They Nevertheless, four major techniques are in use,
achieved a classification accuracy of 81 %. which are generally accepted as safe and effica-
Such techniques have also proven capable of cious: anterior cingulotomy, subcaudate tractotomy,
predicting clinical outcome with MRI data in limbic leucotomy, and anterior capsulotomy [24].
362 J. Houenou and L. Emsell

All these procedures target the limbic territory condition per se. These caveats are not all spe-
and its connections. cific to DTI, but are generally common to all neu-
Another promising surgical approach, partic- roimaging studies of patients in psychiatry.
ularly in the treatment of depression, is deep
brain stimulation (DBS). This technique involves
the targeted stimulation of brain tissue via an Diagnosis and Patient Stratification
electrode in order to modulate neurotransmis- Presently Based on Clinical
sion. In the case of depression, improvements Assessment, Not Biomarkers
have been reported when using DBS to target the
subcallosal cingulate, ventral striatum, and ante- To date, diagnoses in psychiatry are solely based
rior limb of the internal capsule (ALIC). The on clinical assessment. The classification and
ALIC has also been targeted in obsessive- definition of the illnesses rely on guidelines and
compulsive disorder [25]. manuals approved by the psychiatry community
Given the ability of DTI to virtually delineate such as the “Diagnostic and Statistical Manual of
major pathways, it can be used to investigate the Mental Disorders” (DSM; current version DSM-
connectivity profile of ablation and electrode tar- V) of the American Psychiatric Association and
get sites in order to understand more about the the “International Statistical Classification of
biological basis of the therapeutic and unwanted Diseases and Related Health Problems” (ICD;
effects associated with the procedures, and about current version ICD-10) of the World Health
the neural circuitry involved in different aspects Organization.
of psychopathology. For example, recent DTI These classification systems define a mental
tractography studies have found that typical psy- disorder based on a collection of clinical signs
chosurgical lesion and DBS sites share similar and symptoms (“a syndrome”) and their conse-
fiber bundles within various cortical and subcor- quences. As an example, the DSM-V defines a
tical circuits involving the prefrontal cortex and mental disorder as a syndrome that occurs in an
limbic networks, including, for example, the individual, the consequences of which are clini-
medial forebrain bundle and anterior thalamic cally significant distress or disability, that must
radiation [26–28]. not be merely an expectable response to common
As the neurocircuitry of psychiatric disorders stressors and losses or a culturally sanctioned
is unraveled, DTI could also be informative in response to a particular event, that reflects an
guiding neurosurgical placement of the electrode underlying psychobiological dysfunction and is
in DBS (see Chap. 14) and for refining psycho- not primarily a result of social deviance or con-
surgical targets. Although presently such applica- flicts with society. Other accepted validity crite-
tions are very much in their infancy, in the future, ria for psychiatric disorders include those
DTI or advanced versions of the technique such established by Robins and Güze in 1970 that are
as HARDI (see Chap. 21) may rejuvenate mod- a common clinical description, the exclusion of
ern surgical interventions in psychiatry [29]. other disorders, longitudinal studies (for stability
over time), familial studies, and laboratory tests.
These definitions and the classification sys-
Special Challenges tems thus rely largely on statistical clustering of
in the Application of DTI symptoms in individuals. No single pathophysi-
in Psychiatry ological process is assumed for a disease defini-
tion such as in other medical fields. The
The application of DTI, and neuroimaging in “underlying psychobiological dysfunction” is
general in psychiatry, is an exciting challenge. vague and secondary, largely because psychiatry
Nevertheless, specific caveats must be kept in presently has no unitary pathophysiological
mind, which are related to the current classifica- model for most diseases (schizophrenia, bipolar
tion systems in psychiatry and to the psychiatric disorder, autism etc.).
18 DTI in Psychiatry 363

The issue with such a situation is that neuro- Finally, an additional layer of complexity
imaging and DTI studies currently investigate comes from the variation in time of the diagnos-
groups of patients based on clinical classifica- tic criteria used. As an example, diagnoses in
tions only. As an example, when we compare a the DSM-III and DSM-IV are not strictly identi-
group of 30 patients with “schizophrenia” with cal, and thus, studies using these different man-
“healthy controls,” one cannot know if the 30 uals cannot be directly compared. Some
patients share a common underlying etiological diagnoses disappear from the classifications,
mechanism or various physiopathological pro- while others arise.
cesses. This may explain some false negatives On the other hand, neuroimaging may help to
(because of the inclusion of patients with hetero- better define homogeneous and valid diagnostic
geneous neurobiology). This situation may also groups, by identifying clear physiopathological
explain the heterogeneity of the results if differ- processes involved. The initial goal of the
ent groups studying the same “disease” have DSM-V revision was indeed to define illnesses
included non-comparable groups. Indeed, the by using the new knowledge stemming from bio-
DSM-V authors state that an inter-rater kappa for logical, including neuroimaging studies. To
most diagnoses between 0.4 and 0.6 would be a achieve this goal, large studies comparing
realistic goal, and 0.2 and 0.4 would be accept- patients across diagnoses are recommended.
able [30]. Therefore, one cannot assume that
groups of patients with an identical diagnosis are
similar between studies. Psychiatric and Medical Comorbidity,
In addition, the stability of psychiatric diag- including Alcohol
noses over time is also open to debate. A very and Substance Abuse
recent study explored this question in a cohort of
470 first-admission patients with psychotic dis- Heterogeneous results have been obtained in neu-
orders who were systematically assessed at roimaging studies of psychiatric conditions.
baseline and during a 10-year follow-up [31]. Several sources of heterogeneity can be identi-
Diagnoses were based on best-estimate consen- fied. Amongst them, the heterogeneity of the
sus. In this report, 50.7 % of study participants’ clinical samples recruited is a crucial issue. One
diagnoses changed at some point during the source of heterogeneity comes from the classifi-
study. Therefore, a study scanning patients with cation systems used (see previous paragraph).
“first-episode schizophrenia” may include But potential biases are specifically present in
patients with first-episode schizophrenia, but neuroimaging of patients with psychiatric ill-
also patients with other future diagnoses such as nesses: comorbidity, heterogeneity of the illness,
bipolar disorder. medication, impact of illness duration, and epi-
Boundaries of diagnoses are also unclear. The sodes and impact of symptoms.
distinction between schizophrenia and bipolar
disorder has been debated since 1896, when Emil Comorbidity
Kraepelin proposed that a fundamental dichot- Patients suffering from a psychiatric illness often
omy exists between those two diagnoses (the exhibit high rates of psychiatric and somatic
“Kraepelinian dichotomy”). The existence of comorbidities. In schizophrenia, anxiety and
mixed clinical forms (“schizoaffective disorders”), depressive symptoms are very common with an
shared genetic vulnerability, diagnosis instability, estimated prevalence of 29 % for PTSD and 23 %
and common risk factors have led several authors for OCD. Depression occurs in 50 % of patients
to consider those two illnesses as belonging to with schizophrenia and 47 % also have a lifetime
the same fundamental process [32, 33]. Some diagnosis of substance abuse [34]. In patients
authors even include autism in this picture with bipolar disorder, substance use comorbidi-
(Kanner, himself, firstly described autism as ties are present in up to 72 % of patients, along
“early-onset schizophrenia”). with anxiety or multiple comorbidities [35].
364 J. Houenou and L. Emsell

For somatic conditions, cardiovascular dis- hallucinations in patients with schizophrenia


eases are far more frequent than in general popu- have already been associated with specific DTI
lation in patients with bipolar disorder [36]. In findings [40].
schizophrenia, most of the common medical con- Illness duration, severity, number of episodes,
ditions are more frequent than in general vpopu- and current symptoms may also vary between
lation [37]. The cause for this is unclear. Two samples and are known to have an influence on
hypotheses are proposed: firstly, there is a delay DTI findings in most conditions.
in diagnosis and lack in the care of somatic con- A last source of heterogeneity is the recruit-
ditions in patients with psychiatric illnesses. ment mode. Patients recruited via the press, inpa-
Secondly, some of these somatic conditions are tient or outpatient facilities differ on many
inherent to the pathophysiology of psychiatric demographic and clinical characteristics.
diseases [36].
The very high rates of comorbidities in psy- Medication
chiatric illnesses raise two challenges in DTI Another major confounding variable is psycho-
studies. First of all, the inclusion of patients with tropic medication. For major psychiatric illnesses
comorbidities may introduce a bias in the inter- such as schizophrenia, bipolar disorder, or severe
pretation of the results. The differences found unipolar disorder, virtually all patients are taking
between patients and controls may be caused by one, or more usually, several psychotropic medi-
the psychiatric illness itself or by its comorbidity. cations such as antipsychotics, mood stabilizers,
As an example, DTI differences between patients antidepressants, and benzodiazepines. All these
with bipolar disorder and controls may be linked psychotropic medications may affect brain struc-
to alcohol use disorder in these patients. Indeed, ture. The most common example is the neuro-
even detoxified subjects with alcohol use disor- trophic effect of lithium on grey matter volumes.
der exhibit DTI abnormalities [38], which are There is presently a lack of knowledge regarding
probably of larger magnitude than those of bipo- the effect of these medications on brain white
lar disorder. Even somatic conditions such as dia- matter, but current evidence suggests a limited
betes may bias the DTI results [39]. One solution impact on DTI variables [41].
to this issue may be the inclusion of comorbidity- Similarly to the comorbidity issue, the recruit-
free patients in DTI studies. However, this ment of medication-free patients, apart from
approach introduces a sampling bias as most of being very difficult, may lead to sampling issues.
the patients have comorbidities and therefore,
comorbidity-free patients may not be representa-
tive of typical patient populations. Special Considerations
in the Scanner: The Effect of Motion,
Heterogeneity of Illness Active Symptoms, and Informed
Another source of heterogeneity in the results of Consent
neuroimaging studies is the heterogeneity of the
clinical samples, which probably confounds the Another source of noise and bias that is crucial in
observed results. The clinical characteristics of DTI studies is the compliance of the patients to
the patients studied are diverse, with, for exam- the instructions given by the scanning staff. More
ple, different forms of schizophrenia (with or specifically, head motion is a major source of
without hallucinations etc.) or different subtypes noise in most neuroimaging studies [42]. Patients
of BD (e.g., types I and II, rapid cycling) and dif- are more prone to head motion than controls
ferences in age at onset (early, intermediate, late). because of several factors including minor neuro-
Unipolar depression is probably even more logical signs associated with the disease itself,
diverse. In anxiety disorders, PTSD may arise medication, motivation, and anxiety. In DTI,
from various types of trauma. Some of these clin- head motion is also a source of noise despite
ical features such as the presence or absence of motion correction algorithms [43]. In movement
18 DTI in Psychiatry 365

disorders, FA values have proven to be robust arising from altered structural connectivity has
despite head motion, which is very encouraging been strengthened.
for the psychiatric field [44].
Aside from motion, the nature of psychiatric
symptoms that patients present with during Which Fiber Bundles Are of Interest
scanning may hinder optimal data acquisition. in Psychiatric Disorders?
For example, depressed subjects may be less
motivated to attend scanning sessions or suffer DTI has been applied to the investigation of a
higher levels of anxiety. Manic or actively psy- number of psychiatric disorders to varying
chotic individuals may be too restless or anxious degrees and using a range of analysis methods to
to tolerate scanning. The resulting scans may explore whole brain white matter, specific fiber
suffer more motion artifacts or scanning may be tracts, and tract subregions. The most commonly
terminated before the acquisition is complete. In reported deficits are found in frontal and tempo-
all these cases, the most optimum results will be ral white matter and tracts that subserve the lim-
obtained by employing strategies to increase bic system. Such tracts include the various
patient compliance with the scanning proce- subregions of the corpus callosum (CC), cingu-
dures. It is therefore extremely important that lum bundle (CB), superior (SLF) and inferior
staff scanning subjects with active psychiatric longitudinal fasciculi (ILF), thalamic radiations,
disorders pay special attention to ensuring that and uncinate fasciculus (UF) [1, 46]. Impaired
patients receive clear and complete instructions WM microstructure in these regions is hypothe-
on the scanning procedure and what they can sized to contribute to a breakdown in the regula-
expect to experience whilst in the scanner. Along tion of higher functions relating to cognition,
these same lines, it is vital that the nontherapeu- emotion, and memory, which are typically com-
tic investigation of psychiatric patients conforms promised in psychiatric illness. Some ascending
to an ethical framework that takes into account and descending fiber systems such as the corona
the ability of the patient to provide informed radiata, internal vcapsules, cerebral and cerebel-
consent [45]. lar peduncles feature more predominantly in neu-
rodevelopmental disorders such as autism and
ADHD, and also in schizophrenia, and may
DTI findings in Psychiatric Disorders underlie the psychomotor features of these
illnesses.
Despite the inherent difficulties in acquiring Reported alterations are however by no means
good comparative data in psychiatric popula- limited to these areas and neither are such find-
tions, research in psychiatry has greatly benefited ings universal. This likely reflects the heteroge-
from neuroimaging. Earlier work using CT, MRI, neity of both the clinical populations studied and
and PET fundamentally altered the perception of the methodology employed to investigate them.
psychiatric illness from an intangible, unquantifi- Furthermore, several regions such as the WM of
able, functional disturbance without organic the medial temporal lobe and corpus callosum
pathology, to a collection of disorders for which emerge consistently in meta-analyses of different
measurable neurological changes in brain struc- disorders. This illustrates the lack of specificity
ture and biochemical function could be identified of DTI changes in psychiatric illnesses and may
and visualized. With the ability to investigate be reflective of the considerable overlap in symp-
white matter, DTI continues to advance our tomatology between them. In this context, DTI
understanding of the nature of these structural metrics in isolation cannot be used diagnostically
changes. Since the advent of the technique, the but provide useful additional data in a multi-
role of white matter alterations as a core feature modal framework incorporating for example,
of mental illness pathophysiology has become genetic, neuropsychological, psychosocial, and
apparent, and the concept of psychiatric disease clinical measures.
366 J. Houenou and L. Emsell

DTI Findings in Selected Psychiatric in severe cases, suicidal thoughts. Bipolar disor-
Disorders der is less common, affecting 1 % of the popula-
tion, and is characterized by alternating periods
Schizophrenia of severe depression and hypomania or mania.
During (hypo) manic episodes, patients experi-
Schizophrenia is a disorder of thought, percep- ence elevated mood, increased energy, reduced
tion, emotion, and behavior affecting an esti- need for sleep, talk more quickly, may make
mated 1 % of the population. Patients may unrealistic plans, overspend, engage in risky
experience both “positive” symptoms, such as behavior, become irritable, aggressive, and abuse
hallucinations, delusions, altered thoughts and alcohol and drugs. Some patients may also expe-
feelings of being controlled, and “negative” rience psychosis, a state in which their perception
symptoms characterized by withdrawal, flattened of reality becomes distorted. In this context,
affect, and anhedonia. symptoms of BD and schizophrenia overlap. It is
It is the most widely studied psychiatric disor- interesting that DTI findings in BD also parallel
der using DTI, with over 300 studies listed on those in schizophrenia. However, FA reductions
PubMed at the time of writing (early 2015). are less widely reported across the whole brain in
There are few negative studies, with the majority BD and there are considerably more negatives
reporting FA reductions in more than one brain studies. There are also some reports of regional
region [47]. Although FA reduction in frontal and FA increase [49]. Regionally, FA reductions tend
temporal WM appears most frequently reported, to be found in frontal and temporal WM. Corpus
there are also reports of such decreases in pari- callosum deficits feature strongly, particularly
etal, occipital, and even cerebellar white matter, anterior (genu) and posterior (splenium) projec-
suggesting widespread diffuse whole brain tions [50–52]. FA reductions also predominate in
pathology, consistent with findings of wide- anatomically closely related tracts, such as por-
spread grey matter reductions and functional tions of the SLF, ILF, IFOF, posterior thalamic
impairments detected using other imaging radiation, and cingulum [53]. Such regions are
modalities [10]. A recent meta-analysis of DTI classically associated with emotional regulation,
studies in schizophrenia described two distinct working memory, and facial processing; func-
regions where FA was reduced consistently: one tions that are impaired in BD. Interestingly, these
in the left perigenual WM of the frontal lobe and regions, which emerged consistently in a meta-
a second region, in the medial temporal lobe analysis of 11 DTI studies [46], parallel the two
[48]. The authors postulate that these regions regions identified in the schizophrenia meta-anal-
represent two distinct networks that are compro- ysis described above.
mised in schizophrenia, leading to a disconnec- Findings in MDD are significantly more het-
tion of important fronto-temporal grey matter erogeneous and overlap considerably with
functional areas. changes identified in BD. Strikingly, a recent
meta-analysis [54] not only identified FA defi-
cits in the callosal genu and body but also found
Mood Disorders them in precisely the same posterior WM region
encompassing the right ILF, IFOF, and poste-
The next most widely investigated psychiatric rior thalamic radiation, as the meta-analysis of
conditions aremajor depressive disorder DTI studies in BD and schizophrenia, all per-
(MDD) and bipolar disorder (BD). Depression formed by different authors [46, 48]. Findings
is a common disorder affecting up to one in five diverge somewhat from BD and schizophrenia,
people in their lifetime. It is characterized by where FA reductions in MDD are found in more
extended periods of low mood, sadness, anhedo- dorsal regions of the PFC, compared to more
nia, impaired concentration, altered sleep and ventral and perigenual PFC regions in BD and
appetite, feelings of guilt and worthlessness, and schizophrenia.
18 DTI in Psychiatry 367

Anxiety Disorders The typical behaviors associated with OCD


are hypothesized to reflect cortical dysregulation
Anxiety disorders are common, affecting one in of cortico-thalamo-striatal circuits including the
ten people in their lifetime. They are character- orbitofrontal cortex, cingulate, and caudate [59].
ized by both psychological effects, including Positive DTI findings predominate, with FA
increased worry, irritability, fear, and impaired changes, i.e., reductions and also increases
sleep; and somatic complaints, such as dizziness, reported in tracts associated with these regions,
palpitations, trembling, sweating, rapid breath- for example, in the cingulum bundle, internal
ing, and gastrointestinal disruption. Some anxiety capsule, anterior thalamic radiation, superior lon-
disorders are a permanent and disruptive feature gitudinal fasciculus and inferior fronto-occipital
of a patient’s life such as generalized anxiety fasciculus, as well as in the corpus callosum,
disorder (GAD), obsessive-compulsive disor- frontal and parietal white matter [60, 61].
der (OCD), and posttraumatic stress disorder
(PTSD). Other forms of anxiety disorder arise
only in certain situations, which are typically Personality Disorders
stressful for the affected individual. For example,
panic disorder is characterized by intense, iso- Personality disorders are common, and arise
lated “attacks” that come on quickly, and pho- from the abnormal expression of certain charac-
bias, which give rise to feelings of anxiety and ter traits that diverge from the sociocultural norm
fear when the sufferer is exposed to something of the individual’s environment. Such traits
that is not usually dangerous, such as house spi- include being overly suspicious, impulsive,
ders or traveling by air. Neurobiological models overly emotional, and anxious. These traits may
of anxiety propose a disruption to key networks lead those with the disorder to engage in destruc-
that modulate fear and attention and involve brain tive and harmful behavior to themselves and/or
structures such as the medial prefrontal cortex, others, and they may find it difficult to function in
posterior cingulate cortex, insula, brain stem, hip- healthy relationships and formal educational or
pocampus, and amygdala [55]. The cingulum employment settings.
bundle is central to these networks and features Research using DTI to study personality dis-
prominently in DTI studies of anxiety. orders is limited. Two studies report reduced
Most types of anxiety disorder have been FA in orbitofrontal WM [62, 63], whilst another
investigated using DTI, but studies are less reports no FA decrease in borderline personal-
numerous than in the schizophrenia and mood ity disorder patients compared to a control
disorders. Two studies have investigated panic group [64]. However, interpretation of these
disorder. One ROI analysis of the anterior and findings is compromised by the inclusion of
posterior cingulum reported increased FA in this patients with different comorbid psychiatric
structure [56]. The other utilized a voxel-based conditions and analyses based on suboptimal
approach and found FA decreases in the right DTI data. Only two studies have examined
IFOF, left callosal body, and left SLF [57]. schizotypal personality disorder. The first used
DTI studies of PTSD are limited and include an automated ROI analysis of the uncinate fas-
investigations into the effect of childhood trauma ciculus and cingulum bundle in DTI linescan
measured during childhood and during adult- data and found reduced FA in the uncinate only
hood, and also the effect of adult trauma. A meta- [65]. The second investigated a larger sample in
analysis of seven studies investigating a more elaborate analysis examining WM
trauma-exposed adults identified FA decreases in underlying Brodmann regions of the dorsolat-
nine clusters and increases in six clusters, which eral PFC, cingulate gyrus, and temporal lobe,
included different regions of the cingulum. and found both FA reductions in temporal WM
Interestingly, volumetric reductions have also and posterior cingulum and FA increase in the
been commonly reported in this structure [58]. subgenual PFC [66].
368 J. Houenou and L. Emsell

Neurodevelopmental Disorders tributed to the results, which they discussed in the


context of potential confounds in such DTI stud-
Neuroimaging studies in children have additional ies and to caution against the use of DTI metrics
challenges beyond those described previously as a biomarker for single-subject diagnosis.
(see Chap. 13). Notably, the effect of age is likely
to be an important confound in such studies.
Given the different trajectories of white matter Attention Deficit Hyperactivity
development both within and between typically Disorder
developing children and children affected by psy-
chopathology, cross-sectional studies including Attention Deficit Hyperactivity Disorder
different age groups limit comparability between (ADHD) is a neurodevelopmental, behavioral
studies and generalizability of findings. In spite disorder affecting up to 5 % of school age chil-
of such issues, DTI is providing data supporting dren, characterized by impaired attention and
dyconnectivity models of two key neurodevelop- concentration with increased impulsivity and
mental disorders. hyperactivity. One neurobiological theory pro-
poses that the disorder reflects abnormal fronto-
striatal-cerebellar circuitry. Findings from DTI
Autism appear to support this model with several studies
reporting both FA alterations in a range of tracts
Autism (autism spectrum disorder/ASD) is a per- subserving these regions. For example, a meta-
vasive neurodevelopmental disorder emerging in analysis including nine VBA studies of both
early childhood that is characterized by impaired pediatric and adult populations (173 ADHD
social and communication skills, repetitive, ste- patients and 169 healthy controls) identified five
reotyped behavior, and increased sensitivity to foci of altered FA within the callosal genu, ante-
external stimuli. The dramatic increase in ASD rior corona radiate, internal capsule, and cerebel-
diagnosis in recent years has been mirrored by an lar white matter [70]. ROI studies have also
increase in neuroimaging research investigating reported reduced anisotropy in overlapping
the neurobiological basis of the condition. regions, including the middle cerebellar peduncle
Studies employing active and resting state fMRI [71], corticospinal tract [72], internal capsules,
have demonstrated aberrant functional connec- and corpus callosum [73, 74]. Anisotropy
tivity in ASD, which may be driven by impaired increases have also been reported in frontal and
structural connectivity, i.e., WM pathology [67, temporal white matter [75, 76]. Interestingly, one
68]. However, DTI findings have been heteroge- study of the basal ganglia did not find group dif-
neous with a particular lack of consensus on the ferences in FA or MD, but found an increase in
location of FA and MD changes in ASD. Most FA with age in the ADHD group that was absent
studies have found FA to be reduced; however in the controls, hinting at delayed WM develop-
few studies have reported this reduction in the ment that normalizes in adulthood [77]. Indeed,
same region and other studies have failed to find many cases of ADHD resolve with increasing
FA reductions in these regions [69]. A relatively maturity, whilst others persist into adulthood,
large recent study including 39 young autistic which may reflect different illness subtypes or
children found widespread, minor FA reductions pathophysiological mechanisms.
and MD increases in the order of 1–2 % com-
pared to 39 typically developing children [69].
Notably, this study used two different voxel- Alcohol Use Disorders
based approaches and found discrepancies in the
level of statistical significance in regions reported The harmful use of alcohol is widespread across
between the methods. The authors also identified the globe, with many Eastern European coun-
image artifacts in their data that may have con- tries, Thailand, Korea, and Columbia reporting
18 DTI in Psychiatry 369

prevalence rates of over 10 % in men (WHO). References


In the context of abuse and dependence, alcohol
misuse is a common and significant comorbid- 1. Emsell L, Mcdonald C. The structural neuroimaging
of bipolar disorder. Int Rev Psychiatry.
ity in psychiatric disorders [78]. The damaging
2009;21:297–313.
effect of alcohol on the brain through acute 2. Levitt JJ, Bobrow L, Lucia D, Srinivasan P. A selec-
cytotoxity and the sequelae of chronic overcon- tive review of volumetric and morphometric imaging
sumption is well documented [79]. One DTI in schizophrenia. Curr Top Behav Neurosci.
2010;4:243–81.
ROI based study investigating the acute effects
3. Thermenos HW, Keshavan MS, Juelich RJ, Molokotos
of wine consumption on different brain regions E, Whitfield-Gabrieli S, Brent BK, et al. A review of
at 0.5, 1, 2, and 3 h following consumption neuroimaging studies of young relatives of individu-
reported significant changes in ADC in the cere- als with schizophrenia: a developmental perspective
from schizotaxia to schizophrenia. Am J Med Genet B
bral peduncles, thalamus and frontal WM, and
Neuropsychiatr Genet. 2013;162B:604–35.
in FA in frontal WM over time that may reflect 4. Fornito A, Yucel M, Patti J, Wood SJ, Pantelis
the development of cytotoxic edema and subse- C. Mapping grey matter reductions in schizophrenia:
quent recovery [80]. Another voxel-based study an anatomical likelihood estimation analysis of voxel-
based morphometry studies. Schizophr Res.
investigating 14 adolescent binge drinkers
2009;108:104–13.
(defined as drinking five alcoholic beverages in 5. Whalley HC, Harris JC, Lawrie SM. The neurobio-
one sitting) and equally matched controls logical underpinnings of risk and conversion in rela-
reported widespread FA reductions that tives of patients with schizophrenia. Int Rev
Psychiatry. 2007;19:383–97.
appeared to be dose dependent [81]. Studies of
6. Linden DE. The challenges and promise of neuroim-
“uncomplicated alcoholics” have aimed to aging in psychiatry. Neuron. 2012;73:8–22.
characterize long-term effects of chronic alco- 7. Walterfang M, Malhi GS, Wood AG, Reutens DC,
hol exposure and have reported regional FA Chen J, Barton S, et al. Corpus callosum size and
shape in established bipolar affective disorder. Aust N
reductions in anterior and superior association
Z J Psychiatry. 2009;43:838–45.
bundles and in the corpus callosum [82, 83]. 8. Greicius M. Resting-state functional connectivity in
One study investigating mesencephalic fiber neuropsychiatric disorders. Curr Opin Neurol.
tracts in detoxified subjects reported ADC 2008;21:424–30.
9. Dauvermann MR, Whalley HC, Schmidt A, Lee GL,
increases and an 18 % reduction in recon-
Romaniuk L, Roberts N, et al. Computational neuro-
structed tracts per unit volume between the psychiatry - schizophrenia as a cognitive brain net-
midbrain and pons [38]. Alcohol therefore work disorder. Front Psychiatry. 2014;5:30.
appears to impact upon DTI metrics, whether 10. Fornito A, Zalesky A, Pantelis C, Bullmore
ET. Schizophrenia, neuroimaging and connectomics.
consumed in moderate or large amounts, and
Neuroimage. 2012;62:2296–314.
these effects are measurable in both acute and 11. Kupfer DJ. The increasing medical burden in bipolar
chronic phases of overuse, as well as in states of disorder. JAMA. 2005;293:2528–30.
detoxification. As such, alcohol use should be 12. Singh I, Rose N. Biomarkers in psychiatry. Nature.
2009;460:202–7.
accounted for in DTI investigations and may
13. Houenou J, d’Albis MA, Vederine FE, Henry C,
represent an important confound in DTI studies Leboyer M, Wessa M. Neuroimaging biomarkers in
of psychiatric disorders. bipolar disorder. Front Biosci (Elite Ed).
In summary, DTI is a valuable tool in the field 2012;4:593–606.
14. Lemm S, Blankertz B, Dickhaus T, Muller
of psychiatry, particularly in clinical research. It
KR. Introduction to machine learning for brain imag-
is not without its limitations, and implementing ing. Neuroimage. 2011;56:387–99.
the technique properly in psychiatric populations 15. Duchesnay E, Cachia A, Boddaert N, Chabane N,
requires careful attention. In the future, concur- Mangin JF, Martinot JL, et al. Feature selection and
classification of imbalanced datasets: application to
rent advances in both neuroimaging and biologi-
PET images of children with autistic spectrum disor-
cal psychiatry should converge with advances in ders. Neuroimage. 2011;57:1003–14.
allied disciplines in order to inform and improve 16. Koutsouleris N, Meisenzahl EM, Davatzikos C,
clinical practice and the care of patients with psy- Bottlender R, Frodl T, Scheuerecker J, et al. Use of
neuroanatomical pattern classification to identify
chiatric disorders.
370 J. Houenou and L. Emsell

subjects in at-risk mental states of psychosis and pre- 31. Bromet EJ, Kotov R, Fochtmann LJ, Carlson GA,
dict disease transition. Arch Gen Psychiatry. 2009;66: Tanenberg-Karant M, Ruggero C, et al. Diagnostic
700–12. shifts during the decade following first admission for
17. Iwabuchi SJ, Liddle PF, Palaniyappan L. Clinical util- psychosis. Am J Psychiatry. 2011;168:1186–94.
ity of machine-learning approaches in schizophrenia: 32. Burmeister M, McInnis MG, Zollner S. Psychiatric
improving diagnostic confidence for translational genetics: progress amid controversy. Nat Rev Genet.
neuroimaging. Front Psychiatry. 2013;4:95. 2008;9:527–40.
18. Davatzikos C, Shen D, Gur RC, Wu X, Liu D, Fan Y, 33. Kas MJ, Fernandes C, Schalkwyk LC, Collier
et al. Whole-brain morphometric study of schizophre- DA. Genetics of behavioural domains across the neu-
nia revealing a spatially complex set of focal abnor- ropsychiatric spectrum; of mice and men. Mol
malities. Arch Gen Psychiatry. 2005;62:1218–27. Psychiatry. 2007;12:324–30.
19. Farb NA, Anderson AK, Bloch RT, Segal ZV. Mood- 34. Buckley PF, Miller BJ, Lehrer DS, Castle
linked responses in medial prefrontal cortex predict DJ. Psychiatric comorbidities and schizophrenia.
relapse in patients with recurrent unipolar depression. Schizophr Bull. 2009;35:383–402.
Biol Psychiatry. 2011;70:366–72. 35. Bauer MS, Altshuler L, Evans DR, Beresford T,
20. Wang P, Verma R. On classifying disease-induced pat- Williford WO, Hauger R. Prevalence and distinct cor-
terns in the brain using diffusion tensor images. Med relates of anxiety, substance, and combined comor-
Image Comput Comput Assist Interv. 2008; bidity in a multi-site public sector sample with bipolar
11:908–16. disorder. J Affect Disord. 2005;85:301–15.
21. Ingalhalikar M, Kanterakis S, Gur R, Roberts TP, 36. Leboyer M, Soreca I, Scott J, Frye M, Henry C,
Verma R. DTI based diagnostic prediction of a disease Tamouza R, et al. Can bipolar disorder be viewed as a
via pattern classification. Med Image Comput Comput multi-system inflammatory disease? J Affect Disord.
Assist Interv. 2010;13:558–65. 2012;141:1–10.
22. Ardekani BA, Tabesh A, Sevy S, Robinson DG, 37. Laursen TM, Munk-Olsen T, Gasse C. Chronic
Bilder RM, Szeszko PR. Diffusion tensor imaging somatic comorbidity and excess mortality due to natu-
reliably differentiates patients with schizophrenia ral causes in persons with schizophrenia or bipolar
from healthy volunteers. Hum Brain Mapp. 2011; affective disorder. PLoS One. 2011;6:e24597.
32:1–9. 38. Chanraud S, Reynaud M, Wessa M, Penttila J,
23. Besga A, Termenon M, Grana M, Echeveste J, Perez Kostogianni N, Cachia A, et al. Diffusion tensor trac-
JM, Gonzalez-Pinto A. Discovering Alzheimer’s dis- tography in mesencephalic bundles: relation to mental
ease and bipolar disorder white matter effects building flexibility in detoxified alcohol-dependent subjects.
computer aided diagnostic systems on brain diffusion Neuropsychopharmacology. 2009;34:1223–32.
tensor imaging features. Neurosci Lett. 39. Antenor-Dorsey JA, Meyer E, Rutlin J, Perantie DC,
2012;520:71–6. White NH, Arbelaez AM, et al. White matter micro-
24. Mashour GA, Walker EE, Martuza RL. Psychosurgery: structural integrity in youth with type 1 diabetes.
past, present, and future. Brain Res Brain Res Rev. Diabetes. 2013;62:581–9.
2005;48:409–19. 40. Allen P, Modinos G, Hubl D, Shields G, Cachia A,
25. Ward HE, Hwynn N, Okun MS. Update on deep brain Jardri R, et al. Neuroimaging auditory hallucinations
stimulation for neuropsychiatric disorders. Neurobiol in schizophrenia: from neuroanatomy to neurochem-
Dis. 2010;38:346–53. istry and beyond. Schizophr Bull. 2012;38:695–703.
26. Gutman DA, Holtzheimer PE, Behrens TE, Johansen- 41. Hafeman DM, Chang KD, Garrett AS, Sanders EM,
Berg H, Mayberg HS. A tractography analysis of two Phillips ML. Effects of medication on neuroimaging
deep brain stimulation white matter targets for depres- findings in bipolar disorder: an updated review.
sion. Biol Psychiatry. 2009;65:276–82. Bipolar Disord. 2012;14:375–410.
27. Schlaepfer TE, Bewernick BH, Kayser S, Madler B, 42. Van Dijk KR, Sabuncu MR, Buckner RL. The influ-
Coenen VA. Rapid effects of deep brain stimulation ence of head motion on intrinsic functional connectiv-
for treatment-resistant major depression. Biol ity MRI. Neuroimage. 2012;59:431–8.
Psychiatry. 2013;73:1204–12. 43. Jones DK, Cercignani M. Twenty-five pitfalls in the
28. Schoene-Bake JC, Parpaley Y, Weber B, Panksepp J, analysis of diffusion MRI data. NMR Biomed.
Hurwitz TA, Coenen VA. Tractographic analysis of 2010;23:803–20.
historical lesion surgery for depression. 44. Muller HP, Sussmuth SD, Landwehrmeyer GB,
Neuropsychopharmacology. 2010;35:2553–63. Ludolph A, Tabrizi SJ, Kloppel S, et al. Stability
29. Henderson JM. “Connectomic surgery”: diffusion effects on results of diffusion tensor imaging analysis
tensor imaging (DTI) tractography as a targeting by reduction of the number of gradient directions due
modality for surgical modulation of neural networks. to motion artifacts: an application to presymptomatic
Front Integr Neurosci. 2012;6:15. Huntington’s disease. PLoS Curr. 2011;3:RRN1292.
30. Kraemer HC, Kupfer DJ, Clarke DE, Narrow WE, 45. Gupta UC, Kharawala S. Informed consent in psy-
Regier DA. DSM-5: how reliable is reliable enough? chiatry clinical research: a conceptual review of
Am J Psychiatry. 2012;169:13–5. issues, challenges, and recommendations. Perspect
Clin Res. 2012;3:8–15.
18 DTI in Psychiatry 371

46. Vederine FE, Wessa M, Leboyer M, Houenou J. A 60. Benedetti F, Giacosa C, Radaelli D, Poletti S, Pozzi E,
meta-analysis of whole-brain diffusion tensor imag- Dallaspezia S, et al. Widespread changes of white
ing studies in bipolar disorder. Prog matter microstructure in obsessive-compulsive disor-
Neuropsychopharmacol Biol Psychiatry. 2011;35(8): der: effect of drug status. Eur Neuropsychopharmacol.
1820–6. 2013;23:581–93.
47. Kyriakopoulos M, Frangou S. Recent diffusion tensor 61. Bora E, Harrison BJ, Fornito A, Cocchi L, Pujol J,
imaging findings in early stages of schizophrenia. Fontenelle LF, et al. White matter microstructure in
Curr Opin Psychiatry. 2009;22:168–76. patients with obsessive-compulsive disorder.
48. Ellison-Wright I, Bullmore E. Meta-analysis of diffu- J Psychiatry Neurosci. 2011;36:42–6.
sion tensor imaging studies in schizophrenia. 62. Carrasco JL, Tajima-Pozo K, Diaz-Marsa M, Casado
Schizophr Res. 2009;108:3–10. A, Lopez-Ibor JJ, Arrazola J, et al. Microstructural
49. Heng S, Song AW, Sim K. White matter abnormalities white matter damage at orbitofrontal areas in border-
in bipolar disorder: insights from diffusion tensor line personality disorder. J Affect Disord. 2012;
imaging studies. J Neural Transm. 2010;117:639–54. 139:149–53.
50. Sarrazin S, Poupon C, Linke J, Wessa M, Phillips M, 63. Grant JE, Correia S, Brennan-Krohn T, Malloy PF,
Delavest M, et al. A multicenter tractography study of Laidlaw DH, Schulz SC. Frontal white matter integ-
deep white matter tracts in bipolar I disorder: psy- rity in borderline personality disorder with self-
chotic features and interhemispheric disconnectivity. injurious behavior. J Neuropsychiatry Clin Neurosci.
JAMA Psychiatry. 2014;71:388–96. 2007;19:383–90.
51. Emsell L, Langan C, Van Hecke W, Barker GJ, 64. Rusch N, Weber M, Il’yasov KA, Lieb K, Ebert D,
Leemans A, Sunaert S, et al. White matter differences Hennig J, et al. Inferior frontal white matter micro-
in euthymic bipolar I disorder: a combined magnetic structure and patterns of psychopathology in women
resonance imaging and diffusion tensor imaging with borderline personality disorder and comorbid
voxel-based study. Bipolar Disord. 2013;15:365–76. attention-deficit hyperactivity disorder. Neuroimage.
52. Emsell L, Leemans A, Langan C, Van Hecke W, 2007;35:738–47.
Barker GJ, McCarthy P, et al. Limbic and callosal 65. Nakamura M, McCarley RW, Kubicki M, Dickey CC,
white matter changes in euthymic bipolar I disorder: Niznikiewicz MA, Voglmaier MM, et al. Fronto-
an advanced diffusion magnetic resonance imaging temporal disconnectivity in schizotypal personality
tractography study. Biol Psychiatry. 2013;73: disorder: a diffusion tensor imaging study. Biol
194–201. Psychiatry. 2005;58:468–78.
53. Emsell L, Chaddock C, Forde N, Van Hecke W, 66. Hazlett EA, Goldstein KE, Tajima-Pozo K, Speidel
Barker GJ, Leemans A et al. White matter microstruc- ER, Zelmanova Y, Entis JJ, et al. Cingulate and tem-
tural abnormalities in families multiply affected with poral lobe fractional anisotropy in schizotypal person-
bipolar I disorder: a diffusion tensor tractography ality disorder. Neuroimage. 2011;55:900–8.
study. Psychol Med. 2013;10:2139–50 67. Muller RA, Shih P, Keehn B, Deyoe JR, Leyden KM,
54. Liao Y, Huang X, Wu Q, Yang C, Kuang W, Du M, Shukla DK. Underconnected, but how? A survey of
et al. Is depression a disconnection syndrome? Meta- functional connectivity MRI studies in autism spec-
analysis of diffusion tensor imaging studies in patients trum disorders. Cereb Cortex. 2011;21:2233–43.
with MDD. J Psychiatry Neurosci. 2013;38:49–56. 68. Verly M, Verhoeven J, Zink I, Mantini D, Peeters R,
55. Etkin A. Functional neuroanatomy of anxiety: a neu- Deprez S, et al. Altered functional connectivity of the
ral circuit perspective. Curr Top Behav Neurosci. language network in ASD: role of classical language
2010;2:251–77. areas and cerebellum. Neuroimage Clin. 2014;
56. Han DH, Renshaw PF, Dager SR, Chung A, Hwang J, 4:374–82.
Daniels MA, et al. Altered cingulate white matter con- 69. Walker L, Gozzi M, Lenroot R, Thurm A, Behseta B,
nectivity in panic disorder patients. J Psychiatr Res. Swedo S, et al. Diffusion tensor imaging in young
2008;42:399–407. children with autism: biological effects and potential
57. Lai CH, Wu YT. Fronto-occipital fasciculus, corpus confounds. Biol Psychiatry. 2012;72:1043–51.
callosum and superior longitudinal fasciculus tract 70. van Ewijk H, Heslenfeld DJ, Zwiers MP, Buitelaar
alterations of first-episode, medication-naive and late- JK, Oosterlaan J. Diffusion tensor imaging in atten-
onset panic disorder patients. J Affect Disord. tion deficit/hyperactivity disorder: a systematic review
2013;146:378–82. and meta-analysis. Neurosci Biobehav Rev.
58. Daniels JK, Lamke JP, Gaebler M, Walter H, Scheel 2012;36:1093–106.
M. White matter integrity and its relationship to 71. Bechtel N, Kobel M, Penner IK, Klarhofer M,
PTSD and childhood trauma--a systematic review and Scheffler K, Opwis K, et al. Decreased fractional
meta-analysis. Depress Anxiety. 2013;30:207–16. anisotropy in the middle cerebellar peduncle in chil-
59. Fineberg NA, Potenza MN, Chamberlain SR, Berlin HA, dren with epilepsy and/or attention deficit/hyperactiv-
Menzies L, Bechara A, et al. Probing compulsive and ity disorder: a preliminary study. Epilepsy Behav.
impulsive behaviors, from animal models to endopheno- 2009;15:294–8.
types: a narrative review. Neuropsychopharmacology. 72. Hamilton LS, Levitt JG, O’Neill J, Alger JR, Luders
2010;35:591–604. E, Phillips OR, et al. Reduced white matter integrity
372 J. Houenou and L. Emsell

in attention-deficit hyperactivity disorder. issues and findings from the NESARC. Curr Opin
Neuroreport. 2008;19:1705–8. Psychiatry. 2012;25:165–71.
73. Pavuluri MN, Yang S, Kamineni K, Passarotti AM, 79. Harper C. The neuropathology of alcohol-related
Srinivasan G, Harral EM, et al. Diffusion tensor imag- brain damage. Alcohol Alcohol. 2009;44:136–40.
ing study of white matter fiber tracts in pediatric bipo- 80. Kong LM, Zheng WB, Lian GP, Zhang HD. Acute
lar disorder and attention-deficit/hyperactivity effects of alcohol on the human brain: diffusion tensor
disorder. Biol Psychiatry. 2009;65:586–93. imaging study. AJNR Am J Neuroradiol.
74. Cao Q, Sun L, Gong G, Lv Y, Cao X, Shuai L, et al. 2012;33:928–34.
The macrostructural and microstructural abnormali- 81. McQueeny T, Schweinsburg BC, Schweinsburg AD,
ties of corpus callosum in children with attention defi- Jacobus J, Bava S, Frank LR, et al. Altered white mat-
cit/hyperactivity disorder: a combined morphometric ter integrity in adolescent binge drinkers. Alcohol
and diffusion tensor MRI study. Brain Res. Clin Exp Res. 2009;33:1278–85.
2010;1310:172–80. 82. Pfefferbaum A, Rosenbloom M, Rohlfing T, Sullivan
75. Peterson DJ, Ryan M, Rimrodt SL, Cutting LE, EV. Degradation of association and projection white
Denckla MB, Kaufmann WE, et al. Increased regional matter systems in alcoholism detected with quantita-
fractional anisotropy in highly screened attention- tive fiber tracking. Biol Psychiatry. 2009;65:680–90.
deficit hyperactivity disorder (ADHD). J Child 83. Harris GJ, Jaffin SK, Hodge SM, Kennedy D,
Neurol. 2011;26:1296–302. Caviness VS, Marinkovic K, et al. Frontal white mat-
76. Davenport ND, Karatekin C, White T, Lim ter and cingulum diffusion tensor imaging deficits in
KO. Differential fractional anisotropy abnormalities alcoholism. Alcohol Clin Exp Res.
in adolescents with ADHD or schizophrenia. 2008;32:1001–13.
Psychiatry Res. 2010;181:193–8.
77. Silk TJ, Vance A, Rinehart N, Bradshaw JL,
Cunnington R. Structural development of the basal
ganglia in attention deficit hyperactivity disorder: a Suggested Reading
diffusion tensor imaging study. Psychiatry Res.
2009;172:220–5. Thomason ME, Thompson PM. Diffusion imaging, white
78. Hasin D, Kilcoyne B. Comorbidity of psychiatric and matter, and psychopathology. Annu Rev Clin Psychol.
substance use disorders in the United States: current 2011;7:63–85.
Diffusion Tensor Imaging
in Traumatic Brain Injury 19
Süleyman Sener, Paul M. Parizel,
and Andrew I.R. Maas

ever, changing: in low and middle income coun-


Learning Points tries, injuries are mainly caused by road traffic
incidents and involve vulnerable road users
• Challenges in the use of DTI in TBI. (pedestrians, cyclists), who are mostly young
• DTI as a diagnostic tool for severity of TBI. men. In higher income countries, injuries are
• DTI as an outcome predictor. more often caused by falls and involve older
patients. These patients more frequently have
comorbidities, and often take medication, includ-
Introduction ing anticoagulants and platelet aggregation inhib-
itors. Both comorbidity and medications may
Traumatic brain injury (TBI) is defined as an influence the disease process. TBI does not only
alteration in brain function, or other evidence of affect the patient, but also has a direct impact on
brain pathology, caused by an external force [1]. their families, relatives, caretakers, and social
TBI is considered a silent epidemic, giving rise to environment. The costs related to TBI are high,
many new victims each year, and represents a not only because of long-term hospitalization and
substantial socioeconomic and health problem. rehabilitation but also because of indirect costs
Worldwide, the incidence of TBI continues to resulting from loss of productivity in previously
increase, and the WHO predicts that road traffic healthy individuals due to mortality and disabil-
incidents will be among the top three causes of ity. The financial burden for TBI is estimated at
death and disability by the year 2020. In Europe, over 60 billion dollars a year in the USA [2].
approximately 2.5 million new cases of TBI are Despite its frequent occurrence and significant
diagnosed each year. The epidemiology is, how- personal and societal impact, our understanding
of TBI is still limited. The injured brain is like a
black box, from which it is difficult to extract
S. Sener, MD (*) • A.I.R. Maas information. Advanced MR Imaging, and in par-
Department of Neurosurgery, Antwerp University ticular Diffusion Tensor Imaging (DTI), offers
Hospital and University of Antwerp, opportunities to investigate this black box, and to
Wilrijkstraat 10, 2650 Edegem, Antwerp, Belgium
extract information that can be used to better
e-mail: suleyman.sener@uza.be
characterize damage, track disease processes,
P.M. Parizel, MD, PhD
and to establish a more accurate prognosis.
Department of Radiology, Antwerp University
Hospital and University of Antwerp, In the acute phase, structural brain damage is
Edegem, Antwerp, Belgium best assessed by CT scanning, as this is rapidly

© Springer Science+Business Media New York 2016 373


W. Van Hecke et al. (eds.), Diffusion Tensor Imaging, DOI 10.1007/978-1-4939-3118-7_19
374 S. Sener et al.

Fig. 19.1 TBI in a 57-year-old man, injured in a car acci- In addition, there is a small right parietal subdural hema-
dent. GCS on admission was 11/15 Noncontrast CT of the toma, but the midline structures are not displaced. The MRI
brain upon admission (a) and MRI of the brain on day 7 scan on day 7 reveals hemorrhagic contusions in the right
after the injury, including fat-saturated turbo FLAIR (b) temporal and right frontal lobes, surrounded by edema. On
and susceptibility weighted imaging (SWI) (c) were per- the SWI scan, the hemorrhagic nature of these contre-coup
formed. The coup side is indicated by a left parieto-occipital contusions is clearly seen, and there is evidence of multiple
subgaleal hematoma and infiltration of the subcutaneous other hemorrhagic foci in the brain parenchyma, as well as
fat (arrow). On the contre-coup side, there are hemor- small amounts of intraventricular blood in the trigone and
rhagic contusions in the right frontal and temporal lobes. occipital horns of the lateral ventricles

available and can be performed quickly. This is and existing contusions may increase in size in
important, as rapid detection and—if indicated— up to 40 % of cases. This lesion progression
prompt removal of an intracranial hematoma is mainly occurs within 6–9 h after injury [6].
one of the major principles upon which care for Follow-up imaging is therefore essential.
the TBI patient is based. CT scanning further per- The disadvantage of CT scanning is that it
mits a broad characterization of TBI by the only captures limited information on the full
Marshall CT classification [3]. This classification extent of structural damage and does not provide
offers a descriptive approach, which is important, any insight into function. Magnetic Resonance
as TBI is a very heterogeneous disease encom- (MR) imaging can provide better insight into the
passing a broad range of pathologies [4]. The extent and severity of primary and secondary
Rotterdam CT score was developed from a prog- brain damage (Fig. 19.1).
nostic perspective and combines different CT fea- Specific MRI sequences, i.e., susceptibility
tures into a sum score. This score has been shown weighted imaging (SWI) and diffusion weighted
to be more strongly related to outcome than the imaging (DWI), are more sensitive for detecting
Marshall CT classification [5]. Prediction of out- structural changes in the brain, particularly
come, however, is better performed by combining smaller lesions, such as microhemorrhages, dif-
different variables (clinical, radiological, and fuse axonal injury (DAI), and traumatic axonal
laboratory) into a multidimensional model. injury (TAI) (Fig. 19.2) [7]. Diffusion tensor
TBI is a dynamic process, and pathology imaging (DTI) provides valuable additional
evolves over time. In patients with contusions, information about white matter lesions and struc-
new lesions may develop in up to 16 % of cases tural damage, and is therefore particularly suited
19 Diffusion Tensor Imaging in Traumatic Brain Injury 375

Fig. 19.2 TBI in a 19-year-old man, injured in a motor prominent susceptibility artifact in the right frontal region
vehicle accident. The patient was admitted to the emer- is caused by the ventricular shunt. Five days later, on day
gency department with a GCS of 9/15. MR imaging was 12, the parenchymal lesions are unchanged, but there is a
performed on day 7 (a, d), day 12 (b, e), and after 6 slight decrease in the amount of intraventricular hemor-
months (c, f), using fat-saturated turbo FLAIR (a, b, c) rhage. After 6 months (c, f) the intracerebral lesions have
and susceptibility weighted imaging (SWI) (d, e, f). The regressed on the FLAIR image, but there remain multifo-
MRI scan on day 7 (a, d) reveals left frontal and bilateral cal, punctate microhemorrhagic foci on the SWI sequence.
capsulo-lenticular contusions, as well as intraventricular In addition, a mild cortical atrophy has occurred
hemorrhage and multiple scattered hemorrhagic foci. A

to characterize the presence and degree of injury cognitive impairment and disability after TBI and
and understand pathophysiological mechanisms thus a determining factor for outcome [11–18].
in neurotrauma. For example, in mild TBI patients with impaired
MR imaging can visualize abnormalities, con- executive function, reduced FA values were seen
sistent with axonal injury in up to 30–50 % of in the dorsolateral prefrontal cortex (DLPFC)
patients, in whom CT showed no apparent struc- [19]. A study by the NICER consortium demon-
tural damage (Fig. 19.3) [8–10]. It is important to strated that microstructural changes were observed
detect such lesions since TAI is a major cause of up to 5 years after severe TBI [20].
376 S. Sener et al.

Fig. 19.3 Mild TBI in a


35-years-old patient who
fell from the stairs, with
GCS of 14/15 (E4M6V4)
on admission. CT scan
(a) upon admission
showed no intracranial
abnormalities. MR
imaging—gradient echo T2
sequence—(b) 6 weeks
after initial injury shows
remnants of subarachnoid
hemorrhage in the left
parieto-temporal lobe
(arrow)

This chapter will elaborate on the role of dif- axonal transport mechanisms. Both hypo- and
fusion tensor imaging in traumatic brain injury hypermetabolism may occur at different stages
for research and clinical practice. after injury. Disturbances of the mitochondrial
transition pore are thought to be a main cause of
mitochondrial failure after TBI. Inflammatory
Role of DTI in TBI cascades are activated, some of which may be
protective, others—when in excess—detrimen-
Pathophysiology tal. Much insight into these mechanisms can be
gained from MR Imaging. DWI and apparent
The pathophysiology of TBI is highly complex diffusion coefficient (ADC) mapping can pro-
and involves multiple pathophysiologic pro- vide information about ischemia and edema,
cesses. Importantly, TBI should not be seen as whilst MR spectroscopy can provide insight into
an event, but as a progressive disease, in which metabolic derangements.
further damage may occur over hours, days,
weeks, months, or even years. Secondary dam-
age is preventable and potentially treatable. DTI in Diagnosis, Characterization,
Detection, quantification, and tracking of such and Classification of TBI
secondary damage is thus of paramount impor-
tance. Secondary damage may include brain DTI can provide important indirect information
swelling due to vascular engorgement or brain about neuronal integrity, continuity, and connec-
edema, which may be intracellular (cytotoxic) tivity of neural pathways, even when traditional
or vasogenic (extracellular). Cerebral ischemia, MRI sequences appear normal. The diffusion
considered one of the most common problems tensor characterizes the magnitude of water dif-
after TBI, can occur locally (in the penumbra of fusion (ADC and mean diffusivity, MD), its
a contusion) or more generalized and may be directional nonuniformity (fractional anisot-
exacerbated by systemic insults, such as low ropy, radial and axial diffusivity, FA, RD, AD),
blood pressure or inadequate blood oxygen- and its orientation (the tensor eigenvectors). The
ation. Traumatic axonal injury, formerly consid- exact mechanisms of the nature of water diffu-
ered a mechanical disruption of axons, has now sion in both grey and white matter are not com-
been shown to result from metabolic failure of pletely understood. However the organization of
19 Diffusion Tensor Imaging in Traumatic Brain Injury 377

tissue structure including the presence of myelin, severe TBI in both whole brain and ROIs, indica-
microtubules, and organelles, as well as the tive of potential myelin and axonal damage.
contribution of intra- and extracellular water Notably, differences in the effect of TBI sever-
(edema formation) play a significant role. This ity on AD and RD may reflect different underly-
is illustrated in a study by Newcombe et al. [21] ing pathophysiological mechanisms related to the
in subjects with moderate to severe head injury degree of tissue injury.
where MRI was performed at a median of 32 h Recently, Van der Eerden and the
after injury. Results showed a decreased FA in Neuroimaging for Coma Emergence and
the white matter, which was attributed predomi- Recovery (NICER) consortium [25] published
nantly to increased radial diffusivity, consistent the results of a multicenter study investigating
with edema. DTI changes in cardiac arrest and TBI patients,
In 2012, Shenton et al. published a review of using 19 predetermined ROIs. They found a
MRI and DTI focusing on mild traumatic brain decrease in axial diffusivity in cardiac arrest
injury. This article gives a good overview of stud- patients and increase in radial diffusivity in
ies performed on mild TBI, with all analyzed severe TBI patients. In the cardiac arrest patients,
data showing some degree of subtle brain dam- abnormalities are mostly seen in the cerebral
age in mild TBI. The authors noted a wide range hemispheres. For the TBI patients the abnormali-
of variability in DTI studies and difficulties in ties were found in both the central brain struc-
data interpretation, due to the use of different tures and cerebral hemispheres. The changes in
scanning protocols and timings at DTI. axial diffusivity could be related to primary axo-
A meta-analysis of 28 studies by Aoki et al. nal damage due to energy depletion caused by
[22] in mild TBI showed that the posterior part of ischemia. It was hypothesized that the increase in
the corpus callosum is more vulnerable to dam- radial diffusivity in TBI patients was related to
age compared to the anterior part. Compared to myelin damage and edema. The moderate
healthy volunteers, there was a significant decrease in axial diffusivity suggests axonal
decrease in FA and increase in mean diffusivity damage due to direct impact at trauma or isch-
in the corpus callosum in TBI patients. emic changes due to secondary mechanism
In a study by Rutgers et al. [23] on 21 patients caused by intracranial lesions. However, relating
with mild TBI, in whom MRI was performed at changes in the axial and radial diffusivities to
an average of 5.5 months after injury, the cerebral specific microstructural features is fraught with
lobar white matter showed regions with reduced challenges, particularly in the context of tissue
FA values in almost 62 % of the study popula- injury. As a consequence, these hypotheses for
tion. Further decrease of FA was observed in 23.6 the observed DTI changes remain tentative.
% of the patients in the cingulum or the corpus Kasahara et al. [26] performed DTI in healthy
callosum. Since abnormalities in the internal cap- controls and patients with mild TBI and
sule, fornix, brain stem, and cerebellum were not DAI. Decreased axonal and radial diffusivity was
frequently seen in their case study of mild TBI, found in the DAI patients. In mild TBI patients,
the authors suggest these lesions may be more normal radial diffusivity and increased axial dif-
associated with severe TBI. fusivity values were observed, indicating possi-
Kraus et al. [24] performed a DTI study in ble axonal abnormality.
chronic traumatic brain injury patients with mild,
moderate, and severe injury. The study included a
total of 20 patients with mild and 17 patients with DTI and Prognosis in TBI
moderate/severe TBI as well as 18 healthy con-
trols. DTI was performed at least 6 months after DTI may also be used as a prognostic predictor in
injury (average of 107 months after injury), using traumatic brain injury.
both whole brain analysis and region of interest A DTI voxel-based analysis by Perlbarg et al.
analysis. Results showed an increased radial and [27] investigating the prediction of 1-year out-
axial diffusivity in patients with moderate and come in severe TBI showed no significant ADC
378 S. Sener et al.

differences between the favorable and unfavor- axonal degeneration after injury, and can poten-
able outcome groups. However, significant tially serve as a prognostic factor and aid the
changes were observed in the FA values between surgeon and the treating physician in their
these two groups, with a decrease in the FA decision-making.
value in the unfavorable outcome group. Nevertheless, there are challenges, both in
Tollard and colleagues [28] performed a study interpreting literature data and in conducting
with DTI and magnetic resonance spectroscopy studies on patients following TBI. For example,
(MRS) for outcome prediction in severe trau- despite broad interest, most reports on DTI in
matic brain injury. Forty-three patients with TBI have been restricted to relatively small case
severe TBI and 15 control subjects underwent series, have included some selection bias, and
MRI with DTI and MRS sequences. The imaging have only rarely studied patients early after
was performed in the subacute phase after trauma injury. Furthermore, the multiple assumptions
(24 ± 11 days after injury). DTI was performed that underpin the algorithms used in post-
using symmetrical regions of interest (ROI) anal- processing of DTI data are not always applicable.
ysis in the left and right mesencephalon, tempo- Practical challenges associated with conducting
ral and occipital white matter, anterior and DTI studies in TBI populations include transpor-
posterior centrum semiovale, and anterior and tation, logistic issues concerning scanning, tim-
posterior part of the pons. ing of DTI scanning, and standardization of data
These regions were predetermined regardless acquisition and analysis protocols.
of the presence of morphologic lesions.
The results revealed significantly lower FA
values in patients with unfavorable outcome (i.e., Transportation and Scanning Issues
Glasgow Outcome Scale, GOS 1–3). Lower FA
values were also seen in the temporal white mat- In the acute phase of TBI, transport and scan-
ter and the centrum semiovale for patients with ning of a critically ill patient poses substantial
favorable outcome (i.e., GOS 4–5) compared to logistical challenges. Transportation and scan-
the control group. The authors suggest this may ning is thwarted by the need for mechanical
be because these lesions have no effect on the ventilation and continuous monitoring. Since
recovery of consciousness. these patients are treated on the intensive care
In this study, MRS was analyzed as single unit, transportation itself may carry risks. For
voxel spectroscopy in the posterior pons and instance, the intra-hospital transportation of
axial chemical shift imaging (CSI) at the basal critically ill patients increases the risk for sig-
ganglia. Interestingly, using DTI and MRS sepa- nificant adverse events, such as inadvertent
rately showed lower specificity (85 % and 75 % hyperventilation or hypotension with limited
respectively) and sensitivity (79 % and 75 % options to intervene in case of problems [29]. In
respectively) for predicting unfavorable outcome addition, installation of an ICU patient on a
at 1-year post-injury, than combining the tech- MRI scan is time consuming. Scanning time for
niques (97 % specificity and 86 % sensitivity). a brain MRI, including DWI, SWI, and DTI, is
This highlights the added value of combining also significantly longer compared to a CT scan
DTI with other imaging modalities to improve its (45 min vs. 2 min).
clinical utility. A major cause for disruption of DTI analysis
is motion artifacts, resulting in loss of signal.
Correction of the motion artifacts however is
Challenges for the Use of DTI in TBI complex, but feasible using various methods [30].
Since TBI patients may have been operated
DTI is well suited to provide information related on, any surgical material such as titanium clips,
to the continuity and connectivity of neuronal fixation materials, hemoclips, and surgical sta-
pathways. It can provide insight into micro- ples can cause loss of signal and will affect DTI
structural changes, such as fiber disruption and analysis.
19 Diffusion Tensor Imaging in Traumatic Brain Injury 379

Timing of DTI Scanning before DTI will be a standard scanning sequence


in clinical practice for TBI.
Since we deal with an evolving pathology, it is Standardization is the most difficult issue in
important to determine when to perform a MRI the use of DTI for TBI since analyzing DTI is
scan after TBI. As explained earlier, TBI is a complex. Furthermore, studies of larger numbers
dynamic process and both primary and secondary of patients and multicenter studies that incorpo-
brain damage will cause brain swelling. In the rate DTI are needed to evaluate the relevance of
acute phase after TBI, DTI can be influenced by DTI as a diagnostic and prognostic tool for TBI
edema, which imitates fiber disruption [23], and in the acute and chronic phase. Nevertheless, DTI
by large intracranial lesions. In the subacute holds great promise to better characterize dam-
phase, neuronal degeneration and inflammatory age, track disease processes, and establish a more
reactions due to secondary brain damage are still accurate prognosis in patients with TBI.
ongoing. Valuable information can be missed by
DTI scanning during this period.
References

Standardization of DTI Analysis— 1. Menon DK, Schwab K, Maas AIR. Demographics


and Clinical Assessment Working Group of the inter-
Analyzing DTI national and interagency initiative toward common
data elements for Research on Traumatic Brain Injury
Two major approaches for analyzing DTI in TBI and Psychological Health; Position statement: defini-
exist: (1) region of interest (ROI) analysis and (2) tion of traumatic brain injury. Arch Phys Med Rehabil.
2010;91(11):1637–40.
whole brain (WB) tractography.
2. Finkelstein EA, Corso PS, Miller TR. The incidence
In the region of interest approach, analyses are and economic burden of injuries in the United States.
performed in predetermined regions, generally New York, NY, USA: Oxford University Press; 2006.
chosen within the white matter (because of inter- Xiii. p. 187.
3. Marshall LF, Bowers Marshall S, Klauber MR, Van
est in axonal disruption and water content/diffu-
Berkum Clark M, Eisenberg HM, Jane JA, Luerssen
sion). This gives detailed analysis in these TG, Marmarou A, Foulkes MA. A new classification
selected ROIs, but risks possibly missing areas of of head injury based on computerised tomography.
decreased FA values outside of these regions. Spec Suppl. 1991;75(1s):S14–20.
4. Saatman KE, Duhaime A-C, Bullock R, Maas AIR,
TBI is a very heterogeneous group and with
Valadka A, Manley GT. Workshop scientific team and
WB tractography a more complete overview can advisory panel members. Classification of traumatic
be achieved. Nevertheless, artifacts such as large brain injury for targeted therapies. J Neurotrauma.
intracranial lesions, which can influence DTI 2008;25:719–39.
5. Maas AIR, Hukkelhoven CWPM, Marshall LF,
analysis, may hinder this technique. In addition,
Steyerberg EW. Prediction of outcome in traumatic
specific regional information is lost by using brain injury with computed tomographic characteris-
whole brain tractography results. tics: a comparison between the computed tomographic
It is the combination of all these challenges, i.e., classification and combinations of computed tomo-
graphic predictors. Neurosurgery. 2005;57:1173–82.
problems due to transportation, timing in DTI scan-
6. Sener S, Rozenbeek B, Maas AIR. Surgical manage-
ning, specialized software, and different methods ment of traumatic brain injury—evidences, controver-
in analysis, that make standardization of the anal- sies and perspectives for the futures. Eur Neurol Rev.
ysis one of the most difficult issues to overcome 2011;6(3):196–201.
7. Shenton ME, Hamoda HM, Schneiderman JS, Bouix S,
when using TBI to assess traumatic brain injury.
Pasternak O, Rathi Y, Vu M-A, Purohit MP, Helmer K,
Koerte I, Lin AP, Kikinis R, Kubicki M, Stern RA,
Zafonte R. A review of magnetic resonance imaging
Conclusion and diffusion tensor imaging findings in mild traumatic
brain injury. Brain Imaging Behav. 2012;6:137–92.
8. Lee B, Newberg A. Neuroimaging in traumatic brain
DTI remains for now mostly a research tool. Even imaging. NeuroRx. 2005;2:372–83.
though various studies have yielded promising 9. Mittle RL, Grossman RI, Hiehle JF, Hurst RW,
findings, there are still difficulties to overcome Kauder DR, Gennarelli RA, et al. Prevalence of MR
380 S. Sener et al.

evidence of diffuse axonal injury in patients with mild in mild traumatic brain injury: a diffusion tensor imag-
head injury and normal head CT findings. Am ing study. Am J Neuroradiol. 2008;29:514–9.
J Neuroradiol. 1994;15:1583–9. 24. Kraus MF, Susmaras T, Caughlin BP, Walker CJ,
10. Hammoud DA, Wasserman BA. Diffuse axonal inju- Weeney JA, Little DM. White matter integrity and
ries: pathophysiology and imaging. Neuroimaging cognition in chronic traumatic brain injury: a diffu-
Clin N Am. 2002;12(2):205–16. sion tensor imaging study. Brain. 2007;130:2508–19.
11. Ezaki Y, Tsutsumi K, Morikawa M, Nagata I. Role of 25. Van der Eerden AW, Khalilzadeh O, Perlbarg V,
diffusion-weighted magnetic resonance imaging in Dinkel J, Sanchez P, Vos PE, Luyt C-E, Stevens RD,
diffuse axonal injury. Acta Radiol. 2006;47:733–40. Menjot de Champfleur N, Delmaire C, Tollard E,
12. Kelly AB, Zimmerman RD, Snow RB, Gandy SE, Gupta R, Dormont D, Laureys S, Benali H,
Heier LA, Deck MDF. Head trauma: comparison of Vanhaudenhuyse A, Galanaud D, Puybasset L. For
MR and CT—experience in 100 patients. AJNR Am NICER (Neuro Imaging for Coma Emergence and
J Neuroradiol. 1988;9(4):699–708. Recovery) Consortium. White matter changes in
13. Murray JG, Gean AD, Evans SJ. Imaging of acute head comatose survivors of anoxic ischemic encephalopa-
injury. Semin Ultrasound CT MR. 1996;17:185–205. thy and traumatic brain injury: comparative diffusion-
14. Shaefer PW, Huisman TAGM, Sorensen AG, tensor imaging study. Radiology. 2014;270:506–16.
Gonzalez RG, Schwamm LH. Diffusion weighted 26. Kasahara K, Hashimoto K, Abo M, Senoo A. Voxel-
MR Imaging in closed head injury: high correlation and atlas-based analysis of diffusion tensor imaging
with initial Glasgow Coma Scale score and score on may reveal focal axonal injuries in mild traumatic
Modified Rankin scale at discharge. Radiology. brain injury—comparison with diffuse axonal injury.
2004;233(1):58–66. Magn Reson Imaging. 2012;30:496–505.
15. Smith DH, Meaney DF, Shull WH. Diffuse axonal 27. Perlbarg V, Puybasset L, Tollard E, Lehéricy S, Benali
injury in head trauma. J Head Trauma Rehabil. H, Galanaud D. Relation between brain lesion loca-
2003;18:307–16. tion and clinical outcome in patients with severe trau-
16. Scheid R, Walther K, Guthke T, Preul C, von Cramon matic brain injury: a diffusion tensor imaging study
Y. Cognitive sequelae of diffuse axonal injury. Arch using voxel-based approaches. Hum Brain Mapp.
Neurol. 2006;63:418–24. 2009;30:3924–33.
17. Skandsen T, Kvistad KA, Solheim O, Haavde Strand 28. Tollard E, Galanaud D, Perlbarg V, Sancez-Pena P, Le
I, Folvik M, Vik A. Prevalence and impact of diffuse Fur Y, Abdennour L, Cozzone P, Lehericy S, Chiras J,
axonal injury in patients with moderate and severe Puybasset L. Experience of diffusion tensor imaging
head injury: a cohort study of early magnetic reso- and 1H spectroscopy for outcome prediction in severe
nance imaging findings and 1-year outcome. traumatic brain injury: preliminary results. Crit Care
J Neurosurg. 2010;113:556–63. Med. 2009;37:1448–55.
18. Chelly H, Chaari A, Daoud E, Dammak H, Medhioub 29. Parmentier-Decrucq E, Poissy J, Favory R, Nseir S,
F, Mnif J, Ben Hamida C, Bahloul M, Bouaziz Onimus T, Guerry M-J, Durocher A, Mathieu
M. Diffuse axonal injury in patients with head inju- D. Adverse events during intrahospital transport of
ries: an epidemiologic and prognosis study of 124 critically ill patients: incidence and risk factors. Ann
cases. J Trauma. 2011;71:838–46. Intensive Care. 2013;3:10.
19. Lipton ML, Gulko E, Zimmerman ME, Friedman 30. Le Bihan D, Poupon C, Amadon A, Lethimonnier
BW, Kim M, Gellella E, Gold T, Shifteh K, Ardekani F. Artefacts and pitfalls in diffusion MRI. J Magn
BA, Branch CA. Diffusion-tensor imaging implicates Reson Imaging. 2006;24:478–88.
prefrontal axonal injury in executive function impair-
ment following very mild traumatic brain injury.
Radiology. 2009;252(3):816–24.
20. Dinkel J, Drier A, Khalilzadeh O, Perlbarg V,
Czernecki V, Gupta R, Gomas F, Sanchze P, Dormont
Suggested Reading
D, Galanaud D, Stevens RD, Puybasset L. for NICER
(Neuro Imaging for Coma Emergence and Recovery) Yeh P-H, Oakes TR, Riedy G. Diffusion tensor imaging
Consortium. Long-term white matter changes after and its application to traumatic brain injury: basic
severe traumatic brain injury: a 5-year prospective principles and recent advances. Open J Med Imaging.
cohort. Am J Neuroradiol. 2014;35:23–9. 2012;2:137–61.
21. Newcombe VF, Williams GB, Nortje J, et al. Analysis Hulkower MB, Poliak DB, Rosenbaum SB, Zimmerman
of acute traumatic axonal injury using diffusion tensor ME, Lipton ML. A decade of DTI in traumatic brain
imaging. Br J Neurosurg. 2007;21(4):340–8. injury: 10 years and 100 articles later. AJNR Am
22. Aoki Y, Inokuchi R, Gunshin M, Yahagi N, Suwa J Neuroradiol. 2013;34(11):2064–74.
H. Diffusion tensor imaging studies of mild traumatic Xiong K, Zhu Y, Zhang W. Diffusion tensor imaging and
brain injury: a meta-analysis. J Neurol Neurosurg magnetic resonance spectroscopy in traumatic brain
Psychiatry. 2012;83:870–6. injury: a review of recent literature. Brain Imaging
23. Rutgers DR, Toulgoat F, Cazejust J, Fillard P, Behav. 2014;8(4):487–96. doi:10.1007/s11682-013-9288-2
Lasjaunias P, Ducreaux D. White matter abnormalities [Epub ahead of print].
High Angular Resolution Diffusion
Imaging 20
Shawna Farquharson
and Jacques-Donald Tournier

• HARDI data is characterized primarily by two


Learning Points parameters: the number of unique directions,
and the b-value. HARDI acquisition strategies
• The fundamental limitations of the Diffusion are essentially identical in nature to the stan-
Tensor (DTI) model have important implica- dard DTI acquisition, and differ only in that a
tions for the interpretation of anisotropy as a larger number of unique diffusion-weighting
marker of white matter integrity, and for appli- gradient directions are used, potentially using
cations such as diffusion-based tractography. a larger b-value than would be considered
• Over the past decade, more advanced models optimal for DTI.
have been developed to specifically address • In practice, the number of directions required
the limitations of the DTI model, many of for HARDI is difficult to ascertain exactly
which are based on the high angular resolu- since increasing it will always improve results
tion diffusion-weighted imaging (HARDI) through increasing the overall SNR of the
data acquisition strategy. acquisition. The primary requirement of the
• These HARDI-based approaches can be com- angular sampling is to characterize the relevant
bined with appropriate tracking algorithms to features of the DW signal. In practice, the opti-
provide improved tractography results. mal set of parameters will need to be deter-
mined empirically on a case-by-case basis,
based on the intended HARDI reconstruction
method and its particular requirements.
• HARDI-based reconstruction methods are
now beginning to be used for clinical and neu-
S. Farquharson, MSc
Florey Institute of Neuroscience and Mental Health, roscientific investigations with extremely
University of Melbourne, 245 Burgundy Street, promising results.
Heidelberg, VIC 3084, Australia
J.-D. Tournier, PhD (*)
Florey Institute of Neuroscience and Mental Health,
University of Melbourne, 245 Burgundy Street,
Heidelberg, VIC 3084, Australia
Why We Need to Move Beyond DTI
Biomedical Engineering, Division of Imaging
The development of diffusion tensor imaging
Sciences & Biomedical Engineering, King’s College
London, London, UK (DTI) and quantitative parameters derived from
e-mail: jacques_donald.tournier@kcl.ac.uk the diffusion tensor over the past two decades

© Springer Science+Business Media New York 2016 383


W. Van Hecke et al. (eds.), Diffusion Tensor Imaging, DOI 10.1007/978-1-4939-3118-7_20
384 S. Farquharson and J.-D. Tournier

was a pivotal moment in the field of diffusion The principal orientation of the tensor ellipsoid
Magnetic Resonance Imaging (MRI). Although within each voxel is used as an estimate of the
DTI is still the most commonly used model to fiber orientation, and forms the basis of many
relate the diffusion-weighted MRI signal to the diffusion-based tractography methods [2, 3].
underlying diffusion process, it is now widely The fundamental limitation of the DTI model
acknowledged to be inadequate for this purpose. is that the tensor framework assumes a single
The limitations of the DTI model have important straight fiber orientation within each imaging
implications for the interpretation of anisotropy voxel, and is hence inadequate for the purpose of
as a marker of white matter integrity and for describing diffusion data within voxels contain-
applications such as diffusion-based tractogra- ing complex fiber configurations or multiple fiber
phy. In this chapter we discuss the practical limi- populations [1–4], an issue commonly referred to
tations of the DTI model and review recent as the “crossing fiber problem.” The so-called
developments in the field, many of which are crossing fiber problem refers to any configuration
based on the high angular resolution diffusion- of white matter fibers within a given imaging
weighted imaging (HARDI) data acquisition voxel that is more complex than a single straight
strategy. fiber population, including fanning, bending,
diverging and crossing fibers [5–7], as illustrated
in Fig. 20.1. It is well known in the technical dif-
Key Points About DTI fusion MRI community that using DTI to model
complex fiber orientations results in an incorrect
The physical basis of the DTI model is that if characterization of the constituent fiber popula-
water molecules encounter highly oriented barri- tions (e.g., [1–4, 7–11]), as illustrated in Fig.
ers, then the distance they travel (from a given 20.2. Given that diffusion-weighted data acquired
point for a given time) is on average greater along on clinical scanners are typically limited to a spa-
the structure than across the structure. This intro- tial resolution of 2–3 mm and the relatively small
duces a dependence of the measured diffusion size of most white matter tracts, it is not surpris-
signal on the direction along which it is mea- ing that the so-called “crossing fiber problem”
sured, a phenomenon commonly referred to as remains a significant issue for DTI.
anisotropy [1]. The diffusion tensor ellipsoid is The fact that the diffusion tensor is affected by
used to describe in three dimensions the degree the crossing fiber problem has been known since
of anisotropy within individual voxels, and the its very invention [12]; however, it is only recently
principal axes of this ellipsoid (or principal that the scale of this problem has been fully
eigenvectors) represent the orientation of the appreciated. Recent work demonstrates that mul-
constituent tissue. Scalar measures derived from tiple fiber populations can be detected in over 90
the diffusion tensor—for example the fractional % of imaging voxels in the white matter [13].
anisotropy (FA) and mean diffusivity (MD)—are The scale of this problem has obvious and pro-
commonly used as measures of “tissue integrity.” found implications for both DTI-based tractogra-

Fig. 20.1 The term “crossing fibers” can refer to any sit- also voxels where the fibers themselves are curved, such
uation where the fiber orientation is not unique. This as curving or diverging configurations (right), or any com-
includes obvious cases where the voxel contains two fiber bination of these two (middle)
bundles that cross or pass close to each other (left), but
20 High Angular Resolution Diffusion Imaging 385

Fig. 20.2 A simple illustration of the effect of crossing not in general be aligned with either of the populations
fibers in diffusion tensor imaging. The diffusion tensor present, and the anisotropy is also reduced. On the other
model is a good fit for voxels that contain a single fiber hand, the DW signal itself (bottom row) clearly does con-
orientation (left and middle columns), but fails to capture tain higher-order information in crossing fiber voxels
the orientation information when two distinct orientations (bottom right), which can be used to resolve the different
are present (right); in this case, the major eigenvector will fiber orientations present

phy techniques and DTI-derived measures used investigated the feasibility of DTI-based tractog-
to assess tissue integrity. raphy for the purpose of visualizing tracts of neu-
The impact of crossing fibers has important rosurgical interest demonstrate that these methods
ramifications for the application of tractography result in systematically unreliable and clinically
in particular, as the DTI model provides incorrect misleading tractography information [17, 19].
fiber orientation estimates in regions containing The extent of the crossing fiber problem
complex fiber configurations (see Fig. 20.2) [1]. means that caution also needs to be observed
This has important practical implications, as the when interpreting DTI-derived diffusion indices
majority of white matter tracts will traverse such as fractional anisotropy (FA), particularly if
regions with multiple fiber orientations at some the intention is to use them as surrogate markers
point along their path. Even a single incorrect ori- of white matter “integrity” [11, 21–23]. It is well
entation estimate (or for that matter, even a rela- known that FA values vary greatly over the white
tively small error in orientation estimation) will matter even in healthy controls where there is no
be enough to cause the algorithm to veer off- known change in white matter integrity. Such
course or follow a completely unrelated tract, variations are likely due to the presence of cross-
resulting in the delineation of false negative [13, ing fibers in different parts of the white matter
14] or false positive delineation of white matter leading to reductions in tensor-derived anisot-
pathways [13, 15]. The severe limitations of ropy, as was originally suggested in the early DTI
using the diffusion tensor model for fiber tractog- literature [24]. The profound confounding influ-
raphy have consistently been reported in the neu- ence of crossing fibers on the interpretation of
rosurgical literature [16–20]. Of particular DTI-derived diffusion indices in the presence of
concern is that studies that have specifically pathology is demonstrated in clinical studies that
386 S. Farquharson and J.-D. Tournier

have observed an increase in anisotropy in cases HARDI Methods


where the given pathology or condition would
have been expected to result in a reduction in tis- What Is HARDI?
sue integrity [25]. The diffusion tensor model is
increasingly recognized to be a gross The term HARDI stands for high angular resolu-
over-simplification of the actual anatomy and the tion diffusion imaging, and was originally coined
simplistic interpretation of DTI derived indices by Tuch et al. to refer to the particular acquisition
as a marker of white matter “integrity” is fraught strategy employed in their study, namely the use
with technical challenges [22, 23, 26]. As a con- of dense sampling on the sphere using a single
sequence, there is a growing interest in finding b-value, as illustrated in Fig. 20.3 [4]. Data
clinically feasible alternatives to DTI [27]. acquired in such a way allow the thorough char-
Over the past decade, more advanced models acterization of the angular dependence on the
have been developed to specifically address the DW signal, with no attempt at characterizing its
limitations of the DTI model ([5–7], see reviews radial (b-value) dependence. For this reason, it is
in [8]), many of which are now being used clini- arguably the most efficient acquisition strategy
cally with promising results. In this section, we for the purpose of fiber orientation estimation,
describe some of the concepts and theory behind and hence for tractography.
approaches that are based on high angular resolu- The HARDI acquisition is essentially identi-
tion diffusion-weighted imaging (HARDI) data cal in nature to the standard DTI acquisition, and
[4], developed specifically to provide more robust differs only in that a larger number of unique
fiber orientation estimates for applications such diffusion-weighting gradient directions are used,
as diffusion tractography, and more recently to potentially using a larger b-value than would be
provide more biologically accurate measures of considered optimal for DTI. There is in fact no
tissue microstructure or “integrity.” clear point at which an acquisition strategy

Fig. 20.3 The motivation for HARDI is the need to cap- tical, using a suitable set of DW directions such as those
ture all the features of the DW signal over the sphere. For shown on the left. By measuring the DW signal along
example, the DW signal shown on the right clearly con- these orientations (middle), the DW signal can be recon-
tains angular features, and these features are key to resolv- structed by fitting a surface (right), so that these features
ing crossing fibers. The idea behind HARDI is to sample can be estimated accurately
the orientation space as densely and uniformly as is prac-
20 High Angular Resolution Diffusion Imaging 387

becomes a HARDI sequence or vice-versa. For there is structure present in this case that the ten-
example, the current recommended minimum sor ellipsoid fails to capture. Indeed, the DW sig-
number of directions for robust DTI is 30 [28], nal for the crossing fiber case is to a very good
while higher-order reconstruction methods have approximation the sum of the DW signals that
been applied to data acquired using as few as 20 would be measured for each population indepen-
directions [29]. Nonetheless, a sequence will typ- dently; even by eye, the two contributions can
ically be considered HARDI if the number of easily be appreciated in the combined DW signal.
directions is relatively large (>~40). The aim of HARDI methods is essentially to sep-
It is worth noting that HARDI is not in itself a arate out these two contributions, and thus to
method for estimating fiber orientations; it refers resolve crossing fibers. For this to be possible,
specifically to the acquisition strategy. This can the relevant features of the DW signal need to be
lead to confusion in the literature when authors captured with sufficient accuracy. This requires a
claim to “perform a HARDI reconstruction”— larger number of DW directions than typically
this statement is ambiguous since it makes no acquired in DTI, leading directly to the develop-
mention of the reconstruction method used: it is ment of HARDI approaches.
in fact perfectly possible to perform a DTI recon- The problem of separating the two contribu-
struction using HARDI data. Nonetheless, tions can in essence be written out as a set of
HARDI is the most common data acquisition simultaneous equations: M DW measurements
strategy required for methods that aim to resolve are acquired per voxel, and from these a set of N
crossing fibers, and for this reason the term model parameters need to be estimated. What
HARDI is often used to refer to more advanced these N parameters represent is dependent on the
higher-order methods. particular reconstruction approach used: they
might represent the orientations and volume frac-
tions of a fixed number of potential fiber popula-
Why HARDI? tions, or the coefficients of a more continuous
representation of the diffusion or fiber configura-
The simplest and most intuitive approach to tion, as illustrated in Fig. 20.4. The latter is par-
understanding HARDI is to consider the example ticularly advantageous from a mathematical
shown in Fig. 20.2, with two fiber populations perspective, since describing the orientation
crossing within a voxel. In this example, it is easy information using a continuous distribution
to see that DTI provides a good characterization allows the problem to be expressed and solved
of each of the two fiber populations separately, using extremely fast linear algebra techniques,
but fails when the two are combined. However, if with reconstruction times on the order of seconds
we focus on the DW signal itself, it is clear that for whole-brain datasets. Using a more explicit

Fig. 20.4 The simplest approach to modelling crossing orientations with associated volume fractions (left); this is
fibers in HARDI is to assume that the DW signals from approach used in multi-tensor fitting methods. Another
the various fiber populations add up to give the total mea- approach is to represent the fiber configuration as a con-
sured DW signal for that voxel (middle left). Resolving tinuous distribution, which can be approximated by a
these crossing fibers is then a matter of finding the fiber dense set of fixed orientations and corresponding volume
configuration that best matches the observed data. One fractions (right); this is the approach used in spherical
way to represent this configuration is as a set of discrete deconvolution methods
388 S. Farquharson and J.-D. Tournier

parametric representation (for example partial their diffusivities constant (i.e., assume a fixed
volume and orientation per fiber population) does mean diffusivity and anisotropy). Nonetheless, a
not lend itself to such convenient linear number of methods are based on this general
approaches, and will in general necessitate the approach; some allow for larger numbers of fiber
use of more computationally expensive nonlinear populations, some include an isotropic “CSF”
optimization methods. compartment, and some use advanced Bayesian
Monte Carlo Markhov Chain (MCMC) sampling
techniques to characterize the uncertainty about
Using HARDI to Resolve Crossing the parameters estimates [14, 30–32].
Fibers The Combined Hindered and Restricted
Model of Diffusion (CHARMED)approach is
By far the most common application for HARDI strongly related to the multi-tensor approach in
methods is the estimation of the fiber orientations that it also models each fiber population indepen-
for use in fiber tracking. A number of methods dently [33, 34]. However, it differs substantially
are available to achieve this, but all are based on in that it assumes a more biologically plausible
the same concept: each fiber population (i.e., dis- model of restricted diffusion in cylinders to rep-
tinct orientation) adds its own contribution to the resent the DW signal from each fiber populations,
measured signal, so that the total measured signal rather than a diffusion tensor model. It also
is the sum of the contributions that would have includes an extracellular compartment, itself
been measured for each bundle in isolation. modeled by a diffusion tensor. One limitation is
that it requires a more demanding multi-shell
Multi-Tensor Fitting (i.e., multiple b-values) HARDI data acquisition,
Based on this observation, the most obvious leading to lengthier scan times.
approach to resolving crossing fibers is the multi- One issue with these methods is that they
tensor approach: we assume the voxel contains somehow need to know how many fiber popula-
two fiber populations rather than one, with each tions to include in the model for each voxel. While
population modeled using its own diffusion ten- a number of approaches have been proposed to do
sor. In this case, the minimum additional infor- this [2, 14, 31], any errors in this number will
mation required is the relative amounts of each inevitably lead to errors in the estimated parame-
fiber population (i.e., their volume fractions) and ters. Most of these approaches are based on some
their respective orientations. The problem is then statistical “goodness of fit” measure, and this will
one of estimating the set of parameters (volume in general lead to underestimation of the number
fractions and orientations) that best fit the mea- of fiber populations present, particularly in noisy
sured data [4, 30]. data where any improvements in the model fit will
It turns out that this is not a simple problem to be overwhelmed by the noise. The result is that
solve in practice due to the nonlinear formulation while the algorithm can be said to be “conserva-
of the problem (a sum of exponentials), dictating tive” in that it only includes additional fiber popu-
the use of computationally expensive nonlinear lations if there is good evidence that these are
minimization methods. In addition, the problem is needed, it will inevitably fail to identify a signifi-
generally ill-conditioned, due to the difficulty of cant proportion of crossing fiber voxels and there-
differentiating between changes in anisotropy and fore model them using the single tensor model,
changes in volume fraction of the constituent fiber with all the limitations highlighted above.
populations1; to get around this, most implementa-
tions enforce the condition that each diffusion ten- Spherical Deconvolution
sor be at least axially symmetric, and often hold all The multi-tensor approach can be extended by
switching from a discrete representation (i.e., a
1
This is discussed in more detail in the “constant anisot- small number of distinct fiber populations) to a
ropy” assumption section below. continuous representation of the fiber orientation
20 High Angular Resolution Diffusion Imaging 389

information. One way to understand this is to directions that might be present in each voxel; the
imagine that rather than trying to estimate the FOD is continuous, and distinct fiber orientations
volume fraction and orientation of each fiber will simply be reflected as distinct peaks in the
population, we are now going to model a large FOD. Finally, the FOD is suited to describing
number of fiber populations, each with a fixed fiber configurations containing a range of orien-
orientation. The problem now reduces to figuring tations, such as for example bending or fanning
out the volume fractions of each of these. If we fibers; these are clearly not well characterized
use a sufficiently dense set of orientations that are using a single orientation, as would be used in
uniformly distributed over the sphere, the infor- multi-tensor approaches. In these cases, the cor-
mation can be represented as a distribution of responding peak in the FOD will simply be
volume fractions over the sphere, as illustrated in broadened accordingly, as expected. Spherical
Fig. 20.4. This distribution is commonly referred deconvolution approaches are therefore more
to as the fiber orientation density function (fODF) computationally efficient, while providing a
or simply the fiber orientation distribution (FOD). much more general description of the fiber orien-
Essentially, the FOD is a function defined over tation information. These provide estimates of
the sphere, which represents the amount of fibers the FOD that are suitable for fiber-tracking, as
at any point on the sphere (i.e., aligned with the illustrated for example in Fig. 20.6.
corresponding orientation). It is often displayed A number of different spherical deconvolution
using orientation plots such as those shown in techniques have been proposed [10, 29, 36–39].
Fig. 20.5, whereby the distance from the origin The most stable of these impose a non-negativity
represents the “density” along the corresponding constraint to explicitly prevent or minimize nega-
orientation. Distinct fiber orientations can clearly tive fiber density values, which are clearly non-
be seen as distinct peaks in the FOD. physical; this has been shown to dramatically
The advantages of using such a distribution to improve the robustness to noise of these methods
represent the fiber orientation information are [29, 37]. Some estimate the per-fiber anisotropy
threefold. First, it means that the relationship on a per-voxel basis [36], other assume fixed dif-
between the measured DW signal and the FOD is fusivity values [37–39], while others bypass the
linear, allowing the use of much more efficient diffusion tensor model entirely and measure the
reconstruction methods based on linear algebra. actual per-fiber DW signal profile from the data
Second, there is no need to specify the number of themselves [10, 29].

Fig. 20.5 Fiber orientation distributions obtained from a the centre with fibers running left–right (red lobes). Its
healthy volunteer, estimated using constrained spherical lateral projections can be seen to cross through the fibers
deconvolution [29] as implemented in MRtrix [35]. These of the corona radiata (running inferior–superior; blue
are shown as a coronal projection overlaid on the mean lobes) and the superior longitudinal fasciculus (running
DWI image, with the corpus callosum clearly visible in anterior–posterior; green lobes)
390 S. Farquharson and J.-D. Tournier

Fig. 20.6 Whole-brain fiber-tracking results obtained dinal fasciculus can be seen running anterior–posterior
from a healthy volunteer, generated using a probabilis- (green) and then down into temporal and parietal
tic fiber tracking algorithm on orientations estimated regions. Also visible are the uncinate fasciculus, con-
using constrained spherical deconvolution [29] as necting the temporal pole to the frontal lobes (blue), and
implemented in MRtrix [35]. A 2 mm-thick section the inferior fronto-occipital fasciculus running anterior–
through the tracks is shown as a sagittal projection. posterior (green), connecting the frontal lobe to the
Amongst other white matter tracts, the superior longitu- occipital lobe

The “Constant Anisotropy” Assumption ing fiber effects or other partial volume effects
The overwhelming majority of multi-tensor fit- (e.g., with CSF). This is in fact in line with the
ting and spherical deconvolution techniques will original DTI literature, where the large variations
make the assumption that all fiber bundles can be of anisotropy observed in healthy volunteers
described by diffusion tensors with the same were attributed to differences in the coherence of
intrinsic “constant anisotropy” (or at least axial fiber tract directions [24]. The relationship
symmetry) to reduce the complexity of the prob- between anisotropy (and more recently radial and
lem. At first sight, this might seem to be a gross axial diffusivities) and white matter “integrity”
over-simplification, particular to those steeped in was suggested based on highly controlled single
the more traditional DTI framework, where nerve experiments, where any confounding
tensor-derived metrics are often used as surrogate effects of crossing fibers could explicitly be ruled
markers of white matter “integrity.” Since these out [40–44]. While these studies undeniably
new approaches assume fixed values for these demonstrated changes in various tensor-derived
metrics over all white matter bundles, it is now metrics under different conditions (dysmyelin-
impossible to estimate equivalent measures, ation, axonal injury, changes in axonal diameter,
let alone detect differences between them. etc.), these changes are dwarfed by the changes
However, there are very good reasons to induced by the presence of crossing fibers [1, 23].
believe that this approximation holds in most, if Moreover, and more importantly, while changes
not all cases. The assumption of fixed anisotropy in diffusivities and/or anisotropy can be observed
inherently implies that all observed anisotropy in these experiments, they are not of a magnitude
differences in the brain are due entirely to cross- sufficient to alter the results dramatically; as
20 High Angular Resolution Diffusion Imaging 391

shown in previous studies, while moderate extracting specific microstructural information


changes in the assumed anisotropy will affect the will inevitably require that assumptions and
estimated volume fractions, they have almost no approximations be made. Rather than trying to do
impact on the estimated fiber orientations [10]. In this, it might be best to characterize what we can,
fact, any pathology that would cause severe and avoid making any potentially invalid or unjus-
changes in the anisotropy of the per-fiber DW tified assumptions. For this reason, a number of
signal would probably need to involve destruc- so-called model-free techniques have been pro-
tion of the axonal membrane, with a large posed to characterize the diffusion process itself.
increase in radial diffusivity and corresponding
reduction in the DW signal; this will be inter- q-Space and the Spin Propagator
preted by these models primarily as a dramatic These methods are all in some way based on the
decrease in volume fraction, an interpretation theory of q-space [48], which describes the rela-
that is in fact fairly accurate given the pathology. tionship between the DW signal and the so-called
For this reason, this particular assumption is averagespin propagator (also called the spin dis-
very commonly employed, and with great success. placement probability density function). In sim-
However, the implication is that genuine anisot- ple terms, the spin propagator is a function
ropy changes are at best extremely difficult to describing the probability P(r|x,Δ) that a water
detect from HARDI data (given that they are molecule initially at x has moved by displace-
essentially indistinguishable from partial volume ment r during the diffusion time Δ of the MR
effects), which essentially rules out the possibility measurement. For free diffusion, this is simply a
of deriving equivalent markers of white matter Gaussian distribution with standard deviation
“integrity”—at least using these approaches. equal to the root mean square displacement of the
Instead, the current trend is to focus on the esti- water molecules (as per Chap. 3). For more com-
mated partial volume fractions, since these seem plex environments, the propagator will have a
to have the most dominant impact on the DW sig- more complex shape, as illustrated in Fig. 20.7.
nal, and can be used as estimates of fiber density— This makes no assumption about whether diffu-
a measure that, although different from the more sion is free or restricted: it simply characterizes
common interpretation of “integrity,” is nonethe- the chances of molecules moving along a given
less clearly clinically very relevant [45–47]. direction by a given amount. In general, this will
depend on the diffusion time Δ: with free diffu-
sion, the distances moved will increase with dif-
Using HARDI to Characterize fusion time. However, in restricted geometries
Diffusion the maximum distance moved will clearly be dic-
tated by the size of the restricting compartment.
Another approach to the crossing fiber problem is The relationship between the DW signal and the
to take a step back and focus on characterizing the spin propagator is relatively simple and not sub-
diffusion process itself, rather than the fiber con- ject to any particular biological assumptions.2
figuration directly. The motivation for such Clearly, the diffusion propagator provides the
approaches is to avoid over-interpretation of the most complete and accurate description of the
measured data. The relationship between the mea- diffusion process, and contains all the informa-
sured DW signal and the actual fiber configuration tion that can be extracted from diffusion MRI,
and associated tissue composition is obviously which is the reason why a number of methods
extremely complex: white matter is not made up of attempt to estimate it.
neatly arranged impermeable cylinders, but con-
tains a wide range of different cells of different
sizes and shapes, each with their own internal 2
Although there are assumptions on the way data are
microstructure. Clearly, the DW signal depends on acquired, as discussed in the section entitled The narrow
a myriad of different factors, and any attempt at pulse approximation below.
392 S. Farquharson and J.-D. Tournier

Fig. 20.7 Illustration of the relationship between the full three-dimensional density function, the characteriza-
fiber configuration (left), the average spin propagator tion of which requires a vast amount of data. In contrast,
(middle), and the diffusion ODF (right). In a crossing fiber the diffusion ODF (right) provides a more condensed ver-
voxel (left), water molecules will tend to diffuse more sion of spin propagator, which essentially describes the
readily along the fiber orientations than across them. This probability of a water molecules moving along any given
corresponds to a spin propagator with pronounced orientation. As expected, the peaks of the dODF point
“ridges” along the fiber orientations (middle). This is a along the fiber orientations

According to q-space, the DW signal is related Nonetheless, this complete characterization is


to the spin propagator via a simple Fourier exactly what Diffusion Spectrum Imaging
transform. In practice, this means that to estimate (DSI) aims to achieve [49]. To do this, 515 image
the spin propagator along one dimension, a num- volumes are acquired, each with a distinct
ber of DW images need to be acquired, each with q-vector with a maximum b-value of 17,000 s/
a different q-value.3 To get an accurate estimate, mm2. This is clearly a much more demanding
a sufficient number of distinct q-values need to acquisition than can realistically be accommo-
be acquired, up to a sufficiently large maximum dated in routine clinical imaging, and for this rea-
q-value to ensure near-complete nulling of the son much of the recent development in this area
DW signal. These two requirements alone are has been focused on obtaining relevant features
difficult to achieve: for a full three-dimensional of the spin propagator (namely the diffusion ori-
characterization of the spin propagator, the num- entation density function; dODF) using the much
ber of distinct q-vectors (and hence image vol- more clinically achievable HARDI acquisition.
umes) required is of the order of 1000 (approx.
10 per image axis). These requirements also dic- The Diffusion Orientation Density
tate the use of very large gradients and/or long Function (dODF)
echo times (to achieve the very large maximum Essentially, the dODF is a simplified version of
q-values needed), leading to a reduction in the the full spin propagator, which provides only the
SNR. Clearly, the full characterization of the spin probability of a water molecule moving along a
propagator is extremely challenging. given direction; any information about how far it
moved is discarded. This much more compact
3
version of the spin propagator is obviously still
The q-value is a measure of the strength of the diffusion
sufficiently informative, especially for the pur-
weighting, akin to the b-value. It is given by q = (γ/2π)δG,
where δ and G are the DW gradient pulse duration and poses of resolving crossing fibers, motivating the
amplitude respectively, and γ is the magnetogyric ratio. development of these HARDI-based methods.
20 High Angular Resolution Diffusion Imaging 393

Q-ball Imaging (QBI) was the first method mally narrow. More specifically, the distance
proposed to estimate the dODF [50]. It performs a moved by molecules during the application of
Funk–Radon Transform on the HARDI data to each DW gradient pulse should be much smaller
map the DW signal per-voxel onto the corresponding than the distance moved between the pulses. The
dODF. This has the advantage of being a linear idea is that each DW gradient pulse respectively
operation, and was subsequently shown to be much “tags” and “untags” water molecules based on
more efficiently and robustly implemented using their current location, so that the only relevant
spherical harmonics [51, 52]. More recently, quantity is their actual displacement between
Constant Solid Angle Q-ball Imaging (CSA- these two events. For this approximation to hold
QBI) has been proposed to obtain a more accurate, in white matter, water molecules should not be
sharper estimate of the dODF [53]. allowed to diffuse by any distance approaching
Other techniques based on q-space that have the axonal diameter. Even if we assume an accept-
been proposed include the Diffusion Orientation able distance to be 1 μm (which is already larger
Transform (DOT), which provides a contour of than many axons) and a diffusion coefficient of
the spin propagator evaluated at a particular value 10−3 mm2/s (which is smaller than would typically
of the displacement—in other words, it provides be assumed), the DW pulse duration would need
the probability of a water molecule moving along to be less than 0.5 ms. To achieve any reasonable
any given direction by a fixed distance [54]. b-value with such a short pulse duration would
Another early technique is Persistent Angular require gradients strengths and rise-times orders
Structure MRI (PAS-MRI), which evaluates an of magnitude larger than can be used in the clinic.
ODF from relatively modest data by imposing a In practice, the minimum DW pulse duration than
maximum entropy constraint [55]. See Fig. 20.8 can realistically be achieved on a human system is
for an illustration of the results obtained using of the order of 10 ms. Clearly, the narrow pulse
three different dODF estimation methods. approximation cannot hold on a clinical system.
Thankfully, the impact of breaking this assump-
The Narrow Pulse Approximation tion is actually relatively trivial for most clinical
The theory of q-space does rely on one approxi- applications. While this does invalidate some of
mation: that the duration of the DW gradient the quantitative estimates that might otherwise
pulses is negligible—in other words, the DW gra- have been derived using these methods, it does not
dient pulses can be considered to be infinitesi- affect the overall angular structure of the spin

Fig. 20.8 An illustration of the results obtained using three the corona radiata (inferior–superior) and the superior longi-
different dODF estimation methods: Q-ball imaging (QBI; tudinal fasciculus (anterior–posterior) [Reprinted from
left), constant solid angle QBI (CSA-QBI; middle), and the Aganj I, Lenglet C, Sapiro G, Yacoub E, Ugurbil K, Harel
diffusion orientation transform (DOT; right). These are dis- N. Reconstruction of the orientation distribution function in
played as coronal projections overlaid on the fractional single- and multiple-shell q-ball imaging within constant
anisotropy map, showing the well-known crossing of the solid angle. Magn Reson Med. 2010 Aug;64(2):554–66.
lateral projections of the corpus callosum (left–right) with With permission from John Wiley & Sons.]
394 S. Farquharson and J.-D. Tournier

propagator [49]. In fact, some studies suggest that since the diffusion ODF is inevitably broader than
such long diffusion times might in fact be benefi- the corresponding fiber ODF—in other words, the
cial when estimating the orientation and density of diffusion ODF cannot be sharper than the fiber
fiber bundles [47, 56]. The failure to meet the nar- configuration from which it emanated. In fact,
row pulse approximation is therefore unlikely to methods have been proposed to “sharpen” the dif-
have any detrimental impact, and indeed many fusion ODF to recover the fiber ODF using a spher-
successful techniques have been proposed that are ical deconvolution operation, a process shown to be
based on q-space, as described previously. ultimately equivalent to the “standard” model-
based spherical deconvolution [58].
Extracting Useful Information Another potential benefit of the dODF is to
from the dODF derive measures of tissue microstructure. However,
Methods that aim to characterize the diffusion there is relatively little on that topic in the litera-
propagator have the advantage of being “model- ture to date. The main scalar index that has been
free”: no assumptions are made with respect to extracted from the dODF is the generalized frac-
the microstructure, and the only information pro- tional anisotropy (GFA) index [50], which is
vided is strictly based only the measurements essentially the standard deviation of the dODF
themselves. While this may seem to be an attrac- relative to its RMS amplitude. Unfortunately, it
tive property, in practice this may actually cause has no clear biological interpretation, depends on
problems when applied in clinical and neurosci- the data acquisition parameters and technique used
entific studies. The reason for this is essentially to reconstruct the dODF, and is also sensitive to
the very reason that these methods are claimed to the fiber configuration: the GFA is inherently
be advantageous: being model-free, the informa- lower in crossing fiber regions, making it unsuit-
tion they do provide is not what most researchers able as a measure of white matter “integrity.” To
are actually interested in. As a consequence, end- date, no readily interpretable metric has been pro-
users will typically resort to ad-hoc methods to posed that can be derived directly from the dODF
extract the information they need, in ways that to characterize white matter tissue.
are often demonstrably inferior and/or actually For these reasons, while the “model-free”
involve a model of some sort. nature of these approaches does initially seem
For instance, the extraction of fiber orientations appealing, it is unclear how these advantages can
from the dODF is typically done by finding the actually be translated through to routine investiga-
peaks of the dODF. While this may initially seem tions. The diffusion ODF simply does not provide
sensible, it suffers from a number of limitations. the information typically desired in routine stud-
First, the power to resolve closely aligned fiber ori- ies, be it for fiber-tracking or the characterization
entations is typically reduced when using dODF- of white matter “integrity.” While there may be
based methods [6, 7, 57–59]. Second, this cases where the model-free nature of these meth-
introduces a bias in the estimated orientations when ods is advantageous, in most cases the fiber ODF
the fibers do not cross at 90° [59, 60]. Finally, peak- is the information required, particularly for fiber-
finding inherently imposes a model on the form of tracking applications—in which case it seems
the fiber orientation distribution: it assumes that more appropriate to use the model-based tech-
fiber bundles are arranged in distinct bundles with niques introduced in the previous section.
discrete fiber orientations. It explicitly does not
allow for curvature or divergence within a voxel.
While tensor-fitting methods admittedly also suffer HARDI in Practice
from this limitation, methods that recover the full
fiber ODF do not make any such assumption. Data Acquisition
While newer dODF methods such as CSA-QBI
and DOT do provide better resolving power than HARDI is characterized primarily by two param-
the original QBI approach, they still cannot provide eters: the number of unique directions, and the
better separation than model-based techniques, b-value. The optimal values for both of these
20 High Angular Resolution Diffusion Imaging 395

parameters are difficult to ascertain in general, ing either will increase the scan time, and increas-
since different reconstruction methods will have ing the spatial resolution will also lower the SNR,
different requirements. Nonetheless, it is generally potentially compromising the performance of the
acknowledged that higher b-values (b = 2500– HARDI reconstruction. In practice, the optimal
3000 s/mm2) allow better estimation of fiber orien- set of parameters will need to be determined
tations [9, 10]. While the DW images undeniably empirically on a case-by-case basis, based on the
look much noisier at these b-values, they actually intended HARDI reconstruction method and its
exhibit near-optimal contrast in the angular particular requirements.
domain, which is what is required to resolve the
different contributions from the various fiber
orientations present (Fig. 20.9). Note however that Clinical Applications
good results can still be obtained using b = 1000
s/mm2 data, provided they are of sufficient quality; The advantages of using more advanced HARDI-
indeed many HARDI studies have been published based methods specifically developed to provide
using b-values in this range [14, 29, 30, 58]. more robust estimates of the fiber orientations are
The number of directions required for HARDI now being realized for applications such as diffu-
is difficult to ascertain exactly since increasing it sion tractography, and more recently in the
will always improve results through increasing assessment of tissue microstructure. Despite the
the overall SNR of the acquisition. The primary wealth of studies in the technical literature that
requirement of the angular sampling is to charac- focus on the validation of the various methods
terize the relevant features of the DW signal. At a providing illustrative examples of white matter
b-value of ~3000 s/mm2, it can be shown that pathways in healthy volunteers, to date, there are
these angular features can be adequately charac- comparatively few studies that have utilized
terized using a minimum of 45 directions [61]. In HARDI-based methods for clinical applications.
practice however, a larger number is generally It is generally conceded that the lack of transla-
recommended to increase the overall SNR, since tion of HARDI-based methods to the clinical
most reconstruction methods will give poor research setting has largely been due to software
results on such data at a typical spatial resolution availability, technical expertise, and in some
of ~2 mm. There is clearly a trade-off between cases clinically impractical scans times. It may
spatial resolution and angular resolution: increas- also be due to a lack of awareness of the full

Fig. 20.9 A simple illustration of the influence of b-value much more clearly depicted at higher b-values, and the
on the measured DW data. Using low b-values (right) pro- presence of the multiple fiber orientations is much more
vides little signal attenuation, and hence good SNR in the obvious, particularly when looking at the scaled-up version
DW images. However, there is little contrast in angular of the results (bottom row). However, this does come with a
domain: the DW signal is very smooth, making is difficult significant reduction in the overall DW signal. In practice,
to make out the two fiber orientations present in this exam- the optimal b-value is a compromise between angular con-
ple. In contrast, the relevant features of the DW signal are trast on the one hand, and signal to noise on the other
396 S. Farquharson and J.-D. Tournier

impact of the limitations of DTI, of the wide- patients studied [62] (Fig. 20.10). Whilst this
spread nature of crossing fiber regions, or of the early study highlights improved tractography
availability of clinically practical alternatives. In results even with the earliest of HARDI-based
this next section, we describe some of the more techniques, there was inconsistency in the ability
recent work focussed specifically on clinical of the multi-tensor model to depict fibers extend-
applications of HARDI-based techniques. ing laterally to the face region. Yamada et al. [62]
attributed this shortfall to the fact that the study
Targeted Tractography relied on relatively low b-value data of 1000 s/
for Neurosurgery mm2, and only 32 directions. Whilst the study
Development of white matter fiber-tracking tech- may have benefited from the acquisition of higher
niques more than a decade ago represented an quality data to improve fiber orientations esti-
extraordinary achievement in neuroscience, with mates through increasing the overall SNR, it is
the potential to have huge impact on the diagno- likely that the failure of the multi-tensor tech-
sis and treatment of brain disorders. From a neu- nique may simply be because the model is lim-
rosurgical perspective in particular, targeted ited to fitting only two tensors in voxels through
tractography was expected to become the ulti- regions, such as the centrum semiovale, which
mate tool for surgical planning as it promised the are known to contain more than two fiber
ability to noninvasively map specific structural populations.
connections in the human brain in vivo in patients Berman [16] presented a more complete delin-
with brain lesions. Since its inception, there have eation of the corticospinal pathways in a case
been hundreds of studies presented in the neuro- used to demonstrate the potential capabilities of
surgical literature demonstrating the potential for q-ball tractography for the purpose of neurosur-
mapping white matter pathways that support elo- gical navigation. Although this article was lim-
quent function for the purposes of planning and ited to the presentation of illustrative cases, it was
navigation, see Chap. 21 for review. However, clear that that the HARDI-based tractography
over 90 % of the tractography literature to date is method had the capacity to provide a more accu-
based on DTI-based techniques despite its known rate description of fiber tracts through regions
practical limitations. There have also been more such as the centrum semiovale than that previ-
recent appeals by some members of the neurosur- ously shown using single and multi-tensor based
gical community to consider DTI-based fiber- studies [19, 63–69]. The practical advantages of
tracking techniques obsolete for the purpose of using HARDI-based methods for the delineation
neurosurgical planning [27]. of the corticospinal pathways have recently been
Despite the lack of translation to the clinical more systematically evidenced in the neurosur-
setting, a number of studies in the neurosurgical gery literature, in a study based on constrained
literature have clearly demonstrated the advan- spherical deconvolution (CSD) [10, 29]. The data
tages of using various HARDI-based tractogra- presented demonstrated that the CSD-based trac-
phy techniques (as opposed to DTI-based tography technique, when directly compared to
methods) specifically for delineating tracts of both deterministic and probabilistic DTI-based
neurosurgical interest. An early HARDI-based tractography techniques, consistently resulted in
tractography study comparing initial clinical more biologically reliable tractography
experience using single tensor technique versus information that provided improved estimates of
the multi-tensor approaches to depict the motor safety margins which may be useful in neurosur-
pathways, illustrated that although there were gical procedures—see illustrative cases provided
“perceived” advantages of using the single tensor in Fig. 20.11 [70].
technique (i.e., instantaneous calculation of trac- It should be emphasized that the more accu-
tography maps), the multi-tensor appeared to rate representation of the corticospinal pathways
offer better depiction of motor tracts from the using the CSD-based tractography method com-
face and tongue regions in over half of the pared to DTI-based tractography presented in
20 High Angular Resolution Diffusion Imaging 397

Transaxial view
of multi-tensor
tractography

3-dimensional
coronal view

Single-tensor Multi-tensor
Fig. 20.10 Example of single tensor vs. multi-tensor Hoogenraad FGC, Holthuizen R, Akazawa K, Ito H, et al.
tractography in a 58-year-old woman with glioblastoma Multitensor Tractography Enables Better Depiction of
multiforme. Note that the pyramidal fibers of lesional side Motor Pathways: Initial Clinical Experience Using
(right) are not depicted using single-tensor tractography, Diffusion-Weighted MR Imaging with Standard b-Value.
whereas they are well shown by using multitensor tractog- AJNR Am J Neuroradiol. 2007 Oct 1;28(9):1668–73.
raphy. These fibers are noted to have substantial anterior With permission from American Society of
displacement [Reprinted from Yamada K, Sakai K, Neuroradiology]

this study was consistent across a range of diffu- tracts, given that such tracts will inevitably tra-
sion acquisition schemes, and included DWI data verse through voxels containing substantial con-
typically considered optimal for both DTI- and tributions from two of more fiber populations at
CSD-based methods (Fig. 20.12) [71, 72]. some point along their path.
Although the configuration of tracks identified by Over the past few years, HARDI-based trac-
the CSD-based method appears to be better tography has also been used to demonstrate
defined when using higher b-value DWI data improvements in the delineation of other func-
acquired using a higher number of diffusion tionally important tracts including the optic radi-
directions (as expected—see above), it is clear ations and the arcuate fasciculus. Recent studies
from the data presented in this study that the depicting Meyer’s loop of the optic radiation
application of this HARDI based model to the using HARDI data and a multi-tensor tractogra-
simpler diffusion acquisition schemes also results phy technique [73] demonstrate that, although
in a considerable improvement in tractography there is biological variability in the distance from
results, particularly in crossing fiber regions (Fig. the temporal pole (TP) to Meyer’s loop (ML),
20.12). While this study was confined to the multi-tensor tractography can achieve results that
investigation of the corticospinal pathways, it is are more consistent with anatomical dissection
likely that the results will apply to most fiber studies [74] than previously reported [75]. These
398 S. Farquharson and J.-D. Tournier

Fig. 20.11 Examples of CSD-based (red) vs. DTI-based DTI-based tractography results in Case B suggest that
(blue) tractography results with segmented pathology only the medial aspect of the lesion impinges on the cor-
volumes (green) overlaid on coronal T1-weighted images ticospinal tracts (i and ii), whereas the CSD-based trac-
in a 49-year-old woman with a large left-sided temporo- tography results suggest that the lesion is enveloped by
parietal AVM (Case A), and a 24-year-old woman with a medial and lateral projections of corticospinal fibers (iii
right focal cortical dysplasia situated in the right poste- and iv) [Reprinted from Farquharson S, Tournier J-D,
rior frontal lobe (Case B). Note that the DTI-based trac- Calamante F, Fabinyi G, Schneider-Kolsky M, Jackson
tography results in Case A suggest a clear margin GD, et al. White matter fiber tractography: why we need
surrounding the lesion (i and ii), whereas the CSD-based to move beyond DTI: Clinical article. Journal of
tractography results indicate that lateral projections of Neurosurgery. 2013 Jun;118(6):1367–77. With permis-
the corticospinal pathway may be at risk (iii and iv). The sion from AANS]

findings are important because it had been previ- more accurate marker, which could be used to
ously suggested that patients with a shorter better predict postoperative VFD.
TP-ML distance were at greater risk of a visual Given the increasing demand for accurate neu-
field deficit (VFD) following anterior temporal rosurgical tractography information and the ever-
lobe resection (ATLR), while the data presented present challenges that complicate tractography
by Winston et al. suggests that there is no differ- in the neurosurgical setting (the variation in anat-
ence in TP-ML distance between patients devel- omy of individual patients, disease-related
oping a VFD and those who do not [73]. In fact, change, and the inevitable intraoperative shift of
the direct comparison of visual field deficits prior anatomy during neurosurgical procedures, to
to, and at 3 and 12 months post-surgery and the name just a few), it is imperative we continue to
preoperative tractography data co-registration identify optimal strategies for targeted tractogra-
with the postoperative anatomical data in the 20 phy for the purpose of neurosurgical navigation in
ATLR patients studied suggest that the size of the individual patients [76, 77]. Although HARDI-
resection and the degree of damage to ML is a based techniques such as the so-called high defi-
20 High Angular Resolution Diffusion Imaging 399

Fig. 20.12 Comparison of tractography results obtained whom tracks were identified in any given voxel, using
using DTI combined with a deterministic algorithm and DTI (left column) and CSD (right column) (range 0–12).
CSD combined with a probabilistic algorithm across a Note that CSD based tractography results in a consider-
range of diffusion acquisition protocols that differed in the able improvement in the delineation of corticospinal path-
number of diffusion directions and b-value used, as indi- ways even if using the most basic of diffusion acquisition
cated in the figure. Panel (a) shows coronal T1-weighted schemes [Reprinted from Farquharson S, Tournier J-D,
images overlaid with tractography results using DTI (left Calamante F, Fabinyi G, Schneider-Kolsky M, Jackson
column) and CSD (right column) from a representative GD, et al. White matter fiber tractography: why we need
normal control subject. Panel (b) shows a coronal FA tem- to move beyond DTI: Clinical article. Journal of
plate image overlaid with frequency maps representing Neurosurgery. 2013 Jun;118(6):1367–77. With permis-
the number of subjects (out of 12 control subjects) in sion from AANS]

nition fiber tracking method (HDFT) (which HARDI-based tractography methods represents
combines diffusion spectrum imaging (DSI) [49] significant technical advancement that allows
with generalized q-sampling imaging for estima- more biologically accurate delineation of white
tion of fiber orientations [50]) are already being matter pathways than was previously achievable
used to facilitate innovative neurosurgical appli- using DTI-based tractography techniques.
cations [18], there is at present only anecdotal evi-
dence to support the use of one HARDI-based Assessment of Structural Connectivity
tractography approach over another. Despite the One of the advantages of utilizing HARDI-based
lack of more rigorous clinically based studies into methods developed specifically to attain more
the accuracy and reproducibility of these more robust fiber orientation estimates is that it is now
advanced methods, it is clear from the current possible to perform more reliable tractography-
body of neurosurgical evidence that the advent of based comparisons throughout the brain. One of
400 S. Farquharson and J.-D. Tournier

Fig. 20.13 Comparison of DTI and QBI tractography DTI in both subjects with pAgCC and controls [Reprinted
results in a control subject (top) and a subject with partial from Wahl M, Strominger Z, Jeremy RJ, Barkovich AJ,
agenesis of the Corpus Callosum pAgCC (bottom) using Wakahiro M, Sherr EH, et al. Variability of Homotopic
(1) DTI tractography performed on a DTI acquisition at and Heterotopic Callosal Connectivity in Partial Agenesis
b = 1000 s/mm2 (a, d), (2) DTI tractography on a HARDI of the Corpus Callosum: A 3 T Diffusion Tensor Imaging
acquisition at b = 3000 s/mm2 (b, e), and (3) QBI tractog- and Q-Ball Tractography Study. AJNR Am J Neuroradiol.
raphy is shown for the same HARDI acquisition at 2009 Feb 1;30(2):282–9. With permission from American
b = 3000 s/mm2 (c, f). Note that QBI tractography typi- Society of Neuroradiology]
cally recovered more extensive fibers for each tract than

the first applications of HARDI-based fiber healthy subjects. The DTI technique was only
tracking to assess differences in structural con- able to delineate a subset of these tracts due to its
nectivity in a patient population was performed in ability to tract through crossing regions (Fig.
to investigate inter-hemispheric connections in 20.13). Although the authors concede that the
patients with partial agenesis of the corpus cal- small cohort of subjects studied with pAgCC pre-
losum (pAgCC) [78]. This early study directly cluded any generalization of results to this phe-
compared results from DTI and Q-ball imaging notypically heterogeneous population, the
(QBI) reconstruction methods in six individuals variability of these aberrant structural connec-
with pAgCC and eight control subjects. The QBI tions in these few subjects was thought to be
data presented in this study revealed that individ- clinically important because individuals with
uals with pAgCC had unexpectedly highly vari- agenesis of the corpus callosum often present
able callosal fiber connectivity, which included with a broad range of behavioral and neurocogni-
many heterotopic tracts that were not seen in the tive deficits [78].
20 High Angular Resolution Diffusion Imaging 401

Recent connectivity studies using HARDI- The super resolution TDI tractography maps
based methods have identified subtle differences were used in conjunction with targeted tractogra-
in the healthy brain that may be critical to our phy maps to specifically enable visualization and
understanding and interpretation of deviations assessment of cerebello-cerebral connections.
from normal structural connectivity. One of the The findings from this interesting study go fur-
largest studies based on HARDI data investigated ther than previous diffusion tensor-based studies,
structural differences in 569 healthy twins [79]. which have been successful in identifying pri-
This study utilized an orientation density func- mary sites of white matter degeneration, to iden-
tion (ODF)-based tractography technique, to tify secondary sites of degeneration that involve a
measure “fiber density” (i.e. the volume of network of cortical and subcortical regions. The
streamlines successfully generated with a region) authors suggest that these findings demonstrate
as a marker of structural connectivity [79]. While that FRDA pathology extends beyond well-
the use of volume as a measure of connectivity is known primary sites of cerebellar and brainstem
not without its confounds [26], the results sug- degeneration; and that such results provide an
gest that the proportions of fibers intersecting the explanation for the non-motor symptoms seen in
left and right hemispheres vary significantly with the disease, such as depression and cognitive dif-
age. In a further study by the same group, which ficulties (Fig. 20.14) [82].
aimed to define the normal trajectory of struc-
tural connectivity from early adolescence to early Assessment of Structural Integrity
adulthood, it was also observed that increases Recent developments in the field have emerged to
and decreases in densities across the maturing allow group-wise whole-brain analysis of diffu-
brain are not distributed evenly [80]: the frontal sion data, in a manner robust to crossing fibers.
cortex had a disproportionate decrease in fiber Apparent fiber density (AFD) is one such method,
density whilst the temporal lobe showed an which uses the fiber orientations distributions
increase in fiber density with age in healthy par- (FOD) computed using constrained spherical
ticipants aged of 12–30 years. deconvolution (CSD) to identify tract-specific dif-
One of the limitations of current methods to ferences in fiber density [83]. This approach was
assess structural connectivity using diffusion- applied in a recent study investigating HARDI
weighted data, compared to traditional histopathol- data collected in patients with probable or definite
ogy, is that the spatial resolution achievable within motor neurone disease (MND) and age matched
a clinically feasible time frame is limited. A recent controls [47]. The results of this study show a sig-
technical advance that may have important future nificant decrease in AFD, specifically within vox-
applications is the development of a post-process- els and orientations corresponding to the
ing technique known as Track Density Imaging corticospinal tract (CST) and corpus callosal (CC)
(TDI) [81]. TDI is an approach that uses the spatial fibers, in the group data of MND patients com-
information from tractography streamlines to pared to healthy control subjects—pathways that
improve spatial resolution beyond that by which are known to be affected in upper motor neuron
the data were originally acquired. The resultant impairment (Fig. 20.15) [47]. The advantage of
super-resolution track density images reveal using this approach is the ability to identify not
enhanced anatomical detail with high SNR allow- only the location, but also the orientation along
ing direct visualization of sub-structures in regions which differences in fiber density can be observed.
such as the thalamus and cerebellar peduncles [81]. Using the FOD as a measure of apparent fiber
The super resolution properties of TDI have density may prove to be a more accurate and read-
recently been appreciated in a study of patients ily interpretable marker of tract “integrity”
with Friedreich ataxia (FRDA) where TDI was throughout the brain than DTI-derived measures
performed to increase the resolution to 500 μm, of diffusion anisotropy, particularly in regions
four times the native imaging resolution [82]. containing multiple fiber populations, since the
402 S. Farquharson and J.-D. Tournier

Fig. 20.14 example of group-averaged track density spinal tract (d) between patients with FRDA and control
images (TDI) super resolved to 500 μm from HARDI data subjects [Reprinted from Zalesky A, Akhlaghi H, Corben
acquired at 2 mm3 in healthy controls and patients with LA, Bradshaw JL, Delatycki MB, Storey E, et al.
Friedreich ataxia (FRDA). Note the visually evident dif- Cerebello-cerebral connectivity deficits in Friedreich
ferences in structural connections from regions such as ataxia. Brain Struct Funct. 2014; 219(3): 969–981. With
the anterior thalamic radiation (a), brain-stem and dentate permission from Springer-Verlag.]
nucleus (b), middle cerebellar peduncle (c) and cortico-

fiber density estimates for one tract are no longer development in the field of diffusion MRI. These
confounded by the presence of other tracts that methods are now beginning to be used for clinical
might be traversing the same region. and neuroscientific investigations with extremely
promising results. Although further validation is
necessary, it is clear that the development and
Conclusion application of HARDI-based methods have
moved the field one important step closer to one
The advent of HARDI-based methods specifi- of the great challenges for the upcoming decade—
cally developed to provide more robust estimates to noninvasively map the structural connectivity
of the fiber orientations represents an exciting of the human brain.
20 High Angular Resolution Diffusion Imaging 403

Fig. 20.15 example of Apparent Fiber Density (AFD) right primary motor cortices [Reprinted from Raffelt D,
results of patients with motor neuron disease (MND) Tournier J-D, Rose S, Ridgway GR, Henderson R, Crozier
compared to health control subjects. Note that a signifi- S, et al. Apparent Fiber Density: A novel measure for the
cant decrease in AFD was detected in MND patients analysis of diffusion-weighted magnetic resonance
within two distinct clusters: Cluster 1 extends from the images. Neuroimage. 2012 Feb 15;59(4):3976–94. With
lower brain stem to the internal capsule, and cluster 2 cor- permission from Elsevier.]
responds to corpus callosal fibers connecting the left and
404 S. Farquharson and J.-D. Tournier

References degeneration and their dependence on white matter


architecture. Neuroimage. 2001;13(6):1174–85.
16. Berman J. Diffusion MR, tractography as a tool for
1. Alexander AL, Hasan KM, Lazar M, Tsuruda JS,
surgical planning. Magn Reson Imaging Clin N Am.
Parker DL. Analysis of partial volume effects in
2009;17(2):205–14.
diffusion-tensor MRI. Magn Reson Med. 2001;45(5):
17. Farquharson S, Tournier J-D, Calamante F, Fabinyi G,
770–80.
Schneider-Kolsky M, Jackson GD, et al. White matter
2. Alexander DC, Barker GJ, Arridge SR. Detection and
fiber tractography: why we need to move beyond
modeling of non-Gaussian apparent diffusion
DTI. J Neurosurg. 2013;118:1367.
coefficient profiles in human brain data. Magn Reson
18. Fernandez-Miranda JC, Pathak S, Engh J, Jarbo K,
Med. 2002;48(2):331–40.
Verstynen T, Yeh F-C, et al. High-definition fiber trac-
3. Frank LR. Anisotropy in high angular resolution
tography of the human brain: neuroanatomical valida-
diffusion-weighted MRI. Magn Reson Med. 2001;
tion and neurosurgical applications. Neurosurgery.
45(6):935–9.
2012;71(2):430–53.
4. Tuch DS, Reese TG, Wiegell MR, Makris N, Belliveau
19. Kinoshita M, Yamada K, Hashimoto N, Kato A,
JW, Wedeen VJ. High angular resolution diffusion
Izumoto S, Baba T, et al. Fiber-tracking does not
imaging reveals intravoxel white matter fiber hetero-
accurately estimate size of fiber bundle in pathologi-
geneity. Magn Reson Med. 2002;48(4):577–82.
cal condition: initial neurosurgical experience using
5. Seunarine KK, Alexander DC. Multiple fibres:
neuronavigation and subcortical white matter stimula-
beyond the diffusion tensor. In: Johansen-Berg H,
tion. Neuroimage. 2005;25(2):424–9.
Behrens TEJ, editors. Diffusion MRI: from quantita-
20. Nimsky C, Ganslandt O, Fahlbusch R. Implementation
tive measurement to in-vivo neuroanatomy. San
of fiber tract navigation. Neurosurgery. 2006;58(4
Diego, CA: Elsevier; 2009. p. 56–74.
Suppl 2):ONS–292–303. discussion ONS–303–304.
6. Tournier JD. The biophysics of crossing fibres. In:
21. Jones DK, Cercignani M. Twenty-five pitfalls in the
Jones DK, editor. Diffusion MRI: theory, methods,
analysis of diffusion MRI data. NMR Biomed.
and applications. New York, NY: Oxford University
2010;23(7):803–20.
Press; 2010. p. 465–82.
22. Vos SB, Jones DK, Jeurissen B, Viergever MA,
7. Tournier J-D, Mori S, Leemans A. Diffusion tensor
Leemans A. The influence of complex white matter
imaging and beyond. Magn Reson Med. 2011;
architecture on the mean diffusivity in diffusion ten-
65(6):1532–56.
sor MRI of the human brain. Neuroimage. 2012;
8. Alexander DC. Multiple-fiber reconstruction algo-
59(3):2208–16.
rithms for diffusion MRI. Ann N Y Acad Sci. 2005;
23. Wheeler-Kingshott CAM, Cercignani M. About
1064(1):113–33.
“axial” and “radial” diffusivities. Magn Reson Med.
9. Alexander DC, Barker GJ. Optimal imaging parame-
2009;61(5):1255–60.
ters for fiber-orientation estimation in diffusion
24. Pierpaoli C, Basser PJ. Toward a quantitative assess-
MRI. Neuroimage. 2005;27(2):357–67.
ment of diffusion anisotropy. Magn Reson Med.
10. Tournier J-D, Calamante F, Gadian DG, Connelly
1996;36(6):893–906.
A. Direct estimation of the fiber orientation density func-
25. Douaud G, Jbabdi S, Behrens TEJ, Menke RA, Gass
tion from diffusion-weighted MRI data using spherical
A, Monsch AU, et al. DTI measures in crossing-fibre
deconvolution. Neuroimage. 2004;23(3):1176–85.
areas: increased diffusion anisotropy reveals early
11. Vos SB, Jones DK, Viergever MA, Leemans A. Partial
white matter alteration in MCI and mild Alzheimer’s
volume effect as a hidden covariate in DTI analyses.
disease. Neuroimage. 2011;55(3):880–90.
Neuroimage. 2011;55(4):1566–76.
26. Jones DK, Knösche TR, Turner R. White matter integ-
12. Basser PJ, Pierpaoli C. Microstructural and physio-
rity, fiber count, and other fallacies: the do’s and don’ts
logical features of tissues elucidated by quantitative-
of diffusion MRI. Neuroimage. 2013;73:239–54.
diffusion-tensor MRI. J Magn Reson B. 1996;111(3):
27. Fernandez-Miranda JC. Editorial: beyond diffusion
209–19.
tensor imaging. J Neurosurg. 2013;118(6):1363–6.
13. Jeurissen B, Leemans A, Tournier J-D, Jones DK,
28. Jones DK. The effect of gradient sampling schemes
Sijbers J. Investigating the prevalence of complex
on measures derived from diffusion tensor MRI: a
fiber configurations in white matter tissue with diffu-
Monte Carlo study. Magn Reson Med. 2004;51(4):
sion magnetic resonance imaging. Human Brain
807–15.
Mapping [Internet]. 2012 May 19 [cited 2012 Jun 15];
29. Tournier J-D, Calamante F, Connelly A. Robust deter-
Available from: http://www.ncbi.nlm.nih.gov/
mination of the fibre orientation distribution in diffu-
pubmed/22611035
sion MRI: non-negativity constrained super-resolved
14. Behrens TEJ, Berg HJ, Jbabdi S, Rushworth MFS,
spherical deconvolution. Neuroimage. 2007;35(4):
Woolrich MW. Probabilistic diffusion tractography
1459–72.
with multiple fibre orientations: what can we gain?
30. Parker GJM, Alexander DC. Probabilistic Monte
Neuroimage. 2007;34(1):144–55.
Carlo based mapping of cerebral connections utilising
15. Pierpaoli C, Barnett A, Pajevic S, Chen R, Penix L,
whole-brain crossing fibre information. Inf Process
Virta A, et al. Water diffusion changes in Wallerian
Med Imaging. 2003;18:684–95.
20 High Angular Resolution Diffusion Imaging 405

31. Hosey T, Williams G, Ansorge R. Inference of mul- orientational anisotropy, a true-tract specific index to
tiple fiber orientations in high angular resolution dif- characterize white matter diffusion. Human Brain
fusion imaging. Magn Reson Med. 2005;54(6): Mapping [Internet]. 2012 Apr 5 [cited 2012 Apr 12];
1480–9. Available from: http://www.ncbi.nlm.nih.gov/
32. Hosey TP, Harding SG, Carpenter TA, Ansorge RE, pubmed/22488973
Williams GB. Application of a probabilistic 46. Jbabdi S, Behrens TEJ, Smith SM. Crossing fibres in
double-fibre structure model to diffusion-weighted tract-based spatial statistics. Neuroimage. 2010;
MR images of the human brain. Magn Reson Imaging. 49(1):249–56.
2008;26(2):236–45. 47. Raffelt D, Tournier J-D, Rose S, Ridgway GR,
33. Assaf Y, Basser PJ. Composite hindered and restricted Henderson R, Crozier S, et al. Apparent fibre density:
model of diffusion (CHARMED) MR imaging of the a novel measure for the analysis of diffusion-weighted
human brain. Neuroimage. 2005;27(1):48–58. magnetic resonance images. Neuroimage. 2012;
34. Assaf Y, Freidlin RZ, Rohde GK, Basser PJ. New 59(4):3976–94.
modeling and experimental framework to characterize 48. Callaghan PT. NMR imaging, NMR diffraction and
hindered and restricted water diffusion in brain white applications of pulsed gradient spin echoes in porous
matter. Magn Reson Med. 2004;52(5):965–78. media. Magn Reson Imaging. 1996;14(7-8):701–9.
35. Tournier J, Calamante F, Connelly A. MRtrix: diffu- 49. Wedeen VJ, Hagmann P, Tseng W-YI, Reese TG,
sion tractography in crossing fiber regions. Int Weisskoff RM. Mapping complex tissue architecture
J Imaging Syst Technol. 2012;22(1):53–66. with diffusion spectrum magnetic resonance imaging.
36. Anderson AW. Measurement of fiber orientation dis- Magn Reson Med. 2005;54(6):1377–86.
tributions using high angular resolution diffusion 50. Tuch DS. Q-ball imaging. Magn Reson Med. 2004;
imaging. Magn Reson Med. 2005;54(5):1194–206. 52(6):1358–72.
37. Dell’Acqua F, Rizzo G, Scifo P, Clarke RA, Scotti G, 51. Descoteaux M, Angelino E, Fitzgibbons S, Deriche
Fazio F. A model-based deconvolution approach to R. Regularized, fast, and robust analytical Q-ball
solve fiber crossing in diffusion-weighted MR imag- imaging. Magn Reson Med. 2007;58(3):497–510.
ing. IEEE Trans Biomed Eng. 2007;54(3):462–72. 52. Hess CP, Mukherjee P, Han ET, Xu D, Vigneron
38. Kaden E, Knösche TR, Anwander A. Parametric DB. Q-ball reconstruction of multimodal fiber orien-
spherical deconvolution: inferring anatomical connec- tations using the spherical harmonic basis. Magn
tivity using diffusion MR imaging. Neuroimage. Reson Med. 2006;56(1):104–17.
2007;37(2):474–88. 53. Aganj I, Lenglet C, Sapiro G, Yacoub E, Ugurbil K,
39. Seunarine KK, Alexander DC. Linear Persistent Harel N. Reconstruction of the orientation distribu-
Angular Structure MRI and non-linear Spherical tion function in single- and multiple-shell q-ball
Deconvolution for Diffusion MRI. Proceedings of the imaging within constant solid angle. Magn Reson
International Society for Magnetic Resonance in Med. 2010;64(2):554–66.
Medicine. Seattle, Washington, USA; 2006. p. 2767. 54. Özarslan E, Shepherd TM, Vemuri BC, Blackband SJ,
40. Beaulieu C, Allen PS. Determinants of anisotropic Mareci TH. Resolution of complex tissue microarchi-
water diffusion in nerves. Magn Reson Med. tecture using the diffusion orientation transform
1994;31(4):394–400. (DOT). Neuroimage. 2006;31(3):1086–103.
41. Beaulieu C, Allen PS. Water diffusion in the giant 55. Jansons KM, Alexander DC. Persistent angular struc-
axon of the squid: implications for diffusion-weighted ture: new insights from diffusion magnetic resonance
MRI of the nervous system. Magn Reson Med. imaging data. Inverse Problems. 2003;19(5):
1994;32(5):579–83. 1031–46.
42. Beaulieu C, Does MD, Snyder RE, Allen PS. Changes 56. Yeh C-H, Tournier J-D, Cho K-H, Lin C-P, Calamante
in water diffusion due to Wallerian degeneration in F, Connelly A. The effect of finite diffusion gradient
peripheral nerve. Magn Reson Med. 1996;36(4): pulse duration on fibre orientation estimation in diffu-
627–31. sion MRI. Neuroimage. 2010;51(2):743–51.
43. Song S-K, Sun S-W, Ju W-K, Lin S-J, Cross AH, 57. Alexander DC, Seunarine KK. Mathematics of cross-
Neufeld AH. Diffusion tensor imaging detects and ing fibers. In: Jones DK, editor. Diffusion MRI: the-
differentiates axon and myelin degeneration in mouse ory, methods, and applications. New York, NY:
optic nerve after retinal ischemia. Neuroimage. Oxford University Press; 2010.
2003;20(3):1714–22. 58. Descoteaux M, Deriche R, Knösche TR, Anwander
44. Song S-K, Sun S-W, Ramsbottom MJ, Chang C, A. Deterministic and probabilistic tractography based
Russell J, Cross AH. Dysmyelination revealed on complex fibre orientation distributions. IEEE Trans
through MRI as increased radial (but unchanged Med Imaging. 2009;28(2):269–86.
axial) diffusion of water. Neuroimage. 2002;17(3): 59. Tournier JD, Yeh C-H, Calamante F, Cho K-H,
1429–36. Connelly A, Lin C-P. Resolving crossing fibres using
45. Dell’acqua F, Simmons A, Williams SCR, Catani constrained spherical deconvolution: validation using
M. Can spherical deconvolution provide more infor- diffusion-weighted imaging phantom data.
mation than fiber orientations? Hindrance modulated Neuroimage. 2008;42(2):617–25.
406 S. Farquharson and J.-D. Tournier

60. Zhan W, Yang Y. How accurately can the diffusion Proceedings of the International Society for Magnetic
profiles indicate multiple fiber orientations? A study Resonance in Medicine. Hawai’i, USA; 2009. p. 358.
on general fiber crossings in diffusion MRI. J Magn 73. Winston GP, Daga P, Stretton J, Modat M, Symms
Reson. 2006;183(2):193–202. MR, McEvoy AW, et al. Optic radiation tractography
61. Tournier J-D, Calamante F, Connelly A. Determination and vision in anterior temporal lobe resection. Ann
of the appropriate b value and number of gradient direc- Neurol. 2012;71(3):334–41.
tions for high-angular-resolution diffusion-weighted 74. Ebeling U, Reulen H-J. Subcortical topography and
imaging. NMR Biomed. 2013;26(12):1775–86. proportions of the pyramidal tract. Acta Neurochir.
62. Yamada K, Sakai K, Hoogenraad FGC, Holthuizen R, 1992;118(3-4):164–71.
Akazawa K, Ito H, et al. Multitensor tractography 75. Nilsson D, Rydenhag B, Malmgren K, Starck G,
enables better depiction of motor pathways: initial Ljungberg M. Anatomical accuracy and feasibility of
clinical experience using diffusion-weighted mr probabilistic and deterministic tractography of the
imaging with standard b-value. AJNR Am optic radiation. Epilepsia [Internet]. 2010 [cited 2013
J Neuroradiol. 2007;28(9):1668–73. Aug 29]. p. 91. Available from: http://onlinelibrary.
63. Berman JI, Berger MS, Chung SW, Nagarajan SS, w i l ey. c o m . e z p . l i b. u n i m e l b. e d u . a u /
Henry RG. Accuracy of diffusion tensor magnetic doi/10.1111/j.1528-1167.2010.02658.x/abstract
resonance imaging tractography assessed using intra- 76. Nimsky C, Ganslandt O, Merhof D, Sorensen AG,
operative subcortical stimulation mapping and mag- Fahlbusch R. Intraoperative visualization of the pyra-
netic source imaging. J Neurosurg. 2007;107(3):488. midal tract by diffusion-tensor-imaging-based fiber
64. Berman JI, Berger MS, Mukherjee P, Henry tracking. Neuroimage. 2006;1:30(4).
RG. Diffusion-tensor imaging-guided tracking of 77. Nimsky C, Ganslandt O, Fahlbusch R. Implementation
fibers of the pyramidal tract combined with intraop- of fiber tract navigation. Neurosurgery. 2007;61(1
erative cortical stimulation mapping in patients with Suppl):306–17. discussion 317–318.
gliomas. J Neurosurg. 2004;101(1):66–72. 78. Wahl M, Strominger Z, Jeremy RJ, Barkovich AJ,
65. Clark CA, Barrick TR, Murphy MM, Bell BA. White Wakahiro M, Sherr EH, et al. Variability of homotopic
matter fiber tracking in patients with space-occupying and heterotopic callosal connectivity in partial agen-
lesions of the brain: a new technique for neurosurgical esis of the corpus callosum: a 3T diffusion tensor
planning? Neuroimage. 2003;20(3):1601–8. imaging and q-ball tractography study. AJNR Am
66. Itoh D, Aoki S, Maruyama K, Masutani Y, Mori H, J Neuroradiol. 2009;30(2):282–9.
Masumoto T, et al. Corticospinal tracts by diffusion 79. Daianu M, Jahanshad N, Dennis EL, Toga AW,
tensor tractography in patients with arteriovenous mal- McMahon KL, de Zubicaray GI, et al. Left versus
formations. J Comput Assist Tomogr. 2006;30(4):618. right hemisphere differences in brain connectivity:
67. Mikuni N, Okada T, Enatsu R, Miki Y, Hanakawa T, 4-Tesla HARDI tractography in 569 twins. 2012 9th
Urayama S, et al. Clinical impact of integrated func- IEEE International Symposium on Biomedical
tional neuronavigation and subcortical electrical stim- Imaging (ISBI). 2012. p. 526–9.
ulation to preserve motor function during resection of 80. Dennis EL, Jahanshad N, McMahon KL, de Zubicaray
brain tumors. J Neurosurg. 2007;106(4):593–8. GI, Martin NG, Hickie IB, et al. Development of brain
68. Okada T, Miki Y, Kikuta K, Mikuni N, Urayama S, structural connectivity between ages 12 and 30: a
Fushimi Y, et al. Diffusion tensor fiber tractography 4-Tesla diffusion imaging study in 439 adolescents
for arteriovenous malformations: quantitative analy- and adults. Neuroimage. 2013;64:671–84.
ses to evaluate the corticospinal tract and optic radia- 81. Calamante F, Tournier J-D, Jackson GD, Connelly
tion. AJNR Am J Neuroradiol. 2007;28(6):1107–13. A. Track-density imaging (TDI): super-resolution
69. Yamada K, Kizu O, Ito H, Nishimura T. Tractography white matter imaging using whole-brain track-density
for an arteriovenous malformation. Neurology. mapping. Neuroimage. 2010;53(4):1233–43.
2004;62(4):669. 82. Zalesky A, Akhlaghi H, Corben LA, Bradshaw JL,
70. Farquharson S, Tournier J-D, Calamante F, Fabinyi G, Delatycki MB, Storey E, et al. Cerebello-cerebral
Schneider-Kolsky M, Jackson GD, et al. White matter connectivity deficits in Friedreich ataxia. Brain Struct
fiber tractography: why we need to move beyond DTI: Funct. 2014;219(3):969–81.
clinical article. J Neurosurg. 2013;118(6):1367–77. 83. Raffelt D, Tournier J-D, Crozier S, Connelly A,
71. Jones DK, Horsfield MA, Simmons A. Optimal strate- Salvado O, Tournier J-D. Apparent Fibre Density: a
gies for measuring diffusion in anisotropic systems by new measure for high angular resolution diffusion-
magnetic resonance imaging. Magn Reson Med. weighted image analysis. International Society for
1999;42(3):515–25. Magnetic Resonance in Medicine 14th Annual
72. Tournier J-D, Calamante F, Connelly A. How many Scientific Meeting and Exhibition: 2006 Proceedings.
diffusion gradient directions are required for HARDI? Stockholm, Sweden; 2010.
Diffusion Kurtosis Imaging
21
Jelle Veraart and Jan Sijbers

Robert Brown, who observed the everlasting jit-


Learning Points tery motion of grains of pollen suspended in water
under a microscope [1]. The Brownian motion
• DKI extends the DTI model by quantifying was theoretically substantiated by Einstein [2].
the degree of non-Gaussian diffusion He showed that the random motion of numerous
• DKI is clinically feasible and relevant molecules can mathematically be described by a
• DKI is a mathematical model and, as such, the displacement probability distribution function
biophysical interpretation might be challenging (PDF). This mathematical concept is briefly intro-
duced with a simple example (see Box 21.1).

Diffusion Concepts
Free Diffusion
As stated in Chap. 3, any type of molecule in a
fluid (e.g., water) is in constant random motion Einstein showed that the displacement of water
agitated by thermal energy. Given a large collec- molecules in an open body of water such as a
tion of molecules in an environment without glass of water can be described by a Gaussian
boundaries, molecules undergo a random walk probability distribution function [2]. In the
consisting of independent steps with a change of absence of flow—so the water must be still—the
direction after each collision with another mole- Gaussian distribution will be centered around
cule. This motion is named after the botanist zero. The distribution has zero mean. In addition
to the mean, a distribution has another important
property or statistic: the standard deviation. The
J. Veraart, PhD (*)
standard deviation is a measure of the width of
iMinds-Vision Lab, Department of Physics,
University of Antwerp, Universiteitsplein 1, the distribution. Its square is called the variance.
2610, Antwerp, Belgium If the mean displacement is zero, then the vari-
Center for Biomedical Imaging, Department of ance equals the mean squared displacement 〈x2〉,
Radiology, New York University School of Medicine, which is linked to the diffusion coefficient D and
New York, USA
the diffusion time t by the Einstein equation:
e-mail: Jelle.Veraart@uantwerpen.be
J. Sijbers, PhD x 2 = 2nDt , (1)
iMinds-Vision Lab, Department of Physics,
University of Antwerp, Universiteitsplein 1, with n the dimensionality of the diffusion. The
2610, Antwerp, Belgium higher the mobility of the water molecules or the

© Springer Science+Business Media New York 2016 407


W. Van Hecke et al. (eds.), Diffusion Tensor Imaging, DOI 10.1007/978-1-4939-3118-7_21
408 J. Veraart and J. Sijbers

Box 21.1  Displacement PDF another dice and repeat the experiment. Now,
If a fair dice is thrown, then it can fall six we count the sum of the eyes of both dice.
ways. When counting the different outcomes Obviously, it is more likely to throw a seven
of throwing that dice many times, one will than a two. Indeed, there are six combinations
observe that any outcome is equally probable. that will results in a seven, whereas only one
A visual representation might help to grasp combination gives two. All combinations are
that the probability of throwing one is equal to equally likely to be thrown. So, one might
the probability of throwing two, three, four, expect to count six times more sevens than
five, or six. A bar plot showing the different twos. Hence, the PDF will no longer be uni-
counts divided by the total number of throws form. Instead, it will be a bell-shaped distribu-
is given in Fig. 21.1. This graph shows for tion with its peak at seven. By adding more
each possible outcome the probability of dice to the experiment, it will eventually
being thrown during a single experiment. The become a Gaussian distribution. Knowing the
mathematical relation between the possible PDF might be important as it allows predict-
outcomes (discrete or continuous) and the ing the outcome of an experiment with some
respective probabilities is called the probabil- probability.
ity distribution function (PDF). In case of a Similarly, rather than counting eyes on
single dice, the PDF will be uniform because dice, one can also measure the distance trav-
all outcomes are equally probable. Let us add eled by a molecule given a diffusion time.

Fig. 21.1 (Left–Middle–Right) The probabilities of throwing n eyes with 1, 2, and 5 dice, respectively, are shown

longer the diffusion time, the wider the Gaussian large chambers of the ventricular system.
distribution—or the greater the average traveled However, biological tissues such as the brain
distance will be. white matter are highly heterogeneous media that
consist of various individual compartments (e.g.,
intracellular, extracellular, neurons, glial cells,
Hindered and Restricted Diffusion and axons) and barriers (e.g., cell membranes and
myelin sheaths). Therefore, the random move-
Obviously a glass of water is a poor model to ment of water molecules is hindered and/or
describe the diffusion in biological tissue. Given restricted by compartmental boundaries and
typical diffusion times in diffusion-weighted other molecular obstacles (see Fig. 21.2). There
MRI—about 50–100 ms—free diffusion can is no doubt that molecules’ mobility is reduced
only be expected in the cerebrospinal fluid in the by their interactions with compartments and
21  Diffusion Kurtosis Imaging 409

Fig. 21.2 Schematical
representation of
hindered (green) and
restricted diffusion (red).
The black circles represent
impermeable boundaries

b­arriers. However, diffusion is only termed


restricted if molecules that are confined in a
MEAN SQUARED DISPLACEMENT

bounding structure, which they are not likely to N


SIO
N

leave, collide with this structural boundary dur- FU


SIO

F
ing the diffusion time. Typically, the diffusion of DI
ED
FU

R
water molecules confined within the intra-axonal DE
DIF

N
spaces is expected to be restricted. Indeed, given HI
EE
FR

a diffusion time of 50 ms, a freely diffusing water


molecule would displace on average 25 μm
whereas the diameter of myelinated axons varies
between 1 and 20 μm. RESTRICTED DIFFUSION
Like free diffusion, hindered diffusion can
still be described by a Gaussian distribution.
However, the width of the distribution will be DIFFUSION TIME
smaller than one might expect based on proper-
Fig. 21.3  Mean squares displacements are shown as a
ties of the tissue water itself. It is common to function of the diffusion time for free (blue), hindered
refer to the observed diffusion coefficient as the (green), and restricted (red) diffusion
apparent diffusion coefficient (ADC; Dapp) to
indicate that the diffusion coefficient strongly
depends on interactions of the diffusing mole- clearly indicates the difference between the dif-
cules with the underlying microstructure, rather ferent types of diffusion processes (see Fig. 21.3).
than on intrinsic diffusion properties [3]. In case of free and hindered diffusion, the mean
Restricted diffusion, on the other hand, can no squared displacement linearly increases with the
longer accurately be described by a Gaussian dis- diffusion time. The coefficient representing this
tribution [4–9]. Plotting the mean squared dis- linear relation is given by the diffusion coeffi-
placement as a function of the diffusion time cient, multiplied by 2n, with n the dimension.
410 J. Veraart and J. Sijbers

For free diffusion, the diffusion coefficient is


larger than for hindered diffusion. Indeed, for
the same diffusion time, hindered molecules
will have displaced less than free diffusing mol-
ecules. For restricted diffusion, on the other
hand, the mean squared displacement converges
to an upper bound, which relates to the size of
the bounding microstructure. Given the size of a
typical diffusion-­ weighted MRI voxel (about
2 × 2 ×2–3 × 3 × 3  mm3), the biological tissue
within a voxel is expected to contain a mixture of
hindered and restricted compartments [10, 11].
Therefore, the primary assumption made in diffu-
sion tensor imaging, i.e., Gaussian diffusion [12],
does not hold in most voxels [13]. Hence, a more
advanced diffusion model is required, that, apart
from the diffusion coefficient, also describes the Fig. 21.4  Distributions with varying kurtosis, but with
deviation from a Gaussian distribution. A distri- the same mean and variance are shown
bution’s deviation from a Gaussian distribution
can be quantified by the excess kurtosis, hereaf- Gaussian diffusion. For diffusion, a kurtosis of
ter shortened to kurtosis [14]. The knowledge of −3/7 is a practical minimum [15]. Such a distri-
the kurtosis of the displacement distribution in bution would indicate fully restricted diffusion
addition to its variance improves the description within spherical pores with the same radius.
of the underlying diffusion process [15]. In the However, fully restricted diffusion is not
next paragraph, the kurtosis of an arbitrary distri- expected in biological tissue. As stated previ-
bution is briefly introduced. ously, one rather expects the imaged volume to
consist of a mixture of hindered and restricted
compartments [10, 11]. Therefore, it is generally
Kurtosis assumed that the kurtosis will only take positive
values in diffusion-­weighted MRI [16].
The kurtosis K is a dimensionless statistical met- It is clear that the knowledge of the kurtosis
ric that quantifies the deviation from Gaussianity definitely contributes to a more accurate descrip-
of an arbitrary distribution. In Fig. 21.4, a num- tion of the underlying diffusion process. In the
ber of distributions that have the same mean next section, it will be explained how kurtosis
(which is zero) and variance (which is one), but parameters can be computed from diffusion-­
different kurtosis are shown. Indeed, the weighted MR images.
(excess) kurtosis varies from −1.2 to 3. One can
observe that the kurtosis is a measure of peaked-
ness or sharpness of an arbitrary distribution. Diffusion Kurtosis Coefficient
Given that a Gaussian distribution has zero kur-
tosis, a positive kurtosis indicates that distribu- The natural logarithm of the diffusion weighted
tion is more peaked than a Gaussian distribution. MR signal, S, can be approximated by an expan-
In terms of diffusion, small displacements are sion in terms of the b-value [15]:
more probable compared to hindered diffusion. A
1
negative kurtosis, on the other hand, is less log S (b) » log S (0) - bDapp + b 2 Dapp
2
K app , (2)
6
peaked than a Gaussian distribution. If the diffu-
sion is described by such a distribution, small with S(0) the nondiffusion-weighted signal, Dapp
displacements are less likely compared to the apparent diffusion coefficient and Kapp the
21  Diffusion Kurtosis Imaging 411

Fig. 21.5  Following diffusion-weighted signals (left), as the accuracy of the fit. This is mainly noticeable at inter-
well as their log-transformation (right) are shown as a mediate b-values. At high b-values, severe approximation
function of the b-value: measured values (red dots), DTI errors become dominant. Therefore, DKI is a low to inter-
model (blue), and DKI model (green). Owing to the non-­ mediate b-value technique
Gaussian diffusion the addition of the b2-term improves

apparent kurtosis coefficient. Note that the term kurtosis coefficient. Indeed, to accurately model
apparent kurtosis coefficient is used in analogy to hindered diffusion, a three-dimensional (3D)
the apparent diffusion coefficient to indicate the Gaussian diffusion model that relies on a second
dependency of the measured parameter to mea- order, symmetric diffusion tensor D, instead of
surement variables such as diffusion time. If dif- the scalar Dapp, is needed [19]. This widely used
fusion is assumed to be Gaussian, or equivalently, diffusion tensor imaging (DTI) model has six
to have zero kurtosis, then the last term nullifies degrees of freedom, describing the shape and ori-
and the equation reduces to the basic DTI for- entation of a 3D ellipsoid. Like the directional
mula [12]. Note that the additional kurtosis term dependence of Dapp can be captured by a diffusion
depends on b squared. Hence, unlike Gaussian tensor, the directional dependence of Kapp can be
diffusion, the logtransformed diffusion-weighted represented by a tensor as well: the diffusion kur-
signal will not decay linearly with the b-value tosis tensor W [15, 20]. This fourth rank 3D ten-
(Fig. 21.5). Alternatively, one can say that the sor, which is fully symmetric, has only 15
diffusion-weighted signal will decay non-mono-­ components that are independent. The general-
exponentially. Just as Dapp characterizes the diffu- ization of Eq. (2) to 3D results in:
sion coefficient in the direction parallel to the
log S (b, g ) » log S (0) - båi =1 å j =1 gi g j Dij
3 3
orientation of diffusion sensitizing gradients, Kapp
characterizes the diffusional kurtosis in the same 1 æ1 3 ö 3
+ b 2 ç åi =1 Dii ÷ åi =1 å j =1
3
direction. 6 è3 ø
åk =1 ål =1 gi g j g k glWijkl , (3)
3 3

Diffusion Kurtosis Tensor


with g = [ g x , g y , g z ] the applied diffusion gradi-
In the brain white matter, molecular diffusion is ent direction. One immediately recognizes the
more likely to be hindered and restricted perpen- DTI model as being the first two terms on the
dicular to the axonal fibers than parallel to them right-hand side of Eq. (3) [19]. Fitting this model
because of the geometry of the underlying micro- voxelwise to a set of diffusion MR images to esti-
structure [17, 18]. Therefore, the diffusion is mate D and W, and as such, directly quantifying
anisotropic and, as such, the diffusion cannot be the direction-dependent diffusion and kurtosis
described adequately by a single diffusion and information, is called Diffusion Kurtosis Imaging
412 J. Veraart and J. Sijbers

[15, 21, 22]. The technique is a straightforward ­ etrics of diffusional non-Gaussianity, comple-
m
mathematical extension of DTI as the cumulant mentary to the diffusion metric obtained with
expansion framework overarches both models DTI [15].
[23]. Given both diffusional tensors, the apparent The most commonly used kurtosis metrics are
diffusion and kurtosis coefficients along an arbi- mean kurtosis, radial kurtosis, and axial kurtosis.
trary direction can be evaluated to study the dif- The axial kurtosis is the evaluation of the appar-
fusion properties in any direction. ent kurtosis tensor along the principle diffusion
direction [24]. The radial kurtosis is the average
apparent kurtosis coefficient, measured in the
Diffusion Kurtosis Parameters equatorial plane [25], whereas the mean kurtosis
is the overall average apparent diffusion kurtosis
In DTI, the principle axes of the ellipsoidally coefficient [20]. Furthermore, a kurtosis anisot-
shaped diffusion tensor and their corresponding ropy metric has been proposed [25]. The comple-
lengths are determined by the eigenvectors and mentariness of the kurtosis and diffusion
eigenvalues of the diffusion tensor D [19]. The measures is indicated in the scatter plots of
first eigenvalue equals the diffusivity along the Fig.  21.6, which show the weak correlation
main direction of diffusion, and is called axial between directional diffusion and kurtosis met-
diffusivity. The average of the second and third rics, observed in the white matter of the healthy
eigenvalue is called the radial diffusivity. The human brain (cf. [15]). This implies that two vox-
measure quantifies the average diffusivity in the els with equal mean diffusivity not necessarily
equatorial plane, i.e., the plane perpendicular to have the same mean kurtosis. As such, kurtosis
the principal diffusion direction. The average of measures provide additional information regard-
all three eigenvalues is the mean diffusivity. ing the underlying diffusion process. Typical val-
Another rotationally invariant scalar measure is ues of the DKI metrics for the healthy human
the fractional anisotropy. It quantifies the degree brain are presented by Lätt et al. [26]. The param-
of anisotropy of the apparent diffusion tensor eter maps are shown in Fig. 21.7. The diffusion
[12]. On the one hand, DKI provides a more kurtosis metrics are potentially more sensitive to
objective and accurate quantification of these local (microstructural) tissue properties [15].
scalar metrics in the sense that the dependence of Furthermore, it has been shown that the diffusion
the estimated diffusivity b-value is eliminated or kurtosis metrics are less sensitive to certain con-
at least strongly reduced [13]. On the other hand, founding effects and thereby serve as a more
it provides additional rotationally invariant robust biomarker. One study, for example,

Fig. 21.6  Scatter plots show the correlation between and axial diffusivity. The corresponding metrics are only
(left) mean kurtosis and mean diffusivity, (middle) radial weakly correlated. The Spearman’s rank correlation coef-
kurtosis and radial diffusivity, and (right) axial kurtosis ficients are −0.07, −0.65, and −0.13, respectively
21  Diffusion Kurtosis Imaging 413

Fig. 21.7  The main diffusion and kurtosis parameter 3 × 10−3] mm2/s, while the range of the corresponding kur-
maps, obtained from the healthy human brain, are shown. tosis metrics was [0, 1.5]. The anisotropy maps are
The range of the mean, axial and radial diffusivity is [0, bounded by [0, 1]

showed that the mean kurtosis in gray matter is the applied diffusion gradient. Typically, the
altered substantially less by CSF contamination highest b-value is somewhat higher than in DTI
than either of the conventional DTI metrics [27]. acquisitions. Indeed, the maximal b-value should
be chosen carefully as it defines a trade-off
between the accuracy and precision of diffusion
Diffusion Kurtosis Imaging: parameter estimators. While for DTI diffusion-­
Acquisition weighted images are typically acquired with
rather low b-values, about 1000 s/mm2, some-
DKI is a straightforward extension of DTI in what stronger diffusion sensitizing gradients need
terms of data acquisition. Indeed, the same to be applied for DKI as the quadratic term in the
weighted imaging sequences can be b-value needs to be apparent. It has been shown
diffusion-­
used to record the images. However, since the that b-values of about 2000 s/mm2 are sufficient
apparent diffusion tensor has 6 independent ele- to measure the degree of non-Gaussianity with an
ments and the kurtosis tensor has 15 independent acceptable precision [21]. Nevertheless, several
elements, the DKI model has a total of 21 inde- studies reported b-values up to 3000 s/mm2 and
pendent tensor parameters. As for DTI, the noise-­ even more (e.g., [28, 29]). The assumption that
free nondiffusion-weighted signal must be the diffusion-weighted signal is monotonically
estimated as well. Hence, at least 22 diffusion-­ decreasing with the b-value imposes an analytical
weighted images need to be acquired for DKI. upper bound on the maximal b-value [30]:
Let us recall that for DTI only seven diffusion-­ bmax £ 3 / ( Dapp K app ). (4)
weighted images were required. It can further be
shown that there must be at least three distinct Indeed, as can be seen in Fig. 21.5, the DKI
b-values, which only differ in the magnitude of model has a global minimum at b = bmax. For
414 J. Veraart and J. Sijbers

larger b-values the diffusion-weighted signals  iffusion Kurtosis Imaging: Post


D
predicted by the DKI model start increasing Processing
again. This is in disagreement with the assump-
tion that the diffusion-weighted signals keep In general, data correction is the first step of the
decaying with increasing b-value. Therefore, the diffusion MR data post-processing pipeline.
DKI model is only accurate in a limited b-value First, the relatively long acquisition times cause
range. The upper bound of that range is difficult subject motion to become more probable, and, as
to determine. However, typical diffusion and kur- such, neccessitate the alignment of all diffusion-­
tosis values, observed in the human brain, are weighted images before the model parameters
Dapp ≈ 1 μm2/ms and Kapp ≈ 1. Those values justify are estimated [31]. Commonly, such a correction
the use of b-values up to 3000 s/mm2 for studies is applied by a single rigid transformation of the
involving the human brain [21]. Given the lim- diffusion-weighted images to a single reference
ited maximal gradient magnitude, such b-values image, often a nondiffusion-weighted image.
are often achieved by increasing the gradient The use of a rigid transformation is motivated by
duration. As the correctness of the DKI model the expected lack of scaling and shearing of the
relies on the short gradient pulse (SGP) condi- volumes, whereas translations and rotations are
tion, the use of high b-values will render the mea- assumed to be global for brain imaging. However,
sured apparent kurtosis metrics more approximate note that other diffusion applications might
[15]. Furthermore, high b-valued diffusion- require more complex motion correction strate-
weighted images suffer from low signal-to-noise gies. To preserve the orientation information
ratio (SNR) due to the severe signal attenuation. captured in diffusion MR data, the diffusion gra-
Since SNR has a direct impact on the precision of dient directions need to be rotated accordingly
the diffusion quantification, the acquisition of [32]. Second, rapidly switching diffusion gradi-
diffusion MR images along more diffusion direc- ents will generate eddy currents in nearby con-
tions than strictly necessary is advisable. ductors. These currents will perturb the spatial
Although in theory only 15 distinct diffusion encoding locally and, as such, the reconstructed
(gradient) directions are required [15], in prac- diffusion-­weighted image will be geometrically
tice, a minimum of 30 directions for each b-value distorted [33]. The strength of the distortions—
is fairly common [21]. A wide range of DKI data stretch or compression of the image along the
acquisition protocols in line with these consider- phase encoding direction—increases with the
ations are possible. Depending on the set of diffu- diffusion encoding amplitude. Although the
sion parameters one is interested in, a specific effect of eddy currents can be minimized at the
acquisition protocol (i.e. b-values and gradient acquisition stage [34], residual distortions still
directions) that is optimal in terms of highest need correction. A common strategy is to correct
achievable precision on the measurements of subject motion and eddy current distortions
interest can be computed [25]. In Table 21.1, the simultaneously by a global affine transforma-
minimal acquisition requirements for DKI are tion. Recently, several techniques dedicated to
listed and compared to DTI. motion/eddy current correction of high b-valued
diffusion-­weighted images have been presented
Table 21.1  Minimal acquisition requirements for DTI [35, 36]. Such corrections need to be followed
and DKI by signal modulation according to the volumet-
DTI DKI ric change [37, 38]. The signal modulation factor
Number of diffusion-weighted 7 22 will be b-value dependent as the strength of the
images distortions depends on the magnitude of the
Number of b-values 2 3 diffusion-­encoding gradient. Not correcting such
b-value range (s/mm2) [0–1500] [0–3000] a b-value dependent signal change will bias the
Noncoplanar gradient 6 15 kurtosis parameters [13, 39]. Nonetheless, the
directions signal modulation step is widely ignored, partly
21  Diffusion Kurtosis Imaging 415

motivated by a lack of this post-processing step ringing, or misalignment of the diffusion-­


in most software packages. weighted images [40]. It is well known that the
After data correction—and optional local diffusivity should be positive along all directions.
smoothing to reduce the Gibbs phenomena—the This condition holds if the apparent diffusion ten-
DKI model parameters, i.e., the tensor elements, sor is positive definite. In DTI, one often imposes
need to be estimated. The most popular strategies the non-negative definiteness constraint on the
are nonlinear least squares (NLS), weighted lin- diffusion tensor [43]. The same constraint can be
ear least squares (WLLS), and linear least squares imposed during a DKI analysis. In addition, two
(LLS). Technical details on the implementation constraints on the apparent kurtosis coefficients
of these methods are provided in [40, 41]. It is might be imposed. First, while the theoretical
important to grasp that not all estimation strate- lower bound on the kurtosis of a probability dis-
gies show the same performance in terms of tribution is minus two, previous studies suggest
accuracy and precision [37, 38, 41]. Typically, that directional kurtoses should typically be
the WLLS estimator might be preferred to the ­positive [16, 40]. Second, an upper bound on the
other estimators [41]. First, within the class of kurtosis that depends on the maximal b-value, cf.
linearized strategies, the WLLS estimator is defi- Eq. (4), is often imposed during the estimation of
nitely the better choice as it accounts for the sig- the DKI parameters.
nal dependency of the variance in the
log-transformed diffusion-weighted data [42].
Therefore, the WLLS estimator will show higher  iffusion Kurtosis Imaging:
D
precision than the LLS estimator. Second, the Applications
WLLS estimator is potentially very accurate,
especially compared to the NLS estimator. Despite DKI being a recently developed tech-
Indeed, if Rician distributed diffusion-weighted nique, an exponential growth of publications
MR data has a minimum SNR of two, then the already suggests that DKI will become a new
WLLS estimator is unbiased, that is, the estima- important imaging modality in detecting micro-
tor has no systematic estimation error [41]. Third, structural changes in human living tissue that are
the WLLS estimator has a closed-form solution. not revealed by the Gaussian DTI model.
Therefore, unlike the iterative NLS strategy, the Preliminary, yet promising results have shown
WLLS estimator is computationally efficient and that DKI measures may be able to better differen-
not prone to getting stuck in a local optimum. In tiate between high-grade and low-grade cerebral
practice, however, the SNR might drop below 2 gliomas than DTI measures alone [44, 45].
because of high b-values or high spatial resolu- Furthermore, clinical studies indicate that DKI
tions. In that case, the above-mentioned estima- has the potential to improve the early diagnosis
tors will show a systematic overestimation of the of, or to gain more insight into pathologies such
kurtosis parameters and, as such, one might pre- as Parkinson Disease [29, 46], attention-deficit
fer using more advanced estimators that explic- hyperactivity disorder [47], temporal lobe epi-
itly account for the actual MR data distribution lepsy [48], traumatic brain [49], Alzheimer’s dis-
[38, 39]. An overview of these estimators, their ease [50], and cerebral infarction [51–53].
strengths, and limitations is beyond the scope of However, its potential use is not restricted to the
this introductory chapter. brain. The greater relative contrast of kurtosis
As the diffusion of water molecules is a physi- metrics for cancerous sextants also suggests the
cal property of the tissue being measured, diffu- potential clinical advantage of incorporating
sional tensor estimates must be physically and DKI into liver and prostate MR imaging protocol
biologically meaningful. Unfortunately, in many [54, 55]. Additionally, one study has reported on
cases, diffusion and kurtosis values might lie out- the sensitivity of the DKI metrics to abnormali-
side a physically acceptable range due to the ties in the lung, i.e., within the bronchioles and
presence of noise, imaging artifacts such as Gibbs bronchi, using hyperpolarized 3HE imaging [56].
416 J. Veraart and J. Sijbers

Furthermore, microstructural changes associated an intra-axonal space, consisting of parallel


with human development and aging have been impermeable cylindrical axons and an extra-­
studied with DKI [57]. This study showed differ- axonal space. The diffusion in both compart-
ent mean kurtosis patterns for different age ments is assumed to be anisotropic and Gaussian.
ranges, indicating that DKI is able to detect The white matter model links the DKI metrics to
changes in microstructural complexity in both microstructural properties such as the axonal
gray and white matter. In complementary water fraction and the tortuosity of the extra-­
research, the practical utility of DKI for the axonal space. In another attempt to gain insight
(early) diagnosis of pathological changes has in the mathematical DKI model, the DKI infor-
been studied using small animal imaging. For mation was matched to the information extracted
example, it has been shown that DKI improved from the biophysical composite hindered and
the early detection of ischaemic lesions in a restricted model of diffusion (CHARMED)model
stroke model for rats, compared to DTI [28]. [10, 11, 16]. In that model, the white matter is
Moreover, DKI may help stratify heterogeneous again assumed to be consisting of two
diffusion-weighted MRI lesions for enhanced ­compartments: (a) a hindered extra-axonal space,
characterization of ischemic tissue injury [58]. and (b) one or more intra-axonal compartments
Studies have also reported on the increased sensi- modeled as impermeable cylinders showing
tivity of kurtosis metrics to changes in the white restricted diffusion perpendicular to the fiber.
and gray matter in rodent models of Huntington The CHARMED model allows the description of
Disease [59, 60], Chronicle mild stress [61], the diffusion weighted-signals in terms of bio-
Alzheimer’s disease [62], traumatic brain injury physical parameters such as extra- and intra-axo-
[63], and brain maturation [64]. It might be nal volume fractions and axonal diffusivities. It
expected that many new potential applications of was shown that those biophysical parameters cor-
DKI will be revealed in the near future. relate with the DKI parameters in areas of higher
intra-voxel directional coherence, and as such,
the CHARMED model might be used the get
 iffusion Kurtosis Imaging:
D more insight into the meaning of the DKI param-
Limitations eters [16]. Those findings, however, only apply
within the limits of the validity of both white
The potential risk of DKI—just as DTI—is the matter models.
over-interpretation of observed changes in diffu-
sional measures. The kurtosis model arises sim-
ply from a mathematical expansion of the References
diffusion-weighted signal as a function of the
b-value and, as such, does not involve any bio- 1. Brown R. A brief account of microscopical observa-
tions made in the months of June, July and August,
physical modeling [16, 65, 66]. From a change in 1827, on the particles contained in the pollen of
kurtosis or diffusivity, one might only conclude plants; and on the general existence of active mole-
that there is something in the tissue microstruc- cules in organic and inorganic bodies. Philos Mag.
ture that is changing the way that molecules can 1828;4:161–1763.
2. Einstein A. Über die von der molekularkinetischen
diffuse. More specific inferences are not substan- Theorie der Wärme geforderte Bewegung von in
tiated without the justification of a biophysical ruhenden Flüssigkeiten suspendierten Teilchen. Ann
model that helps to interpret the biophysical Phys. 1905;322(8):549–60.
meaning of DKI metric changes. Recently, the 3. Le Bihan D, et al. MR imaging of intravoxel incoher-
ent motions: application to diffusion and perfusion in
two-tensor model has been studied to elucidate neurologic disorders. Radiology. 1986;161(2):401–7.
the underpinnings of DKI contrast [65, 67]. In 4. Assaf Y, et al. Non-mono-exponential attenuation of
that model, it is assumed that brain white matter water and N-Acetyl aspartate signals due to diffusion
consists of two non-exchanging compartments: in brain tissue. J Magn Reson. 1998;131(1):69–85.
21  Diffusion Kurtosis Imaging 417

5. Beaulieu C, et al. Determinants of anisotropic water 24. Hui ES, et al. Towards better MR characterization of
diffusion in nerves. Magn Reson Med. 1994;31(4): neural tissues using directional diffusion kurtosis
394–400. analysis. Neuroimage. 2008;42(1):122–34.
6. King MD, et al. q-space imaging of the brain. Magn 25. Poot DHJ, et al. Optimal experimental design for dif-
Reson Med. 1994;32(6):707–13. fusion kurtosis imaging. IEEE Trans Med Imaging.
7. Niendorf T, et al. Biexponential diffusion attenuation 2010;29(3):819–29.
in various states of brain tissue: implications for 26. Lätt J, et al. Regional values of diffusional kurtosis
diffusion-­weighted imaging. Magn Reson Med. estimates in the healthy brain. J Magn Reson Imaging.
1996;36(6):847–57. 2013;37(3):610–8.
8. Stanisz GJ, et al. An analytical model of restricted dif- 27. Yang AW, et al. Effect of cerebral spinal fluid suppres-
fusion in bovine optic nerve. Magn Reson Med. sion for diffusional kurtosis imaging. J Magn Reson
1997;37(1):103–11. Imaging. 2013;37(2):365–71.
9. Stanisz GJ, et al. Diffusional anisotropy of T2 compo- 28. Grinberg F, et al. Diffusion kurtosis imaging and log-­
nents in bovine optic nerve. Magn Reson Med. normal distribution function imaging enhance the
1998;40(3):405–10. visualisation of lesions in animal stroke models. NMR
10. Assaf Y, et al. New modeling and experimental frame- Biomed. 2012;25(11):1295–304.
work to characterize hindered and restricted water dif- 29. Wang J-J, et al. Parkinson disease: diagnostic utility
fusion in brain white matter. Magn Reson Med. of diffusion kurtosis imaging. Radiology. 2011;261(1):
2004;52(5):965–78. 210–7.
11. Assaf Y, et al. Composite hindered and restricted 30. Lazar M, et al. Estimation of the orientation distribu-
model of diffusion (CHARMED) MR imaging of the tion function from diffusional kurtosis imaging. Magn
human brain. Neuroimage. 2005;27(1):48–58. Reson Med. 2008;60(4):774–81.
12. Basser PJ. Inferring microstructural features and the 31. Jenkinson M, et al. A global optimisation method for
physiological state of tissues from diffusion-weighted robust affine registration of brain images. Med Image
images. NMR Biomed. 1995;8(7-8):333–4. Anal. 2001;5(2):143–56.
13. Veraart J, et al. More accurate estimation of diffusion 32. Leemans A, et al. The B-matrix must be rotated when
tensor parameters using diffusion Kurtosis imaging. correcting for subject motion in DTI data. Magn
Magn Reson Med. 2011;65(1):138–45. Reson Med. 2009;61(6):1336–49.
14. Balanda KP, MacGillivray HL, et al. Kurtosis: a criti- 33. Horsfield M. Mapping eddy current induced fields for
cal review. Am Stat. 1988;42(2):111–9. the correction of diffusion-weighted echo planar
15. Jensen JH, et al. Diffusional kurtosis imaging: the images. Magn Reson Imaging. 1999;17(9):1335–45.
quantification of non-gaussian water diffusion by 34. Reese TG, et al. Reduction of eddy-current-induced
means of magnetic resonance imaging. Magn Reson distortion in diffusion MRI using a twice-refocused
Med. 2005;53(6):1432–40. spin echo. Magn Reson Med. 2003;49(1):177–82.
16. De Santis S, et al. Using the biophysical CHARMED 35. Andersson J et al. 2012. A comprehensive Gaussian
model to elucidate the underpinnings of contrast in process framework for correcting distortions and
diffusional kurtosis analysis of diffusion-weighted movements in diffusion images. In Proceedings of the
MRI. MAGMA. 2012;25(4):267–76. international society for Magnetic Resonance in
17. Chenevert TL, et al. Anisotropic diffusion in human Medicine, p. 2426
white matter: demonstration with MR techniques 36. Ben-Amitay S, et al. Motion correction and registra-
in vivo. Radiology. 1990;177(2):401–5. tion of high b-value diffusion weighted images. Magn
18. Moseley ME, et al. Anisotropy in diffusion-weighted Reson Med. 2012;67(6):1694–702.
MRI. Magn Reson Med. 1991;19(2):321–6. 37. Jones DK, et al. Twenty-five pitfalls in the analysis of
19. Basser PJ, et al. MR diffusion tensor spectroscopy diffusion MRI data. NMR Biomed. 2010;23(7):
and imaging. Biophys J. 1994;66(1):259–67. 803–20.
20. Lu H, et al. Three-dimensional characterization of 38. Veraart J, et al. Comprehensive framework for accu-
non-gaussian water diffusion in humans using diffu- rate diffusion MRI parameter estimation. Magn Reson
sion kurtosis imaging. NMR Biomed. 2006;19(2): Med. 2013;81(4):972–84.
236–47. 39. Veraart J, et al. Constrained maximum likelihood esti-
21. Jensen JH, et al. MRI quantification of non-Gaussian mation of the diffusion kurtosis tensor using a Rician
water diffusion by kurtosis analysis. NMR Biomed. noise model. Magn Reson Med. 2011;66(3):678–86.
2010;23(7):698–710. 40. Tabesh A, et al. Estimation of tensors and tensor-­
22. Wu EX, et al. MR diffusion kurtosis imaging for neu- derived measures in diffusional kurtosis imaging.
ral tissue characterization. NMR Biomed. 2010;23(7): Magn Reson Med. 2011;65(3):823–36.
838–48. 41. Veraart J, et al. Weighted linear least squares estima-
23. Kiselev VG. The cumulant expansion: an overarching tion of diffusion MRI parameters: strengths, limita-
framework for understanding diffusion MRI. In: tions, and pitfalls. Neuroimage. 2013;81:335–46.
Jones DK, editor. Diffusion MRI: theory, methods and 42. Salvador R, et al. Formal characterization and exten-
applications. Oxford: Oxford University Press; 2010. sion of the linearized diffusion tensor model. Hum
p. 152–68. Brain Mapp. 2005;24(2):144–55.
418 J. Veraart and J. Sijbers

43. Koay CG, et al. A unifying theoretical and algorithmic for detection and assessment of aggressiveness of
framework for least squares methods of estimation in peripheral zone cancer. Radiology. 2012;264(1):
diffusion tensor imaging. J Magn Reson. 126–35.
2006;182(1):115–25. 56. Trampel R, et al. Diffusional kurtosis imaging in the
44. Van Cauter S, et al. Gliomas: diffusion kurtosis MR lung using hyperpolarized 3He. Magn Reson Med.
imaging in grading. Radiology. 2012;263(2):492–501. 2006;56(4):733–7.
45. Raab P, et al. Cerebral gliomas: diffusional kurtosis 57. Falangola MF, et al. Age-related non-gaussian diffu-
imaging analysis of microstructural differences. sion patterns in the prefrontal brain. J Magn Reson
Radiology. 2010;254(3):876–81. Imaging. 2008;28(6):1345–50.
46. Giannelli M, et al. Diffusion kurtosis and diffusion-­ 58. Cheung JS, et al. Stratification of heterogeneous dif-
tensor MR imaging in Parkinson disease. Radiology. fusion MRI ischemic lesion with kurtosis imaging:
2012;265(2):645–6. author reply 646–7. evaluation of mean diffusion and kurtosis MRI mis-
47. Helpern J, et al. Preliminary evidence of altered gray match in an animal model of transient focal ischemia.
and white matter microstructural development in the Stroke. 2012;43(8):2252–4.
frontal lobe of adolescents with attention-deficit 59. Blockx I, et al. Identification and characterization of
hyperactivity disorder: a diffusional kurtosis imaging Huntington related pathology: an in vivo DKI imag-
study. J Magn Reson Imaging. 2011;33(1):17–23. ing study. Neuroimage. 2012;63(2):653–62.
48. Gao Y, et al. Diffusion abnormalities in temporal
60. Blockx I, et al. Microstructural changes observed with
lobes of children with temporal lobe epilepsy: a pre- DKI in a transgenic Huntington rat model: evidence
liminary diffusional kurtosis imaging study and com- for abnormal neurodevelopment. Neuroimage.
parison with diffusion tensor imaging. NMR Biomed. 2012;59(2):957–67.
2012;25(12):1369–77. 61. Delgado y Palacios R, et al. Magnetic resonance

49. Grossman EJ, et al. Thalamus and cognitive impairment imaging and spectroscopy reveal differential hippo-
in mild traumatic brain injury: a diffusional kurtosis campal changes in anhedonic and resilient subtypes of
imaging study. J Neurotrauma. 2012;29(13):2318–27. the chronic mild stress rat model. Biol Psychiatry.
50. Gong N-J, et al. Correlations between microstructural 2011;70(5):449–57.
alterations and severity of cognitive deficiency in 62. Zhang L, et al. Current neuroimaging techniques in
Alzheimer’s disease and mild cognitive impairment: a Alzheimer’s disease and applications in animal mod-
diffusional kurtosis imaging study. Magn Reson els. Am J Nucl Med Mol Imaging. 2012;2(3):
Imaging. 2013;31(5):688–94. 386–404.
51. Jensen JH, et al. Preliminary observations of increased 63. Zhuo J, et al. Diffusion kurtosis as an in vivo imaging
diffusional kurtosis in human brain following recent marker for reactive astrogliosis in traumatic brain
cerebral infarction. NMR Biomed. 2011;24(5): injury. Neuroimage. 2012;59(1):467–77.
452–7. 64. Cheung MM, et al. Does diffusion kurtosis imaging
52. Hori M, et al. A new diffusion metric, diffusion kurto- lead to better neural tissue characterization? A rodent
sis imaging, used in the serial examination of a patient brain maturation study. Neuroimage. 2009;45(2):
with stroke. Acta Radiol Short Rep. 2012;1(12):1–3. 386–92.
53. Hui ES, et al. Stroke assessment with diffusional kur- 65. Fieremans E, et al. White matter characterization with
tosis imaging. Stroke. 2012;43(11):2968–73. diffusional kurtosis imaging. Neuroimage. 2011;58(1):
54. Rosenkrantz AB, et al. Assessment of hepatocellular 177–88.
carcinoma using apparent diffusion coefficient and 66. Nilsson M, et al. The role of tissue microstructure and
diffusion kurtosis indices: preliminary experience in water exchange in biophysical modelling of diffusion
fresh liver explants. Magn Reson Imaging. 2012; in white matter. MAGMA. 2013;26(4):345–70.
30(10):1534–40. 67. Fieremans E, et al. Monte Carlo study of a two-­

55. Rosenkrantz AB, et al. Prostate cancer: feasibility and compartment exchange model of diffusion. NMR
preliminary experience of a diffusional kurtosis model Biomed. 2010;23(7):711–24.
HHS Public Access
Author manuscript
Exp Neurol. Author manuscript; available in PMC 2016 October 01.
Author Manuscript

Published in final edited form as:


Exp Neurol. 2015 October ; 272: 88–96. doi:10.1016/j.expneurol.2015.05.011.

Diffusion Tensor Imaging in Hemorrhagic Stroke


Neeraj Chaudhary1,2, Aditya S Pandey1,2, Joseph J Gemmete1,2, Ya Hua2, Yining Huang3,
Yuxiang Gu4, and Guohua Xi2
1Department of Radiology, University of Michigan, Ann Arbor
2Department of Neurosurgery, University of Michigan, Ann Arbor
3Department of Neurology, the 1st Affiliated Hospital, Peking University, Beijing
Author Manuscript

4Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai

Abstract
Diffusion Tensor Imaging (DTI) has evolved considerably over the last decade to now be
knocking on the doors of wider clinical applications. There have been several efforts over the last
decade to seek valuable and reliable application of DTI in different neurological disorders. The
role of DTI in predicting outcomes in patients with brain tumors has been extensively studied and
has become a fairly established clinical tool in this scenario. More recently DTI has been applied
in mild traumatic brain injury to predict clinical outcomes based on DTI of the white matter tracts.
The resolution of white matter fiber tractography based on DTI has improved over the years with
increased magnet strength and better tractography post processing. The role of DTI in
Author Manuscript

hemorrhagic stroke has been studied preliminarily in the scientific literature. There is some
evidence that DTI may be efficacious in predicting outcomes of motor function in animal models
of intracranial hemorrhage. Only a handful of studies of DTI have been performed in subarachnoid
hemorrhage or intraventricular hemorrhage scenarios. In this manuscript we will review the
evolution of DTI, the existing evidence for its role in hemorrhagic stroke and discuss possible
application of this non-invasive evaluation technique of human cerebral white matter tracts in the
future.

Keywords
cerebral hemorrhage; diffusion Tensor Imaging; subarachnoid hemorrhage
Author Manuscript

Corresponding Authors: Neeraj Chaudhary, MD, MRCS, FRCR, Departments of Radiology & Neurosurgery, University of Michigan
Health System, B1D330A, 1500 E Medical Center Drive, Ann Arbor, MI – 48109-5030, Phones: Off – 734 763 2082, Admin – 734
763 5842, Fax: 734 615 5111 Or Guohua Xi, MD, Department of Neurosurgery, University of Michigan, 5018 BSRB, 109 Zina
Pitcher Place, Ann Arbor, MI 48109-2200, USA, guohuaxi@umich.edu, Phone: +1-734-764-1207, Fax: +1-734-763-7322.
Disclosure: We declare that we have no conflict of interest.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our
customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of
the resulting proof before it is published in its final citable form. Please note that during the production process errors may be
discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Chaudhary et al. Page 2

Introduction
Author Manuscript

Stroke is one of the most common causes of morbidity and mortality in the developed and
the developing world (1, 2). There are two major stroke subtypes, ischemic stroke and
hemorrhagic stroke. The commonest of them is ischemic stroke (IS) followed by
subarachnoid hemorrhage (SAH), intraventricular hemorrhage (IVH) and then intracerebral
hemorrhage (ICH) in the hemorrhagic subtype (3–6). Amongst the different types of stroke
the incidence of ICH is approximately 13% and SAH is 25–30%. In the SAH group of
patients, surgical treatment in the form of clipping or endovascular coil embolization is
performed to preclude the risk of a subsequent fatal hemorrhage. Similarly in the ICH
patients surgical evacuation is performed in large hemorrhages with mass effect. However,
currently there are very few effective therapeutic options available to reduce the
neurological dysfunction caused by the hemorrhagic insult. The understanding of the
mechanisms of neuronal injury in these clinical settings continues to evolve. Several
Author Manuscript

therapeutic options in the form of neuroprotective agents have been shown to hold promise
in the animal population but have not yet translated to show benefit in human subjects.

While better and more non-human primate models are being devised to improve the
understanding of the underlying pathophysiology of hemorrhagic stroke (7, 8), non-invasive
imaging with MRI and CT of the brain are being utilized with a view to develop biomarkers
to visualize these mechanisms of neuronal injury. Standard MRI of the brain delineates
superficial and deeper brain parenchymal structures in great detail. Then there are other
specialized sequences that have been developed that have not yet entered the routine clinical
domain. These specialized sequences are able to shed light on the normal physiology of
neuronal and axonal function. Diffusion tensor imaging or “tractography” is one such
sequence that specifically looks at directionality and integrity of the axonal fibers of the
Author Manuscript

brain and how they change in the event of neuronal injury due to various etiologies one of
them being hemorrhagic stroke.

Conventional MRI techniques are based on registering signal from a volume of tissue by
repeated rephasing and dephasing of precessing protons in the particular volume imaged.
Diffusion tensor imaging relies on slightly different property of the tissues. Water molecules
in most tissues diffuse equally in all directions (isotropic diffusion) but in white matter tracts
the diffusion is along the direction of the tract (anisotropic). Essentially the main parameter
that is measured in DTI is the degree of fractional anisotropy (FA) in a given voxel of the
precessing proton and its Eigen vector in an ellipsoid domain. In each voxel imaged by DTI
the Eigen vector direction in 3 dimensions has to be acquired in at least nine elements of the
matrix to portray the directionality of the elaborate dataset or “tensor”. At the minimum 6
Author Manuscript

directions of DTI datasets are needed to create color maps of white fiber tracts.
Mathematical calculations and data processing is then utilized to show color coded maps of
different white matter fiber tracts in the brain called fiber tractography (9). In the current
manuscript we will concentrate on reviewing the role of diffusion tensor imaging (DTI) in
hemorrhagic stroke i.e. SAH, IVH and ICH.

Exp Neurol. Author manuscript; available in PMC 2016 October 01.


Chaudhary et al. Page 3

Diffusion tensor imaging – history and development


Author Manuscript

Work on the tractographic sequences began in the 1990s. The first tractographic images
demonstrating the multi-dimensional directional information of the curved neural tracts in
the brain were obtained in 1991 by Filler et al when they submitted their patent descriptions
on imaging a series of patients in the UK (7). Subsequently they were published in the
World Intellectual Property Organization in 1993 ( 7). The very first images of DTI were
published in the proceedings of the Society for Magnetic Resonance in Medicine annual
meeting in Berlin in August 1992 (6, 7). Prior to 1992 the clinical community believed that
the strategy of application of diffusion MRI to trace brain white matter tracts did not have
any potential. The discoveries of Filler, Howe and Richards in London, England and Seattle
demonstrated the feasibility of diffusion MRI in producing linear neural images of the white
matter tracts of the brain in late 1991 and then was reaffirmed by investigators LeBihan and
Basser in Bethesda in 1992 (7).
Author Manuscript

The initial interest in tract tracing was to study the evolution of higher function in the
humans from the early hominoids as for example speech (10, 11). This was the original
impetus to generate images of tracts in the brain. Diffusion nuclear magnetic resonance has
been around for decades. It is yet another source of obtaining contrast in individual voxels
based on the rate of signal decay that related to the degree of water to diffuse anisotropically
(in a primary direction) as opposed to isotropically (in all directions) (12). This is the basis
for diffusion weighted MRI that has been utilized to demonstrate ischemic stroke for several
years now (13, 14). DTI is a fundamental modification in MRI data processing that allows
each voxel to produce not only signal intensity data but also directional data that show the
tensor (3D complex vector) direction in a 3 dimensional space. Each voxel represented by a
single arrow is then collated into an array of directional arrows in the orientation of neural
Author Manuscript

tracts. These directional arrows are then strung together by graphic techniques to produce
linear images of nerve tracts (9).

Traditionally the fiber streamlines were reconstructed utilizing deterministic (15) or


probabilistic methods (16–18). These methods of white fiber analysis have several
limitations. When fiber pathways are being mapped at locations of intersections the
traditional analysis methods are unable to display the full dispersion of the pathways. A
good example of this situation is the mapping of fibers that pass through complex white
matter like the centrum semiovale, where pyramidal tract, superior longitudinal fasciculus
and the corpus callosum are known to intersect. As has been demonstrated by Bucci et al
these issues can be overcome by High Angular Resolution Diffusion Imaging (HARDI)
technique that allows unraveling of the intersecting fibers to enable separate fiber tracking
Author Manuscript

(19). They documented feasibility of this technique in 12 patients with brain tumors by
showing excellent correlation of fiber tractography with intra-operative electrical stimulation
(12). In a further advancement in computational abilities Presseau et al demonstrate the
ability of a new compression format to process several thousands and often millions of fiber
streamlines to be stored on smaller memory space (20). This is a significant advancement
where this compression format has been validated in the several different DTI techniques
including the HARDI technique (Figure 1).

Exp Neurol. Author manuscript; available in PMC 2016 October 01.


Chaudhary et al. Page 4

Clinical application of DTI


Author Manuscript

Since its inception in the 1990s there have been over 2000 studies published exploring the
role of DTI tractography in the brain, although most of them have been in the last 5 years
(7). The role of DTI has been explored in formal outcome studies of patients with traumatic
brain injury, ICH (Figure 2), and surgical guidance in optimizing glioma resection (21–23).
The other clinical scenarios where DTI has been explored are in evaluation of dementia in
Alzheimer’s disease and in identifying subtle lesions that could explain the etiology of
epilepsy (24, 25).

Application of DTI in Traumatic Brain Injury (TBI) has recently gathered strength. A pilot
project has been started called TRACK TBI (transforming research and clinical knowledge
in TBI) to improve understanding of mechanisms of brain injury and factors associated with
improved functional outcomes (26). There are several studies that report on the changes in
Author Manuscript

white matter tract DTI parameters in acute, subacute and chronic time points following mild
TBI (27–35). More recently the TRACK TBI study group looked at the ability of DTI,
amongst other MRI parameters, to predict functional outcomes at 3 and 6 months following
mild TBI (36). They evaluated that in 76 patients with mild TBI the independent predictors
of neurological function outcome were parameters identified on DTI. They demonstrate in
their study that white matter fractional anisotropy (FA) was significantly reduced in CT/
MRI-positive, but not in CT/MRI-negative, mild TBI patients when compared to healthy
control subjects, on a group level. In addition reduced FA values in individual mild TBI
patients although modest, were statistically significant predictors of unfavorable 3- and 6-
month outcome. Moreover, these results remained statistically significant not only in the
inclusive sample of 76 mild TBI patients but also in the subset of 37 mild TBI patients
without prior history of substance abuse or other neuropsychiatric disorder (29).
Author Manuscript

Mechanisms of brain injury in hemorrhagic stroke


The mechanisms of white matter tract injuries in hemorrhagic stroke are better understood
now. One of the commonest mechanisms of neuronal injury following an ICH is due to
Wallerian degeneration in the corticospinal tract in the immediate vicinity and remote from
the site of the hematoma. This injury is from direct injury to the neuronal structures in the
deep white or grey matter. Apart from the mechanical mass effect, inflammation and edema
around the periphery of the hematoma that takes place in the acute period(37, 38) there are
other mechanisms mediating injury to the neural structures of the white matter (39, 40). In
the early phase there is neuronal damage mediated by blood components and the
hemoxygenase enzymes that cleave heme. Heme degradation products then diffuse out into
the tissues causing cellular toxicity (40, 41). This mechanism of cellular injury is also
Author Manuscript

applicable in patients with intraventricular hemorrhage, however, the cellular mechanisms of


injury in patients with subarachnoid hemorrhage is not well understood.

In the background of experiments on ICH animal models there is gathering body of evidence
for early and delayed mechanisms of cellular injury. Currently our ability to be able to
predict neuronal functional outcome in the event of a hemorrhagic stroke is suboptimal. DTI
is one of the non-invasive markers which can predict functional outcome. DTI is now

Exp Neurol. Author manuscript; available in PMC 2016 October 01.


Chaudhary et al. Page 5

reaching a point when the resolution and ability to track fibers optimally is very reliable.
Author Manuscript

Secondary injuries to the brain tissue at the periphery of the hematoma after primary
cerebral hemorrhage are mediated by both the mass effect of the new hemorrhage and the
toxicity that is associated with hematoma degradation and organization and the consequent
release of inflammatory and free radical mediators. Iron has been shown to be responsible
for cerebral toxicity in all types of cerebral hemorrhage including SAH (41). For example,
deferoxamine, an iron chelator, reduces ICH-induced brain damage (39) (Figure 3). Egashira
et al in a recent publication showed that Lipocalin 2 (LCN 2) may play an important role in
the mediation of white matter tract injury following SAH in a vessel perforation wild type
mouse model (42). Lipocalin 2 is an iron transport protein that has been implicated in
cerebral toxicity. The authors demonstrate that the T2 signal on MRI was significantly high
following SAH due to LCN 2 expression as evidenced by significant lack of T2 signal
abnormality on MRI in LCN2 knockout mice (Figure 4). Given this background of
Author Manuscript

developing understanding of white matter tract injury mechanisms in hemorrhagic stroke,


we will now look at the existing evidence for ability of DTI to evaluate injured white matter
tracts due to hemorrhagic stroke. For this purpose a comprehensive search of the literature
was performed on pubmed with search criteria including hemorrhagic stroke, DTI,
tractography, SAH, IVH, and ICH.

Existing evidence for DTI application in hemorrhagic stroke


DTI application in early detection of white fiber injury in animal models
ICH is the most common hemorrhagic stroke subtype that the role of DTI has been explored
the most in. The ipsilateral corticospinal tract (CST) including the pyramidal tract (PY) has
been increasingly studied in cases of stroke induced damage. The CST is an important white
matter tract made up of PY, internal capsule and the cerebral peduncle. It has been shown
Author Manuscript

that the asymmetry in fractional anisotropy (FA) values generated from DTI maps in the
CST correlates with clinical signs and can predict functional outcome (43–48). Furthermore
there has been a recent study that showed a decrease in FA values in the contralateral CST at
7 to 28 days following ICH, suggesting that FA asymmetry may not be a robust assessment
of secondary motor pathway injury (49). Shu-Jan Fan et al suggest that our poor
understanding of the contralateral PY and substantia nigra (SN) changes following ICH as
demonstrated in several other ICH animal model studies can be secondary to use of different
ICH sites and different imaging time points in those studies (23, 43, 46, 49). In their study
they employ a robust rodent ICH model utilizing intra-striatal collagenase infusion to create
the hematoma (50). They then longitudinally quantitatively assessed the changes on DTI in
both contralateral and ipsilateral SN and PY tracts following ICH which were compared
with histological examination. In their study they performed induction of an ICH in the
Author Manuscript

striatum of 14 Sprague Dawley rats. MRI at 7 T was performed at days 1, 3, 7, 14, 42, and
120 days post ICH. The parameters assessed were FA, Mean Diffusivity (MD), Axial and
radial diffusivity. The authors concluded that DTI parameters revealed early changes in both
the ipsilateral and contralateral SN and PY following ICH. There was good correlation of
the evolution of DTI findings with histological examination. The early reduction in radial
diffusivity was surmised to be due to early axonal excitation in bilateral SN secondary to the
ipsilateral excitatory injury. The authors in this study also observed initial decrease in mean

Exp Neurol. Author manuscript; available in PMC 2016 October 01.


Chaudhary et al. Page 6

diffusivity (MD) simultaneously in the ipsilateral and contralateral PY. This phenomenon
Author Manuscript

has been observed in the contralateral CST in ischemic stroke, which has been correlated
with poor outcome (45, 47). In other experiments decreased diffusion has been demonstrated
in cerebral locations remote from the primary site of ICH (51, 52). In yet other experiments
vasospasm that is usually the cause of delayed cerebra ischemia has been implicated in
remote ischemia in ICH involving the ventricles (53, 54). Shu-Jan Fan et al in their study
show that in those ICH rodent models which were associated with ventricular involvement
showed early decreased diffusivity on DTI in bilateral PY. They explain that the
combination of ventricular involvement, mass effect from the hematoma, and vasogenic
edema result in raised intracranial pressure causing relative transient ischemia in distant
brain parenchyma causing cellular dysfunction mediated by cell membrane proton pump
failure, intracellular edema, myelin swelling and cessation of energy dependent axoplasmic
processes. All these cellular correlates can explain the decreased MD in bilateral PY tracts
Author Manuscript

(50).

DTI application in human subjects with ICH and functional outcome at 1 month
Evidence in the literature suggests the role of DTI in human subjects (46). The goal is to be
able to predict the functional outcome in the event of an ICH to enable targeted therapy or to
develop a rehabilitation strategy early. A preserved or recovered pyramidal tract is crucial
for a good neurological functional outcome following an ICH (55). Several attempts have
been made to predict the neurological motor function outcome utilizing various MRI
techniques. It has been shown in ischemic stroke, that the functional outcome bears on the
extent of wallerian degeneration remote from the lesion as picked up on MRI (56, 57).
However, the signal intensity changes appear on MRI approximately 4 weeks from the ictus
(58). There has been some evidence that DTI can show changes in signal intensity in the
Author Manuscript

ipsilateral pyramidal tract earlier than the standard MRI sequences. The FA decreases
progressively in the ipsilateral pyramidal tract in the subacute and the chronic stages of
ischemic stroke, which correlates with motor functional outcome (59–61).

In the clinical scenario of a hemorrhagic stroke a few studies have demonstrated a


correlation between decreasing FA on DTI with functional outcome (43). In these studies
MRI was performed 3 days following the hematoma or removal of it. There are not many
studies that have looked at diffusivity changes in the very early acute phase of the ICH and
whether these changes correlate with functional outcome (62). Kusano et al in 18 patients
with ICH explored the role of DTI and diffusivity changes in 2 days following the onset of
the hematoma and tried to see if the changes correlated with functional outcome (46). They
demonstrate that the measured FA in the ipsilateral cerebral peduncle decreased by 11%
compared to the contralateral side. The ratio of FA (rFA) between the ipsilateral and
Author Manuscript

contralateral side cerebral peduncle was negatively correlated with the paresis grading at day
0 and day 28 and modified Rankin score (mRS) at 28 days. The rFA showed a significant
difference good and poor outcome groups. In addition the authors show that rFA correlated
significantly with motor outcome in comparison to other variables used by them in the ICH
scales. The mean diffusivity (MD) was unchanged in the affected cerebral peduncle. This is
in keeping with prior studies showing unchanged MD on the affected side PY tract in
ischemic stroke patients (44, 60, 63). The authors explain this phenomenon by the fact that

Exp Neurol. Author manuscript; available in PMC 2016 October 01.


Chaudhary et al. Page 7

following an ICH there is loss of axonal structures, membrane disintegration and cellular
Author Manuscript

debris leading to decrease in the first eigenvalue and a relative increase the second and the
third diffusion eigenvalues (46). Thus while there is decreased anisotropy the effect on
orientationally averaged diffusion index is more challenging to predict and hence MD may
remain unchanged in the very early phase i.e. first 2 days of and ICH. Yokoyama et al in
another DTI in ICH study in human population have reported that the mean FA ratio
decreased by 11% on the ipsilateral pyramidal tract in comparison to the contralateral side in
the good recovery group of patients while the reduction in mean FA ratio was 23% in the
group of patients with poor recovery (43). In another study Kuzu et al showed a difference
of 9% in the absolute FA values between the patients with good or poor recovery (61).

DTI application in humans with ICH and correlation with outcome at 6 months
So we see that there is gathering body of evidence in ICH from animal studies to those in
Author Manuscript

humans that reductions in FA in the acute phase correlate with motor function at 1 month.
DaMing Wang et al looked at correlation of DTI with motor function outcome 6 months
following an ICH (64). They evaluated DTI performed at 3 days and 2 weeks following ICH
in 36 patients. The parameters selected for evaluation were FA, rFA and MD measured at
the anterior cerebral peduncle. The results demonstrate that the rFA measured at 2 weeks in
the cerebral peduncle correlated significantly with the mRS at 6 months. The authors further
opine that their results compare well with those of Kusano et al, Puig et al and Feys et al in
so much as non-correlation of the size of the hematoma with motor function outcome (46,
65, 66). As shown in the other studies only the CST DTI showed correlation with motor
function outcome. The authors also suggest that DTI measurements at the cerebral peduncle
are less prone to miscalculations given the perpendicular arrangement of the fiber tracts with
respect to the axial plane of imaging. DaMing Wang et al also recommend further studies to
Author Manuscript

evaluate DTI and diffusion tensor tractography (DTT) correlation with motor function
outcome as has been preliminarily demonstrated by Cho et al in a separate study (67). There
are other studies that have refined the correlation of DTI parameters with long term motor
function (68). Tetsuo Koyama et al in their study demonstrate good correlation of DTI
parameters with upper extremity recovery of motor function 6–7 months following and ICH
(69).

DTI application in humans with ICH correlating with functional outcome at 1 month and 3
months
As we have seen from the previous studies discussed above they explore the role of DTI of
the affected CST in the acute phase of ICH and its correlation with motor function outcome.
The aspect of variations in FA and correlation with motor function outcome has not been
Author Manuscript

well addressed in the existing literature. In another study Chi Cheng Ma et al explored the
dynamic variations of FA measurements on DTI of the affected CST in patients with ICH
over a period of 90 days (70). They performed DTI in 25 ICH patients on day 0, 30 & 90. In
this study all patients underwent a motor function score (MFS) assessment before their MRI
at day 0, 30 and 90 by experienced neurosurgeons blinded to the DTI results. The MRI
sequence utilized was diffusion weighted echo planar imaging with gradients in 6 directions
on all the study patients. The authors observed that the day 0 FA correlated significantly
with MFS at day 90. In addition patients with an initial FA value above 0.45 could be

Exp Neurol. Author manuscript; available in PMC 2016 October 01.


Chaudhary et al. Page 8

anticipated to have a good motor function outcome following ICH with conservative
Author Manuscript

management. The authors also demonstrate that progressive reductions of FA values were
noted in the ipsilateral CST correlating with poor motor function. On the other hand the FA
measurements increased in the good motor function outcome group. In the opinion of the
authors of this study the exact modus operandi of the trend of FA values in the opposite
direction in its correlation with good outcome is not entirely explicit. They further surmise
that wallerian degeneration is induced by direct damage to white matter fibers from the
hematoma followed by cytotoxic and vasogenic edema. The cytotoxic edema along the
tracts is not readily apparent on conventional MRI which can be detected by DTI. In their
study by logistic analysis they showed that the day 0 high FA value was the only one
correlating with good motor function outcome. The MFS score at the outset of the ICH did
not correlate with day 90 MFS scores to predict a good motor function outcome. This means
that some patients with poor motor function in the acute phase would improve in the chronic
Author Manuscript

phase. In addition patients with high MFS of 5 or higher correlates with those patients likely
to require surgical intervention to improve functional outcome (71). Thus Ching Ma et al
surmise in their article that FA measurements may help to select patients who may benefit
from surgical hematoma evacuation in turn enabling physicians with ability to predict motor
recovery in advance and also choose effective treatment strategy.

DTI application in humans with ICH to select ones who will benefit from intervention
In another study Guofeng Wan et al explored the role of DTI in evaluation of functional
outcome in patients who had treatment of the ICH with a minimally invasive procedure (72).
They randomized patients with ICH in the thalamic region into having the minimally
invasive procedure or not. DTI was performed on all these patients once at admission and
then another at 2 weeks following surgical intervention and also at the same time point in
Author Manuscript

the corresponding medically treated group. Along with the aforementioned imaging motor
evoked potentials were also recorded at identical time points. Measurements on DTI were
performed on the ipsilateral CST in the internal capsule and contralateral measurements in
identical anatomical location. The authors in this study found that DTI imaging demonstrate
that the white matter fibers of the CST in the ipsilateral side drop to less than 1/3rd of the
fiber density of the contralateral controls. In the group where hematoma was evacuated by
minimally invasive technique showed more than 2/3rd of the fibers recovered back to normal
while the fiber density measured by tractography was the same in the medically treated
group. Both the motor function and the NIHSS scale was significantly more improved in the
minimally invasive treated group. This study further proves that DTI technology is robust
and fiber tracking can quantify white matter recovery following a spontaneous ICH in the
basal ganglia region. There are at least two other studies that have looked at the role of DTI
Author Manuscript

in detecting recovery of the white matter fibers following ICH and treatment with hematoma
evacuation (73, 74). Heish et al in a case report demonstrate recovery of white matter fibers
in CST damaged by putaminal hematoma correlating with improved motor function post
evacuation of hematoma by minimally invasive techniques (75). Similarly Wu et al (ref 2 in
Guofeng article) demonstrate similar significant correlation in 27 patients treated with
minimally invasive methods to evacuate the hematoma, between improved FA and fiber
density in CST and motor function outcome (74).

Exp Neurol. Author manuscript; available in PMC 2016 October 01.


Chaudhary et al. Page 9

DTI application in animal IVH models


Author Manuscript

There are some animal studies that have demonstrated white matter fiber injury in the peri-
ventricular location on histopathological examination (76–78). Since its inception only one
study has been published showing the value of DTI in IVH (76) in an animal model. In this
study Chua et al in a rabbit pup brain demonstrate that there was good correlation with FA
and ADC value measurements in the corpus callosum (CC), corona radiate (CR) but not so
with the internal capsule (IC). There was significant decrease in FA values at CC and CR
correlating with evidence of white matter fiber damage on histology, while the
measurements at IC did not correlate. The pathological entities revealed in the rabbit pup
brain in the corresponding locations in CC, CR were reduced myelination, gliosis and axonal
injury. In the human population relationship of IVH, PVL and hydrocephalus has been
shown in the immature brain (79–81). Sang Seok Yeo et al were the first ones to explore the
role of DTI in 10 human subjects with a combination of IVH and SAH with IVH (82). They
Author Manuscript

showed that FA value reduction was significant in the fornix, CC, and CR. The ADC values
did not show any significant relationship. The authors of this article surmise that this
neuronal injury in the periventricular region in IVH could be the reason for poor outcome
and high mortality in stroke patients with IVH (83, 84). The authors further suggest that
accurate diagnosis of extent of neuronal injury would be crucial in targeting management
strategies to improve patient functional outcome.

DTI application in humans with SAH


Amongst the hemorrhagic stroke subtypes the white matter injury is most ill understood in
SAH. There are multiple different patho-physiological mechanisms that are at play that
inflict neuronal injury in the event of SAH. These include vasospasm, direct injury from
hematoma, insufficient blood supply to the paracentral sulci, or injury to the CST from the
Author Manuscript

aneurysm (85, 86). The evidence for reliability of DTI in denoting early injury to the CST in
other forms of stroke settings makes it the natural MRI biomarker to study in the clinical
scenario of SAH. Sang Seok Yeo et al have explored this aspect in their study of 22 patients
with SAH. DTI parameters were measured in the CR, posterior limb of the internal capsule,
mid brain, mid pons and medulla in 22 patients and 24 age and sex matched controls at 4
weeks from the ictus (87). They demonstrated significantly low values of FA in ROI within
CST at the midbrain in SAH patients compared to the controls. No significant difference
was demonstrated in the measurements at other anatomical locations. Moreover, no
correlation was observed in the ADC values or between the FA values and the motor
function scores. These observations support the hypothesis that CST at the midbrain can be
injured during SAH due to early hydrocephalus (76). In addition another hypothesis that the
close proximity of blood in the perimesencephalic cistern to the CST in the midbrain makes
Author Manuscript

it a target of damage by SAH is supported by the observations in the aforementioned study


(88). It is interesting that motor function outcome did not correlate with the FA values of
CST in the midbrain. This only suggests that white matter tract injury is not the only
mechanism of injury governing the extent of motor function compromise.

Exp Neurol. Author manuscript; available in PMC 2016 October 01.


Chaudhary et al. Page 10

Future directions
Author Manuscript

Although there is reasonable evidence that has accumulated in the last decade that
demonstrates DTI to be sensitive enough to detect white matter tract injury in hemorrhagic
stroke a definite correlation with motor function outcome is still preliminary. In parallel our
understanding of the pathomechanisms of neuronal damage in hemorrhagic stroke is also
increasing. A very good example is the depiction of iron mediated neuronal injury that has
come across through several studies on this topic especially in the non-human primate and
non-primate animal models (39, 89). The authors of this review firmly believe large human
population studies are needed to explore correlation of parenchymal tissue iron levels and
damage to white matter tracts detected by DTI and further connection of these biomarkers
with motor function outcome. Large prospective studies are needed in the human population
to explore this reliably. The next stage will be the evaluation of identified therapeutic targets
based on the aforementioned studies to in randomized controlled human population studies
Author Manuscript

to get to meaningful evidence to support therapies to improve functional outcome in patients


with hemorrhagic stroke. Furthermore, in future studies, it is important to determine whether
DTI can detect white matter edema development and resolution, and white matter recovery.

Conclusion
Hemorrhagic stroke is debilitating and can result in mortality and significant morbidity.
Currently no effective therapeutic measure exists that can improve functional outcomes in
this clinical scenario. Physiological support is the only management approach currently
available with which physicians hope to ride off the insult of hemorrhagic stroke. As we
have discussed in the manuscript conventional non-invasive imaging with conventional MRI
is not sensitive enough to obtain information about extent of damage to guide management
Author Manuscript

strategies more effectively. DTI technique, as we have discussed, shows preliminary


evidence that it may hold promise in predicting patients with poor prognosis early on in
hemorrhagic stroke. It may also reveal potential therapeutic targets to ensure good motor
function outcomes in these patients. To begin with there is a huge potential for
demonstrating DTI as a reliable imaging surrogate of impending neuronal toxicity in
patients with hemorrhagic stroke. The next step would be to demonstrate reliable correlation
of DTI with functional outcome to be able to stratify risk in patients with hemorrhagic
stroke.

Acknowledgments
Supported by grants NS-073595, NS-079157, NS-084049, NS-091545, 973 Program-2014CB541600 and an
UMHS-PUHSC Joint Institute grant.
Author Manuscript

References
1. Pendlebury ST. Worldwide under-funding of stroke research. International journal of stroke :
official journal of the International Stroke Society. 2007 May; 2(2):80–4. [PubMed: 18705958]
2. Leak RK, Zheng P, Ji X, Zhang JH, Chen J. From apoplexy to stroke: historical perspectives and
new research frontiers. Prog Neurobiol. 2014 Apr.115:1–5. [PubMed: 24373958]
3. Xi G, Strahle J, Hua Y, Keep RF. Progress in translational research on intracerebral hemorrhage: Is
there an end in sight? Prog Neurobiol. 2014 Apr.115C:45–63. [PubMed: 24139872]

Exp Neurol. Author manuscript; available in PMC 2016 October 01.


Chaudhary et al. Page 11

4. Chen S, Feng H, Sherchan P, Klebe D, Zhao G, Sun X, et al. Controversies and evolving new
mechanisms in subarachnoid hemorrhage. Prog Neurobiol. 2014 Apr.115:64–91. [PubMed:
Author Manuscript

24076160]
5. Pandey AS, Xi G. Intracerebral hemorrhage: a multimodality approach to improving outcome.
Transl Stroke Res. 2014 Jun; 5(3):313–5. [PubMed: 24764218]
6. Zhang JH. Vascular neural network in subarachnoid hemorrhage. Transl Stroke Res. 2014 Aug;
5(4):423–8. [PubMed: 24986148]
7. Tso MK, Macdonald RL. Subarachnoid hemorrhage: a review of experimental studies on the
microcirculation and the neurovascular unit. Transl Stroke Res. 2014 Apr; 5(2):174–89. [PubMed:
24510780]
8. Wagner KR. Modeling intracerebral hemorrhage: glutamate, nuclear factor-kappa B signaling and
cytokines. Stroke. 2007; 38:753–8. [PubMed: 17261732]
9. Filler A. Magnetic resonance neurography and diffusion tensor imaging: origins, history, and
clinical impact of the first 50,000 cases with an assessment of efficacy and utility in a prospective
5000-patient study group. Neurosurgery. 2009 Oct; 65(4 Suppl):A29–43. [PubMed: 19927075]
10. Filler AG. Axonal transport and MR imaging: prospects for contrast agent development. Journal of
Author Manuscript

magnetic resonance imaging : JMRI. 1994 May-Jun;4(3):259–67. [PubMed: 7520308]


11. Filler AG, Whiteside GT, Bacon M, Frederickson M, Howe FA, Rabinowitz MD, et al. Tri-partite
complex for axonal transport drug delivery achieves pharmacological effect. BMC neuroscience.
2010; 11:8. [PubMed: 20085661]
12. Le Bihan D. Molecular diffusion nuclear magnetic resonance imaging. Magnetic resonance
quarterly. 1991 Jan; 7(1):1–30. [PubMed: 2043461]
13. Kakuda W, Lansberg MG, Thijs VN, Kemp SM, Bammer R, Wechsler LR, et al. Optimal
definition for PWI/DWI mismatch in acute ischemic stroke patients. Journal of cerebral blood flow
and metabolism : official journal of the International Society of Cerebral Blood Flow and
Metabolism. 2008 May; 28(5):887–91.
14. Lansberg MG, Thijs VN, O’Brien MW, Ali JO, de Crespigny AJ, Tong DC, et al. Evolution of
apparent diffusion coefficient, diffusion-weighted, and T2-weighted signal intensity of acute
stroke. AJNR American journal of neuroradiology. 2001 Apr; 22(4):637–44. [PubMed: 11290470]
15. Mori S, Crain BJ, Chacko VP, van Zijl PC. Three-dimensional tracking of axonal projections in the
Author Manuscript

brain by magnetic resonance imaging. Annals of neurology. 1999 Feb; 45(2):265–9. [PubMed:
9989633]
16. Jones DK, Pierpaoli C. Confidence mapping in diffusion tensor magnetic resonance imaging
tractography using a bootstrap approach. Magnetic resonance in medicine : official journal of the
Society of Magnetic Resonance in Medicine/Society of Magnetic Resonance in Medicine. 2005
May; 53(5):1143–9.
17. Lazar M, Alexander AL. An error analysis of white matter tractography methods: synthetic
diffusion tensor field simulations. NeuroImage. 2003 Oct; 20(2):1140–53. [PubMed: 14568483]
18. Parker GJ, Haroon HA, Wheeler-Kingshott CA. A framework for a streamline-based probabilistic
index of connectivity (PICo) using a structural interpretation of MRI diffusion measurements.
Journal of magnetic resonance imaging : JMRI. 2003 Aug; 18(2):242–54. [PubMed: 12884338]
19. Bucci M, Mandelli ML, Berman JI, Amirbekian B, Nguyen C, Berger MS, et al. Quantifying
diffusion MRI tractography of the corticospinal tract in brain tumors with deterministic and
probabilistic methods. NeuroImage Clinical. 2013; 3:361–8. [PubMed: 24273719]
20. Presseau C, Jodoin PM, Houde JC, Descoteaux M. A new compression format for fiber tracking
Author Manuscript

datasets. NeuroImage. 2015 Apr 1.109:73–83. [PubMed: 25592997]


21. Sidaros A, Engberg AW, Sidaros K, Liptrot MG, Herning M, Petersen P, et al. Diffusion tensor
imaging during recovery from severe traumatic brain injury and relation to clinical outcome: a
longitudinal study. Brain : a journal of neurology. 2008 Feb; 131(Pt 2):559–72. [PubMed:
18083753]
22. Wu JS, Zhou LF, Tang WJ, Mao Y, Hu J, Song YY, et al. Clinical evaluation and follow-up
outcome of diffusion tensor imaging-based functional neuronavigation: a prospective, controlled
study in patients with gliomas involving pyramidal tracts. Neurosurgery. 2007 Nov; 61(5):935–48.
discussion 48–9. [PubMed: 18091270]

Exp Neurol. Author manuscript; available in PMC 2016 October 01.


Chaudhary et al. Page 12

23. Yoshioka H, Horikoshi T, Aoki S, Hori M, Ishigame K, Uchida M, et al. Diffusion tensor
tractography predicts motor functional outcome in patients with spontaneous intracerebral
Author Manuscript

hemorrhage. Neurosurgery. 2008 Jan; 62(1):97–103. discussion. [PubMed: 18300896]


24. Chua TC, Wen W, Slavin MJ, Sachdev PS. Diffusion tensor imaging in mild cognitive impairment
and Alzheimer’s disease: a review. Current opinion in neurology. 2008 Feb; 21(1):83–92.
[PubMed: 18180656]
25. Diehl B, Busch RM, Duncan JS, Piao Z, Tkach J, Luders HO. Abnormalities in diffusion tensor
imaging of the uncinate fasciculus relate to reduced memory in temporal lobe epilepsy. Epilepsia.
2008 Aug; 49(8):1409–18. [PubMed: 18397294]
26. Yue JK, Vassar MJ, Lingsma HF, Cooper SR, Okonkwo DO, Valadka AB, et al. Transforming
research and clinical knowledge in traumatic brain injury pilot: multicenter implementation of the
common data elements for traumatic brain injury. Journal of neurotrauma. 2013 Nov 15; 30(22):
1831–44. [PubMed: 23815563]
27. Arfanakis K, Haughton VM, Carew JD, Rogers BP, Dempsey RJ, Meyerand ME. Diffusion tensor
MR imaging in diffuse axonal injury. AJNR American journal of neuroradiology. 2002 May;
23(5):794–802. [PubMed: 12006280]
Author Manuscript

28. Bazarian JJ, Zhong J, Blyth B, Zhu T, Kavcic V, Peterson D. Diffusion tensor imaging detects
clinically important axonal damage after mild traumatic brain injury: a pilot study. Journal of
neurotrauma. 2007 Sep; 24(9):1447–59. [PubMed: 17892407]
29. Chu Z, Wilde EA, Hunter JV, McCauley SR, Bigler ED, Troyanskaya M, et al. Voxel-based
analysis of diffusion tensor imaging in mild traumatic brain injury in adolescents. AJNR American
journal of neuroradiology. 2010 Feb; 31(2):340–6. [PubMed: 19959772]
30. Kim N, Branch CA, Kim M, Lipton ML. Whole brain approaches for identification of
microstructural abnormalities in individual patients: comparison of techniques applied to mild
traumatic brain injury. PloS one. 2013; 8(3):e59382. [PubMed: 23555665]
31. Lipton ML, Kim N, Park YK, Hulkower MB, Gardin TM, Shifteh K, et al. Robust detection of
traumatic axonal injury in individual mild traumatic brain injury patients: intersubject variation,
change over time and bidirectional changes in anisotropy. Brain imaging and behavior. 2012 Jun;
6(2):329–42. [PubMed: 22684769]
32. Mayer AR, Ling J, Mannell MV, Gasparovic C, Phillips JP, Doezema D, et al. A prospective
diffusion tensor imaging study in mild traumatic brain injury. Neurology. 2010 Feb 23; 74(8):643–
Author Manuscript

50. [PubMed: 20089939]


33. Messe A, Caplain S, Paradot G, Garrigue D, Mineo JF, Soto Ares G, et al. Diffusion tensor
imaging and white matter lesions at the subacute stage in mild traumatic brain injury with
persistent neurobehavioral impairment. Human brain mapping. 2011 Jun; 32(6):999–1011.
[PubMed: 20669166]
34. Smits M, Houston GC, Dippel DW, Wielopolski PA, Vernooij MW, Koudstaal PJ, et al.
Microstructural brain injury in post-concussion syndrome after minor head injury. Neuroradiology.
2011 Aug; 53(8):553–63. [PubMed: 20924757]
35. Wilde EA, McCauley SR, Hunter JV, Bigler ED, Chu Z, Wang ZJ, et al. Diffusion tensor imaging
of acute mild traumatic brain injury in adolescents. Neurology. 2008 Mar 18; 70(12):948–55.
[PubMed: 18347317]
36. Yuh EL, Cooper SR, Mukherjee P, Yue JK, Lingsma HF, Gordon WA, et al. Diffusion tensor
imaging for outcome prediction in mild traumatic brain injury: a TRACK-TBI study. Journal of
neurotrauma. 2014 Sep 1; 31(17):1457–77. [PubMed: 24742275]
Author Manuscript

37. Zhao X, Sun G, Zhang H, Ting SM, Song S, Gonzales N, et al. Polymorphonuclear neutrophil in
brain parenchyma after experimental intracerebral hemorrhage. Transl Stroke Res. 2014 Oct; 5(5):
554–61. [PubMed: 24696130]
38. Hatakeyama T, Okauchi M, Hua Y, Keep RF, Xi G. Deferoxamine reduces neuronal death and
hematoma lysis after intracerebral hemorrhage in aged rats. Transl Stroke Res. 2013 Oct; 4(5):
546–53. [PubMed: 24187595]
39. Gu Y, Hua Y, Keep RF, Morgenstern LB, Xi G. Deferoxamine reduces intracerebral hematoma-
induced iron accumulation and neuronal death in piglets. Stroke. 2009 Jun; 40(6):2241–3. Epub
2009/04/18. eng. [PubMed: 19372448]

Exp Neurol. Author manuscript; available in PMC 2016 October 01.


Chaudhary et al. Page 13

40. Zhou Y, Wang Y, Wang J, Anne Stetler R, Yang QW. Inflammation in intracerebral hemorrhage:
from mechanisms to clinical translation. Prog Neurobiol. 2014 Apr.115:25–44. [PubMed:
Author Manuscript

24291544]
41. Xi G, Keep RF, Hoff JT. Mechanisms of brain injury after intracerebral haemorrhage. The Lancet
Neurology. 2006 Jan; 5(1):53–63. [PubMed: 16361023]
42. Egashira Y, Hua Y, Keep RF, Xi G. Acute white matter injury after experimental subarachnoid
hemorrhage: potential role of lipocalin 2. Stroke. 2014 Jul; 45(7):2141–3. [PubMed: 24893611]
43. Yokoyama K, Matsuki M, Shimano H, Sumioka S, Ikenaga T, Hanabusa K, et al. Diffusion tensor
imaging in chronic subdural hematoma: correlation between clinical signs and fractional
anisotropy in the pyramidal tract. AJNR American journal of neuroradiology. 2008 Jun; 29(6):
1159–63. [PubMed: 18356470]
44. Thomalla G, Glauche V, Koch MA, Beaulieu C, Weiller C, Rother J. Diffusion tensor imaging
detects early Wallerian degeneration of the pyramidal tract after ischemic stroke. NeuroImage.
2004 Aug; 22(4):1767–74. [PubMed: 15275932]
45. Domi T, deVeber G, Shroff M, Kouzmitcheva E, MacGregor DL, Kirton A. Corticospinal tract
pre-wallerian degeneration: a novel outcome predictor for pediatric stroke on acute MRI. Stroke; a
Author Manuscript

journal of cerebral circulation. 2009 Mar; 40(3):780–7.


46. Kusano Y, Seguchi T, Horiuchi T, Kakizawa Y, Kobayashi T, Tanaka Y, et al. Prediction of
functional outcome in acute cerebral hemorrhage using diffusion tensor imaging at 3T: a
prospective study. AJNR American journal of neuroradiology. 2009 Sep; 30(8):1561–5. [PubMed:
19556354]
47. DeVetten G, Coutts SB, Hill MD, Goyal M, Eesa M, O’Brien B, et al. Acute corticospinal tract
Wallerian degeneration is associated with stroke outcome. Stroke; a journal of cerebral circulation.
2010 Apr; 41(4):751–6.
48. Liu X, Tian W, Li L, Kolar B, Qiu X, Chen F, et al. Hyperintensity on diffusion weighted image
along ipsilateral cortical spinal tract after cerebral ischemic stroke: a diffusion tensor analysis.
European journal of radiology. 2012 Feb; 81(2):292–7. [PubMed: 21247713]
49. Kwak SY, Yeo SS, Choi BY, Chang CH, Jang SH. Corticospinal tract change in the unaffected
hemisphere at the early stage of intracerebral hemorrhage: a diffusion tensor tractography study.
European neurology. 2010; 63(3):149–53. [PubMed: 20134168]
50. Fan SJ, Lee FY, Cheung MM, Ding AY, Yang J, Ma SJ, et al. Bilateral substantia nigra and
Author Manuscript

pyramidal tract changes following experimental intracerebral hemorrhage: an MR diffusion tensor


imaging study. NMR in biomedicine. 2013 Sep; 26(9):1089–95. [PubMed: 23417762]
51. Qureshi AI. Significance of lesions with decreased diffusion on MRI in patients with intracerebral
hemorrhage. Stroke; a journal of cerebral circulation. 2012 Jan; 43(1):6–7.
52. Garg RK, Liebling SM, Maas MB, Nemeth AJ, Russell EJ, Naidech AM. Blood pressure
reduction, decreased diffusion on MRI, and outcomes after intracerebral hemorrhage. Stroke; a
journal of cerebral circulation. 2012 Jan; 43(1):67–71.
53. Condette-Auliac S, Bracard S, Anxionnat R, Schmitt E, Lacour JC, Braun M, et al. Vasospasm
after subarachnoid hemorrhage: interest in diffusion-weighted MR imaging. Stroke; a journal of
cerebral circulation. 2001 Aug; 32(8):1818–24.
54. Kiphuth IC, Huttner HB, Breuer L, Engelhorn T, Schwab S, Kohrmann M. Vasospasm in
intracerebral hemorrhage with ventricular involvement: a prospective pilot transcranial Doppler
sonography study. Cerebrovascular diseases. 2011; 32(5):420–5. [PubMed: 21986490]
55. Sawlani V, Gupta RK, Singh MK, Kohli A. MRI demonstration of Wallerian degeneration in
Author Manuscript

various intracranial lesions and its clinical implications. Journal of the neurological sciences. 1997
Mar 10; 146(2):103–8. [PubMed: 9077505]
56. Binkofski F, Seitz RJ, Arnold S, Classen J, Benecke R, Freund HJ. Thalamic metbolism and
corticospinal tract integrity determine motor recovery in stroke. Annals of neurology. 1996 Apr;
39(4):460–70. [PubMed: 8619524]
57. Mukherjee P. Diffusion tensor imaging and fiber tractography in acute stroke. Neuroimaging
clinics of North America. 2005 Aug; 15(3):655–65. xii. [PubMed: 16360595]

Exp Neurol. Author manuscript; available in PMC 2016 October 01.


Chaudhary et al. Page 14

58. Inoue Y, Matsumura Y, Fukuda T, Nemoto Y, Shirahata N, Suzuki T, et al. MR imaging of


Wallerian degeneration in the brainstem: temporal relationships. AJNR American journal of
Author Manuscript

neuroradiology. 1990 Sep-Oct;11(5):897–902. [PubMed: 2120993]


59. Buffon F, Molko N, Herve D, Porcher R, Denghien I, Pappata S, et al. Longitudinal diffusion
changes in cerebral hemispheres after MCA infarcts. Journal of cerebral blood flow and
metabolism : official journal of the International Society of Cerebral Blood Flow and Metabolism.
2005 May; 25(5):641–50.
60. Pierpaoli C, Barnett A, Pajevic S, Chen R, Penix LR, Virta A, et al. Water diffusion changes in
Wallerian degeneration and their dependence on white matter architecture. NeuroImage. 2001 Jun;
13(6 Pt 1):1174–85. [PubMed: 11352623]
61. Werring DJ, Toosy AT, Clark CA, Parker GJ, Barker GJ, Miller DH, et al. Diffusion tensor
imaging can detect and quantify corticospinal tract degeneration after stroke. Journal of neurology,
neurosurgery, and psychiatry. 2000 Aug; 69(2):269–72.
62. Qiu TM, Zhang Y, Wu JS. Preliminary application of pyramidal tractography in evaluating
prognosis of patients with hypertensive intracerebral hemorrhage. Acta neurochirurgica
Supplement. 2008; 105:165–70. [PubMed: 19066104]
Author Manuscript

63. Delsing BJ, Catsman-Berrevoets CE, Appel IM. Early prognostic indicators of outcome in
ischemic childhood stroke. Pediatric neurology. 2001 Apr; 24(4):283–9. [PubMed: 11377103]
64. Wang DM, Li J, Liu JR, Hu HY. Diffusion tensor imaging predicts long-term motor functional
outcome in patients with acute supratentorial intracranial hemorrhage. Cerebrovascular diseases.
2012; 34(3):199–205. [PubMed: 23006600]
65. Puig J, Pedraza S, Blasco G, Daunis IEJ, Prados F, Remollo S, et al. Acute damage to the posterior
limb of the internal capsule on diffusion tensor tractography as an early imaging predictor of
motor outcome after stroke. AJNR American journal of neuroradiology. 2011 May; 32(5):857–63.
[PubMed: 21474629]
66. Feys H, Hetebrij J, Wilms G, Dom R, De Weerdt W. Predicting arm recovery following stroke:
value of site of lesion. Acta neurologica Scandinavica. 2000 Dec; 102(6):371–7. [PubMed:
11125752]
67. Cho SH, Kim SH, Choi BY, Cho SH, Kang JH, Lee CH, et al. Motor outcome according to
diffusion tensor tractography findings in the early stage of intracerebral hemorrhage. Neuroscience
letters. 2007 Jun 27; 421(2):142–6. [PubMed: 17566651]
Author Manuscript

68. Koyama T, Tsuji M, Miyake H, Ohmura T, Domen K. Motor outcome for patients with acute
intracerebral hemorrhage predicted using diffusion tensor imaging: an application of ordinal
logistic modeling. Journal of stroke and cerebrovascular diseases : the official journal of National
Stroke Association. 2012 Nov; 21(8):704–11. [PubMed: 21511497]
69. Koyama T, Marumoto K, Miyake H, Ohmura T, Domen K. Relationship between diffusion-tensor
fractional anisotropy and long-term outcome in patients with hemiparesis after intracerebral
hemorrhage. NeuroRehabilitation. 2013; 32(1):87–94. [PubMed: 23422461]
70. Ma C, Liu A, Li Z, Zhou X, Zhou S. Longitudinal study of diffusion tensor imaging properties of
affected cortical spinal tracts in acute and chronic hemorrhagic stroke. Journal of clinical
neuroscience : official journal of the Neurosurgical Society of Australasia. 2014 Aug; 21(8):1388–
92. [PubMed: 24746110]
71. Kuzu Y, Inoue T, Kanbara Y, Nishimoto H, Fujiwara S, Ogasawara K, et al. Prediction of motor
function outcome after intracerebral hemorrhage using fractional anisotropy calculated from
diffusion tensor imaging. Cerebrovascular diseases. 2012; 33(6):566–73. [PubMed: 22688137]
Author Manuscript

72. Wu G, Wang L, Liu J, Mao Y, Qin G. Minimally invasive procedures reduced the damages to
motor function in patients with thalamic hematoma: observed by motor evoked potential and
diffusion tensor imaging. Journal of stroke and cerebrovascular diseases : the official journal of
National Stroke Association. 2013 Apr; 22(3):232–40. [PubMed: 21917481]
73. Hsieh CT, Wu CC, Chiang YH, Chang CF. Stereotactic aspiration of enlarged intracerebral
hematoma caused by intraprocedural perforation of aneurysm during coil embolization. Surgical
neurology. 2008 Jun; 69(6):633–5. discussion 5–6. [PubMed: 18486706]
74. Wu G, Wang L, Hong Z, Li C, Long X, Sheng F. Effects of minimally invasive procedures for
removal of intracranial hematoma on matrix metalloproteinase expression and blood-brain barrier

Exp Neurol. Author manuscript; available in PMC 2016 October 01.


Chaudhary et al. Page 15

permeability in perihematomal brain tissues. Neurological research. 2011 Apr; 33(3):300–6.


[PubMed: 20712922]
Author Manuscript

75. Hsieh CT, Chen CY, Chiang YH, Chang CH, Chang CF. Role of diffusion tensor imaging in a
patient with spontaneous intracerebral hematoma treated by stereotactic evacuation. Surgical
neurology. 2008 Jul; 70(1):75–8. [PubMed: 17707485]
76. Chua CO, Chahboune H, Braun A, Dummula K, Chua CE, Yu J, et al. Consequences of
intraventricular hemorrhage in a rabbit pup model. Stroke; a journal of cerebral circulation. 2009
Oct; 40(10):3369–77.
77. Wasserman JK, Schlichter LC. White matter injury in young and aged rats after intracerebral
hemorrhage. Experimental neurology. 2008 Dec; 214(2):266–75. [PubMed: 18848934]
78. Pang D, Sclabassi RJ, Horton JA. Lysis of intraventricular blood clot with urokinase in a canine
model: Part 2. In vivo safety study of intraventricular urokinase. Neurosurgery. 1986 Oct; 19(4):
547–52. [PubMed: 3491339]
79. de la Monte SM, Hsu FI, Hedley-Whyte ET, Kupsky W. Morphometric analysis of the human
infant brain: effects of intraventricular hemorrhage and periventricular leukomalacia. Journal of
child neurology. 1990 Apr; 5(2):101–10. [PubMed: 2345277]
Author Manuscript

80. Takashima S, Mito T, Houdou S, Ando Y. Relationship between periventricular hemorrhage,


leukomalacia and brainstem lesions in prematurely born infants. Brain & development. 1989;
11(2):121–4. [PubMed: 2712235]
81. Larroque B, Marret S, Ancel PY, Arnaud C, Marpeau L, Supernant K, et al. White matter damage
and intraventricular hemorrhage in very preterm infants: the EPIPAGE study. The Journal of
pediatrics. 2003 Oct; 143(4):477–83. [PubMed: 14571224]
82. Yeo SS, Choi BY, Chang CH, Jung YJ, Ahn SH, Son SM, et al. Periventricular white matter injury
by primary intraventricular hemorrhage: a diffusion tensor imaging study. European neurology.
2011; 66(4):235–41. [PubMed: 21952179]
83. Arboix A, Rodriguez-Aguilar R, Oliveres M, Comes E, Garcia-Eroles L, Massons J. Thalamic
haemorrhage vs internal capsule-basal ganglia haemorrhage: clinical profile and predictors of in-
hospital mortality. BMC neurology. 2007; 7:32. [PubMed: 17919332]
84. Arboix A, Comes E, Garcia-Eroles L, Massons J, Oliveres M, Balcells M, et al. Site of bleeding
and early outcome in primary intracerebral hemorrhage. Acta neurologica Scandinavica. 2002
Apr; 105(4):282–8. [PubMed: 11939940]
Author Manuscript

85. Ferrante L, Acqui M, Mastronardi L, Celli P, Lunardi P, Fortuna A. Posterior inferior cerebellar
artery (PICA) aneurysm presenting with SAH and contralateral crural monoparesis: a case report.
Surgical neurology. 1992 Jul; 38(1):43–5. [PubMed: 1615373]
86. Greene KA, Marciano FF, Dickman CA, Coons SW, Johnson PC, Bailes JE, et al. Anterior
communicating artery aneurysm paraparesis syndrome: clinical manifestations and pathologic
correlates. Neurology. 1995 Jan; 45(1):45–50. [PubMed: 7824133]
87. Yeo SS, Choi BY, Chang CH, Kim SH, Jung YJ, Jang SH. Evidence of corticospinal tract injury at
midbrain in patients with subarachnoid hemorrhage. Stroke; a journal of cerebral circulation. 2012
Aug; 43(8):2239–41.
88. van Gijn J, van Dongen KJ, Vermeulen M, Hijdra A. Perimesencephalic hemorrhage: a
nonaneurysmal and benign form of subarachnoid hemorrhage. Neurology. 1985 Apr; 35(4):493–7.
[PubMed: 3982634]
89. Fujiwara S, Uhrig L, Amadon A, Jarraya B, Le Bihan D. Quantification of iron in the non-human
primate brain with diffusion-weighted magnetic resonance imaging. NeuroImage. 2014 Nov 15;
Author Manuscript

102(Pt 2):789–97. [PubMed: 25192653]

Exp Neurol. Author manuscript; available in PMC 2016 October 01.


Chaudhary et al. Page 16
Author Manuscript
Author Manuscript

Figure 1.
32 direction bilateral normal corticospinal tracts processed image from 3T MRI on a human
subject performed at the authors’ institution. This demonstrates the current ability to perform
diffusion tensor tractography on human subjects.
Author Manuscript
Author Manuscript

Exp Neurol. Author manuscript; available in PMC 2016 October 01.


Chaudhary et al. Page 17
Author Manuscript

Figure 2.
T2 and DTI MRI changes in the brain 6 days after intracerebral hemorrhage.
Author Manuscript
Author Manuscript
Author Manuscript

Exp Neurol. Author manuscript; available in PMC 2016 October 01.


Chaudhary et al. Page 18
Author Manuscript
Author Manuscript
Author Manuscript

Figure 3.
Deferoxamine reduces reddish zone around hematoma at day 3 and day 7 in a pig ICH
model. Pigs received an injection of autologous blood into the right frontal lobe.
Deferoxamine (50 mg/kg, IM) or vehicle was administered 2 hours after ICH and then every
Author Manuscript

12 hours up to 7 days. Animals were killed 3 or 7 days later to examine brain damage after
ICH. Values are means±SD, n=4, # p<0.01 vs. vehicle. (Reproduced with permission from:
Gu Y et al. Stroke, 2009;40:2241–2243)

Exp Neurol. Author manuscript; available in PMC 2016 October 01.


Chaudhary et al. Page 19
Author Manuscript
Author Manuscript
Author Manuscript

Figure 4.
Representative coronal T2 images (A) and NG2, β-amyloid precursor protein (β-APP) and
degraded myelin basic protein (DMBP) immunohistochemistry in white matter of sham and
WT and LCN2−/− mice 24 hours after SAH (B). Quantification of each result
(C). **P<0.01, *P<0.05 vs. WT, and ##P<0.01, #P<0.05 vs. LCN2−/− animals. Values are
means±SD; n=4–6. Scale bar = 100μm. (Reproduced with permission from: Egashira Y et
al. Stroke 2014;45:2141–2143)
Author Manuscript

Exp Neurol. Author manuscript; available in PMC 2016 October 01.


Acknowledgements

The editors would like to extend their gratitude to the following authors for providing additional
glossary contributions to this chapter: (in alphabetical order) Mattan Caan, Thijs Dhollander, Eric
Peterson, Matthew Rowe, Jacques-Donald Tournier, Jelle Veraart, and Sjoerd B Vos.

© Springer Science+Business Media New York 2016 419


W. Van Hecke et al. (eds.), Diffusion Tensor Imaging, DOI 10.1007/978-1-4939-3118-7
Glossary

Louise Emsell and Wim Van Hecke

The language of DTI is complex and sometimes Atlas A reference image constructed by coreg-
ill-defined. The following glossary has been istering and averaging multiple scans in order
assembled by the editors based on a selection of to create a representation of the image charac-
key terms provided by the contributing authors to teristics of a given study population. Atlases
this book as a guide to understanding some key are used for image normalization. Standard
terms related to DTI. Please note that in the atlases can be used to label regions-of- interest
absence of standard terminology, some words or and to report research findings in a common
phrases may be defined differently in other reference space.
sources. Automated tractography Tractography
driven by atlases based on anatomical infor-
Angular resolution The power to unambigu- mation, and requiring no user annotation.
ously resolve closely aligned directions, much Axial diffusivity (or longitudinal diffusiv-
like spatial resolution relates to the ability to ity) Equals the largest eigenvalue, and thus
resolve closely spaced points. May also be represents the ADC value along the main
used to refer to the density of DW sampling direction of diffusion. This is hypothesized to
over the sphere in a HARDI acquisition. relate to the orientation of an axonal bundle in
Anisotropic diffusion Diffusion of atoms the tensor model.
or molecules, which is dependent on Axial Kurtosis (AK, abbreviated form of axial
orientation—i.e., the rate of diffusion is dif- apparent kurtosis coefficient) The apparent
ferent for different directions. kurtosis coefficient, measured along the prin-
Apparent diffusion coefficient The measured cipal direction of diffusion.
diffusion coefficient for water in a medium B0 (or B = 0) A term used to informally refer
in which diffusion is not free as it is impeded to a non diffusion weighted image, acquired
by boundaries such as cellular structures. The as part of a full diffusion weighted imaging
water behaves more like a viscous fluid with a dataset.
lower diffusion coefficient. b-value A measure of the strength of the
Apparent fiber density A framework for the applied diffusion-weighting, and a function of
analysis of diffusion MRI data, which relies the sequence timings and DW gradient ampli-
on estimates of the fiber ODF as obtained tude. This is a generalization of the q-value,
from spherical deconvolution methods and valid under the assumption of free diffusion.
interprets the amplitude of the fiber ODF as Cardiac-gating Gating, or triggering the scan-
approximately proportional to the density ner to acquire data only during specific phases
of the fibers aligned with the corresponding in the cardiac cycle in which pulsation (and
direction. thus pulsation artifacts) is at a minimum
Association fibers White matter connections Cartesian A common method of sampling
between gyri within one hemisphere. k-space using a rectilinear or raster trajectory.

© Springer Science+Business Media New York 2016 421


W. Van Hecke et al. (eds.), Diffusion Tensor Imaging, DOI 10.1007/978-1-4939-3118-7
422 Glossary

Chemical shift artifact Different chemical the diffusion process in each voxel of a DTI
species have different resonance frequencies. image. The diffusion tensor “evaluated” along
These differences lead to mismatches in the fre- any direction to obtain ADC values as repre-
quency (k-space) and spatial domain (image). sented by that tensor.
For example, differences between water and fat Directionally encoded color (DEC)
molecules in the same physical location, mani- maps Maps that use RGB (red, green, blue)
fest as image artifacts characterized by bright encoding to map directions. In DTI, red is
or dark rims shifted relative to this location. typically associated with left-right, green with
Combined Hindered and Restricted Model of back-front and blue with bottom-top.
Diffusion A model for white matter as con- Displacement probability distribution
sisting of hindered axonal compartments and function The mathematical function that
a hindered extra-axonal compartment. Can be expresses the probability that a molecule has
used to extract information from DWI data displaced from point a to point b within a
given multi-shell HARDI data. given period, i.e., the diffusion time.
Commissural fibers White matter connections Dual spin echo (DSE) See twice-refocused
between both hemispheres. spin echo
Constant solid angle Q-ball imaging A Echo planar imaging (EPI) Rapid method of
method to estimate the diffusion ODF based signal readout that acquires multiple lines of
on the theory of q-space, which provides a k-space after a single excitation without any
more meaningful diffusion ODF than the orig- refocusing RF pulses during the readout.
inal QBI method. Eddy current-induced distortions Image dis-
Crossing fibers The situation whereby mul- tortions due to the presence of eddy currents.
tiple white matter bundles cross, fan, bend, The image in usually warped in nonlinear
or kiss within a single voxel. Crossing fibers fashion leading to skewing and shearing dis-
present considerable challenges to DTI tortions that manifest as a bright rim around
because it can only model a single principle reconstructed images.
direction of diffusion. Eddy currents Electrical currents in the con-
Deterministic tractography Tractography ducting materials in the scanner, induced by
based on the estimated average principal dif- the changing magnetic field, and especially the
fusion orientation (or eigenvector), resulting large and fast-switching diffusion-weighting
in one streamline per seed point. gradients.
Diffusion orientation density function A Eigendecomposition The process of obtain-
function on the sphere providing an estimate ing eigenvectors and eigenvalues of a tensor.
of the proportion of spins (e.g., water mol- Computer science provides reliable algo-
ecules) diffusing along any given orientation. rithms for this task.
Diffusion orientation transform A method to Eigenvalues The ADC values of the ten-
estimate a diffusion ODF-like measure, based sor along the directions of the eigenvectors.
on the theory of q-space. The dODF differs in They provide the complete information on the
that it represents the proportion of spins (e.g., shape and size of a tensor, independently of its
water molecules) that have diffused by a given orientation.
absolute distance along any given orientation Eigenvectors A new set of customized axes for
(rather than by any distance as it is typically a tensor, aligned along its specific orientation.
defined). They provide the complete information on the
Diffusion spectrum imaging A method to orientation of a tensor, independently of its
estimate the per-voxel average spin propaga- size and shape.
tor, based on the theory of q-space. This is Fast Spin Echo (FSE) An image acquisition
typically used to estimate the diffusion ODF sequence consisting of a single 90° excitation
by radial projection of the spin propagator. RF pulse followed by a train of 180° refocus-
Diffusion tensor (model) A 3 × 3 symmetric ing RF pulses, often called a Carr-Purcell-
tensor (six unique parameters) representing Meiboom-Gill (CPMG) pulse train. A less
Glossary 423

widely used alternative to EPI, for DTI. FSE number of diffusion-sensitizing gradients are
is slower but more robust to off-resonance applied along a set of uniformly distributed
distortions. directions.
Fat suppression Nulling the signal from Hardware phantoms Physical phantoms that
hydrogen atoms in fat to prevent chemical can be used to test the performance of diffu-
shift artifacts. sion MRI acquisition as well as processing.
Fiber orientation density function A func- Hindered diffusion Diffusion that is impeded
tion on the sphere providing an estimate of by non-enclosing boundaries or obstacles,
the amount of fibers aligned along any given such that the rate of diffusion is reduced, but
orientation. there is no limit on the maximum possible dis-
Field map A map quantifying the deviation of placement of an atom or molecule.
the magnetic field from the desired B0-field Histogram analysis (DTI) A histogram is a
(off-resonance). Can be used for unwarping. frequency distribution that displays the num-
Fractional anisotropy (FA) The standard ber of voxels with a specific value of the dif-
deviation of the eigenvalues, divided by their fusion measure (e.g., FA) obtained within an
root mean square. A measure of how much the ROI (e.g., brain mask). From this histogram,
eigenvalues differ, but carefully normalized the mean, median, peak height, and peak loca-
so it becomes independent of their absolute tion can be extracted and compared statisti-
magnitude. It quantifies how much the tensor cally between groups.
deviates from representing isotropic diffusion. In-plane motion Subject motion in the plane
It has a range between zero and one—from parallel to the slice-orientation
perfect isotropy to perfect anisotropy. Interleaved acquisition Scan in which the
Free diffusion Diffusion unimpeded by slices are acquired in an interleaved fashion,
boundaries or obstacles, such as in the center e.g., first all the odd slices are acquired and
of a glass of water. then all the even slices (1, 3, 5, … 2, 4, 6, …).
Generalized fractional anisotropy A metric Inter-slice instabilities A difference between
to characterize a diffusion or fiber ODF, given the odd and even slices as a result of for
as the standard deviation of the ODF over its example subject motion, during scanning of
RMS value. a single volume in an interleaved acquisition
Geometric distortions Distortions in the Isotropic diffusion Diffusion of atoms or
image, causing a mismatch between the image molecules that is orientationally invariant—
and the geometry or anatomy of the object i.e., the rate of diffusion is the same in every
being scanned. direction.
Gibbs ringing Steep intensity transitions in k-space The spatial frequency spectrum of the
the image cannot be represented correctly in MR image as acquired directly from the MRI
the k-space domain, resulting in artifactual scanner. A Fourier transform can be used to
ringing in the image around this transition convert k-space to an image.
Global tractography A simultaneously global Kurtosis The kurtosis is a dimensionless sta-
and local estimation of a pathway, driven by tistical metric that quantifies the deviation
the data. Less affected by error propagation from Gaussianity of an arbitrary distribution.
than classical deterministic or probabilistic Linear least squares (LLS) A fitting method
approaches. that is fast but not very accurate. It is perfectly
Grey matter One of the major tissue classes suited to generate maps for qualitative use
in the brain, consisting of the neuronal bod- though.
ies (soma) and dendritic projections, as well Mean diffusivity (MD) The average of the
as neuroglia. three eigenvalues. Represents a rotationally
HARDI High angular resolution diffusion invariant average ADC value for each voxel.
imaging—a general type of diffusion-weighted Mean Kurtosis (MK) The average apparent
MRI acquisition, whereby a relatively large kurtosis coefficient
424 Glossary

Multi-shell Referring to a diffusion MRI on the theory of q-space. Relies on the approx-
acquisition with more than one non-zero imation that all spins diffuse by the same fixed
b-value. distance, coupled with a maximum entropy
Multi-shell HARDI An extension of the constraint.
HARDI acquisition consisting of multiple Phase Fundamental property of the MRI
HARDI acquisitions (shells) each its distinct signal, which is used in image generation.
b-value. Unwanted/incorrect phase is the most com-
Multi-shot Refers to the acquisition of MRI mon source of artifacts in EPI.
data whereby segments of k-space are acquired Physically implausible signals Signals that
with different excitations. In DTI, specialist could not exist physically, i.e., signals in the
multi-shot EPI techniques may reduce image diffusion-weighted scans that are higher than
artifacts at the expense of increased scan time in the corresponding voxels of image without,
and relatively complex post-processing. or with a lower, diffusion-weighting.
Multi-tensor fitting A method for estimat- Principal eigenvector The eigenvector associ-
ing the fiber orientations and partial volume ated to the largest eigenvalue. In voxels con-
fractions, based on a model of white matter taining a single coherent bundle of axons, the
whereby each fiber population is modeled by principal eigenvector indicates the local orien-
its own diffusion tensor. tation of the axon bundle.
Narrow pulse approximation The main Probabilistic tractography Tractography
assumption for the theory of q-space to be based on random sampling of the distribu-
valid, whereby the duration of the DW gradi- tion of the diffusion orientation in each voxel,
ent pulses is sufficiently short that motion of resulting in a tract probability map.
the spins during that time can be neglected. Projection fibers White matter connections
Nonlinear least squares (NLS) A fitting between the cortex and lower brain areas, as
method that solves the correctly formulated well as the spinal cord.
fitting problem in DTI. It requires a lot of Propagation of uncertainty Increased local-
computation time and resources though, and ization error in a computed tract based on
can still get stuck in local optima. The non- the error in the orientation estimation of each
linear search allows the incorporation of extra individual voxel.
constraints, such as not allowing for tensors Pulsation Pulsation of the veins and ventri-
with negative eigenvalues. cles as a result of cardiac pulsation. A source
Normalization (Image) The process of bring- of physiological image artifacts that may be
ing images into a common reference space. ameliorated using cardiac-gating during data
See also, Registration. acquisition.
Nyquist ghosting A copy of the image (the Q-ball imaging A method to estimate the dif-
‘ghost’) that is shifted by half a field-of-view fusion ODF, based on the theory of q-space.
in the phase-encoding direction, due to a mis- Relies on large q-values (cf. b-values) and the
calibration in the acquisition of EPI images. Funk-Radon transform to obtain an estimate
Parallel Imaging A method of speeding up of the radial projection of the average spin
image acquisition and reducing off-resonance propagator from HARDI data.
distortions in EPI by acquiring a reduced q-space A theory relating the DW signal mea-
amount of k-space. Common approaches sured over a range of different q-values to the
include GRAPPA and SENSE. Fourier transform of the average spin propa-
Partial Fourier A method of speeding up gator. Forms the basis of all methods that esti-
image acquisition by exploiting the symmetry mate the diffusion ODF, and valid under the
of k-space. In partial-fourier techniques, only narrow pulse approximation.
the lower frequencies and half the higher fre- Quality assurance Procedure to ensure and
quencies in k-space are sampled. check data quality, including scanner perfor-
Persistent Angular Structure MRI A method mance, inspection of the DWIs, tensor fits,
for the estimation of the diffusion ODF, based and any subsequent processing.
Glossary 425

q-value, q-vector A measure of the strength Robust estimation of tensors by outlier


of the applied diffusion-weighting, given as rejection (RESTORE) A nonlinear fitting
the product of the DW gradient pulse ampli- method that can handle outliers (by detecting
tude and its duration. The q-vector refers to and rejecting them). It requires, on average,
the q-value combined with the direction along triple the computation time as compared to
which the corresponding DW gradient is NLS. Data redundancy is also a requirement
applied. in order for a robust outlier detection, whilst
Radial diffusivity (or transverse diffusiv- ensuring enough data to work with after the
ity) Equals the average of the smallest two outlier rejection.
eigenvalues, and thus represents an average ROI (see Region-of-Interest)
ADC for the directions perpendicular to the Rotationally invariant tensor based scalar
orientation of an axonal bundle. measures or properties that are independent of
Radial Kurtosis The average apparent kurto- tensor orientation, and thus describe aspects
sis coefficient, measured in the plane perpen- of its size or shape. Rotationally invariant ten-
dicular to the principal direction of diffusion. sor measures are typically calculated based on
Random walk A path constructed of a series the eigenvalues.
of steps of random deviation, i.e., each step Rotationally variant Dependent or influenced
is random and independent of previous steps; by the orientation of the tensor/microstructure
the location of the path at any time step cannot relative to the diffusion-weighting gradient.
be definitively predicted but only statistically Seed point Single initiation point of tractog-
described. raphy in one voxel. Practically, many seed
Real-time prospective motion correction The points may be generated, e.g., in a cross-
real-time adjustment of the scan parameters, sectional slice.
including slice location and orientation, gradi- Self-diffusion coefficient A measure of the
ents, and readout to correct for subject motion. freedom of movement of any single mol-
Region-of-interest (ROI) An a priori selected ecule of a certain substance, within the sub-
area for investigation, which may be defined stance itself. The self-diffusion coefficient
manually or using an automated approach of water is about 2.2 × 10−3 mm2/s at room
(e.g., based on atlas-labels or tractography). temperature.
Registration (Image) The process whereby Shimming Adapting the magnetic field within
two images are brought into voxel level align- the scanner to improve the field homogeneity
ment. The target image is warped into the Signal dropout Artifacts where the signal is
same “space” as the source or reference image artificially low.
based on different types of information in Signal-to-noise ratio (SNR) The ratio between
either or both images. the measured signal and the background noise
Residual The difference between the measured Single-Shot Refers to the acquisition of a
data and the values evaluated from a model whole 2D image in a single excitation. Single-
fit to the data. A good fit would result in the shot EPI allows a full DTI dataset to be
residuals containing all the measurement noise acquired in a very short amount of time com-
as well as outliers caused by various artifacts. pared to other approaches.
Restricted diffusion Diffusion that is con- Slice dropout Artifacts where the signal in the
strained by impermeable boundaries, such entire slice is artificially low.
that the maximum possible displacement for Smoothing (in VBA) The application of a
a diffusing molecule has a strict limit in any (usually) Gaussian kernel to the image, which
direction, which impinges on the boundary. results in alteration of the voxel-wise metric
Reverse polarity gradient method values according to the full-width half maxi-
(RPGM) Acquires EPI images with oppo- mum of the kernel (typically 3–8 mm). Used
site phase-encoding direction in order to per- to increase signal-to-noise and reduce intra-
form image unwarping. subject variability.
426 Glossary

Spherical deconvolution A method to esti- values or the sum of the diagonal elements of
mate the fiber ODF, based on the assumption the diffusion tensor.
of a canonical fiber population with fixed dif- Track Density Imaging A method for the
fusion properties. reconstruction of high-resolution images based
Spin propagator A function giving the prob- on the results of whole-brain fiber-tracking.
ability that a spin (e.g., a water molecule) (Fiber) Tract Reconstructed pathway on milli-
initially at position x0 will have move to x1 meter scale in DWMRI data using a computer
during the diffusion time τ, i.e., P(x1|x0,τ). algorithm. Often confused with anatomical
Also referred to as the displacement prob- term “white matter fiber bundle.”
ability density function. In diffusion MRI, the Tract-based spatial statistics A popular type
quantity measured is typically the average of voxel-based analysis that evaluates changes
spin propagator, the average probability that a in a skeleton comprising a limited amount of
spin chosen at random from within the voxel white matter, in order to increase sensitivity
will have moved by a distance r, i.e., P(r|τ). by reducing registration error and partial vol-
Streamline Virtual reconstruction of a single ume effects.
pathway through the diffusion field. Twice-refocused spin echo (TRSE) Diffusion
Subject motion Bulk motion of the imaged preparation with two 180° refocusing pulses,
participant during scanning. designed to cancel eddy-currents
Susceptibility The degree of magnetization in Unwarping Image processing methods to
response to a magnetic field. Susceptibility correct or reduce susceptibility-induced
artifacts arise near the interfaces of matter distortions.
with different magnetic susceptibility, e.g., air VBA (see Voxel-based analysis)
and tissue. Voxel-based analysis (DTI) An exploratory
Susceptibility-induced distortions Image dis- approach often used in group analysis to
tortions due to field inhomogeneities that are investigate voxel-wise alterations in DTI
especially present in EPI acquisitions parameters in brain white matter. A VBA pipe-
T2 shine-through A term referring to the fact line typically consists of an image normaliza-
that a DWI (not normalized) represents par- tion step, a smoothing step and statistics that
tially decayed T2 weighted signal. A high control family-wise error. See also TBSS.
intensity in a DWI can thus also be caused by Weighted linear least squares (WLLS) A
an originally high T2 intensity. To rule out this fitting method that is still fast yet more accu-
effect, DWIs should be normalized by a B0, or rate than LLS. It accounts for the logarithmic
ADC maps should be used instead. transformation of the data (and the noise)
TBSS (see Tract-based spatial statistics) up to some extent, but is confronted with a
Template A reference image constructed by chicken-and-egg problem to tackle this issue.
coregistering and averaging multiple scans in Westin measures A set of metrics that charac-
order to create a representation of the image terize the geometric properties of the diffusion
characteristics of a given study population. tensor, including linear (Cl), planar (Cp) and
Templates are used for image normalization. spherical (Cs) components.
Standard templates are often used to report White matter One of the major tissue classes
research findings. in the brain, consisting of tightly packed, pre-
Tensor residuals The ‘residual’ of the dominantly myelinated neuronal axons and
diffusion-weighted signals after tensor esti- associated neuroglia that link functional areas
mation, i.e., the absolute average difference of the central nervous system.
between signal and fit (see also, Residual). White matter bundle Anatomical structure
Through-plane motion Subject motion in the comprising ten thousands (and in some bun-
direction perpendicular to the slice-orientation dles, millions) of axons connecting distant
Trace A measure of average diffusivity in a mainly grey matter brain regions. Although
voxel, calculated by summing the three eigen- this term includes anatomical structures with
Glossary 427

“tract” in their anatomical name, e.g., cortico- locally in all voxels of the brain mask in
spinal tract; it is not synonymous with virtu- order to reconstruct the entire image trac-
ally reconstructed “fiber tracts” or tracks. togram. May be confused with Global trac-
Whole-brain tractography A tractography tography, which is a different and unrelated
approach whereby streamlines are initiated technique.
Index

A Apparent diffusion tensor


Abscess cysts, 311 DTI theory, 47–48
ACM. See Anatomical connectivity mapping (ACM) motivation and implications, 46–47
Acute disseminated encephalomyelitis (ADEM), Arachnoid cysts
332, 339 epidermoid tumors, 312
AD. See Alzheimer’s disease (AD). See Axial FLAIR and DWI, 312
diffusivity (AD) necrotic brain tumor, 309
ADC. See Apparent diffusion coefficient (ADC). Arcuate fasciculus, 266–269
See Diffusion tensor imaging (DTI) Association fibres, 205
ADEM. See Acute disseminated encephalomyelitis Atlas-based tractography
(ADEM) definition, 221
ADHD. See Attention deficit hyperactivity disorder ROIs, 219
(ADHD) rROIs, 221
Affine registration, 187 TRACULA, 220, 222
Alzheimer’s disease (AD). See Mild cognitive ATLR. See Anterior temporal lobe resection (ATLR)
impairment (MCI) Attention deficit hyperactivity disorder (ADHD), 368
American Psychiatric Association, 362 Automated tractography
Anatomical connectivity mapping (ACM), 347 atlas-based, 221–222
Anatomy, 233, 260–270 manual reconstruction, 221
2D, 233 white matter atlas, 221
3D Axial diffusivity (AD), 68, 410
arcuate fasciculus, 265–269 diffusion ellipsoid, 78
cerebral peduncles, 268–270 eigenvalues, 78
cingulum, 260, 261 myelin and axonal density, 78
corpus callosum, 233, 260, 261 Axial FA map color coded, 224
fornix, 260, 262 Axial slices, 233–242
inferior fronto-occipital fasciculus, 263–265 Axonal scale, 222
inferior longitudinal fasciculus, 262, 263
superior fronto-occipital fasciculus, 265–269
superior longitudinal fasciculus, 265–269 B
Anisotropy BD. See Bipolar disorder (BD)
ADC maps, 44, 46 Behavioral variant of frontotemporal dementia
advantages, 43 (BvFTD), 349
complications, 41–43 Biological confounds, 80–81
DWIs, 43, 45 complex tissue architecture, 80–81
ellipsoid eccentricity, 69 crossing fibres
FA, 69–70 AD and RD, 81
gradient table, 43, 44, 46 FA, 80
Anterior temporal lobe resection (ATLR), 396 MD, 81
Apparent diffusion coefficient (ADC), 10, 34, 90, 309 trace, 80–81
advantages, 40–41 demographics, 80
b-value, 40 timing, 80
hindered/restricted diffusion, 39 Bipolar disorder (BD), 366
minimum requirements, 41 Black holes, 335

© Springer Science+Business Media New York 2016 429


W. Van Hecke et al. (eds.), Diffusion Tensor Imaging, DOI 10.1007/978-1-4939-3118-7
430 Index

Brain analysis techniques DW-MRI, 222


anatomical image, 157 foresight, 223, 224
voxels, 157 hypothesized, 222
Brain tumors, DTI, 313–320 large-scale, 222
ADC, 309 measurement, 222, 223
characterization nonexisting, 223
glioblastoma multiforme vs. lymphoma, 314–315 quantify, 222
meningiomas, 315–317 streamline/fibre count, 223, 224
posterior fossa tumors, 317–320 white matter, 222
solitary metastasis vs. glioblastoma multiforme, Constant solid angle-QBI (CSA-QBI), 391
313–314 Constrained spherical deconvolution (CSD), 209
diagnosis and follow-up, 324–326 Coronal slices, 233, 243–251
LGGs and HGGs, 311 Corpus callosum (CC), 135, 233, 260, 261
b-Value Cortical lesions (CLs), 336
effect, 74 Corticospinal (pyramidal) tract (CST)
diffusion weighting, 31–33 cerebral peduncle, 298
BvFTD. See Behavioural variant of frontotemporal HARDI, 299
dementia (BvFTD) motor function, 293
PLIC, 299
thalamocortical fibers, 298
C Creutzfeld–Jakob disease (CJD)
Carr-Purcell-Meiboom-Gill (CPMG), 116 akinetic mutism, 345
Central nervous system (CNS), 331 ataxia and myoclonus, 344
Cerebral peduncles, 268–270 cortical and basal ganglia degeneration, 345
Cerebrospinal fluid (CSF), 332 dementing syndrome, 344
Challenges in TBI, 379 DWI, 344
analyzing DTI FA, 345
ROI, 379 MD, 345
WB, 379 neocortical, limbic and subcortical gray matter
fiber disruption and axonal degeneration, 378 hyperintensities, 344, 345
timing, DTI scanning, 379 sCJD, 344
transportation and scanning issues, 378 vacuolation, 344
CHARMED. See Composite hindered and restricted CSA-QBI. See Constant solid angle-QBI (CSA-QBI)
model of diffusion (CHARMED) CSD, 222
Chemical shift artifact, 142–143 CSF. See Cerebrospinal fluid (CSF)
Children, DTI CSF contamination, diffusion metrics, 82–83
brain development, 279 CST. See Corticospinal (pyramidal) tract (CST)
child-centered approach
SBN, 280
scanning equipment and protocol, 279 D
Cingulum bundle (CB), 260, 261 Data acquisition, 13–15, 232
CIS. See Clinically isolated syndrome (CIS) Data preprocessing, 15, 233
CJD. See Creutzfeld–Jakob disease (CJD) Data requirements, tractography, 208, 209
Clinical practice, DTI b-value, 209–211
children, DTI, 279, 280 field strength, 210, 211
data collection, 276 gradient directions/signal averages, 209–212
ethical considerations, 280 resolution
and research, 275–276 anisotropic resolution, 209
Clinically isolated syndrome (CIS), 335 crossing fibre, 208
CLs. See Cortical lesions (CLs) false positive tracking results, 208
CNS. See Central nervous system (CNS) low spatial resolution, 208
Color-coded super-resolution TDI, 224 ROI placement, 208
Color-coded track-density image (TDI), 224 tracking algorithms, 209
Commissural fibres, 205 U-fibres, 208
Compartment models, 222 voxel, 208
Composite hindered and restricted model of diffusion DBS. See Deep brain stimulation (DBS)
(CHARMED) model, 386, 414 Deep brain stimulation (DBS), 362
Connectivity DRT, 296
anatomically implausible, 222 DTI data, 296
definition, 222 myoclonus dystonia, 296
Index 431

STN and GPi, 296 three-dimensional (3D) Gaussian diffusion model,


VIM, 296 409, 410
Dementia Diffusion kurtosis processing
AD, 343, 346–349 eddy currents, 412
CJD, 344–345 encoding amplitude and gradient, 412
DWI, 344 Gibbs ringing/misalignment, 413
FTLD, 349–352 LLS, 413
Dementia with Lewy bodies (DLB) NLS, 413
vs. AD, 353 subject motion, 412
cingulum and precuneus, 353 water molecules, 413
DTI, 353 WLLS, 413
FA reduction, 353, 354 Diffusion orientation density function (dODF)
Demyelinating diseases, 332 clinical and neuroscientific studies, 392
acquisition protocol CSA-QBI, 391
CSF, 332 DOT, 391
EPI technique, 332 fiber orientations, 392
k-space, 332 GFA, 392
PROPELLER technique, 332 model-free nature, 392
ADEM, 339 PAS-MRI, 391
DTI data analysis, 333–334 QBI, 391
NMO, 338–339 spherical deconvolution operation, 392
pathological abnormalities, 334 tissue microstructure, 392
Dentate-rubral-thalamic tract (DRT), 296 Diffusion orientation transform (DOT), 392
Deterministic tractography Diffusion spectrum imaging (DSI), 390
corpus callosum, 206, 207 Diffusion tensor (DT), 331
curvature threshold, 206, 207 Diffusion tensor imaging (DTI)
FA, 206, 208 acute ischemia and brain lesions, 7
masking, 206 advanced concepts and techniques, 4
parameter values, 206 analysis strategies, 15, 16
principal, 206 analysis technique, role of, 83
seed point, 206, 207 anisotropic diffusion, 8
DICOM. See Digital information and communication in applications of, 11, 13
medicine (DICOM) axonal architecture, 9, 11
Diffusion kurtosis acquisition challenges of interpretation, 79
b-value, 411 clinical applications, 4
diffusion-weighted images, 411 clinical MRI techniques, 8
DTI, 411 in clinical practice, 17–18
SGP, 412 complementary measures, 78–79
Diffusion kurtosis applications data acquisition, 13–15
complementary research, 414 data pre-processing, 15
Gaussian DTI model, 413 decision schemes, 4–5
high-grade and low-grade cerebral gliomas, 413 fiber tracts, presurgical planning work-up, 13
ischemic tissue injury, 414 geometric representation, 10, 12
Diffusion kurtosis imaging (DKI) heterogeneous microstructural environments, 9
acquisition, 411–412 improved directional contrast with, 8, 9
applications, 413–414 interpretation of, 16, 17
arbitrary distribution, 408 isotropic diffusion, 8
coefficient, 408–409 model limitations, 79–80
definition, 405 neurodevelopment/aging, 13
dimensionless statistical metric, 408 practical implementation, 3
free diffusion, 405 prototypal DTI study pipeline, 13, 14
hindered and restricted diffusion, 406–408 quantitative technique, 10–11
limitations software packages, 4
biophysical model, 414 strengths and limitations, 17, 18
CHARMED model, 414 tissue orientation, applied diffusion gradient, 9, 10
white matter model, 414 Diffusion tensor imaging fiber tractography
parameters (see Kurtosis parameters) (DTI FT), 292
PDF, 406 Diffusion tensor information (DTI), 192
post processing (see Diffusion kurtosis standard templates, 193
processing) templates, 193
432 Index

Diffusion-weighted imaging (DWI), 344 image and k-space, 135


EPI-DTI, 98 Image processing stage, 143
hardware limitations, 102 image registration, 132
motion artifacts, 119 interslice instabilities, 133
motion-induced phase error, 119 processing stages, 127
motion sensitivity, 117–120 signal and slice dropouts, 144–145
MRI, 98 table vibrations, 133
nonrigid phase errors, 119 tensor estimation, 136
parallel imaging, 120 DTI findings in psychiatric disorders, 366–369
reconstruction, 120 anxiety disorders
scan parameters, 95 GAD, 367
susceptibility artifacts, 106 OCD, 367
Diffusion-weighted MRI (DWMRI), 205 PTSD, 367
measurements, 76 limbic system, tracts, 365
Diffusivity measurement mood disorders (see Mood disorders, DTI)
AD, 68 neurodevelopmental disorders
MD, 68 ADHD, 368
RD, 68 alcohol use disorders, 368, 369
trace (Tr), 67 autism, 368
Westin measures, 68 personality disorders, 367
Digital information and communication in medicine schizophrenia, 366
(DICOM), 282 white matter, 365
Direction-encoded color (DEC) map, 130–131 DTI FT. See Diffusion tensor imaging fiber tractography
DKI. See Diffusion Kurtosis imaging (DKI) (DTI FT)
DLB. See Dementia with Lewy bodies (DLB) DTI-EPI diffusion gradients, 113
dODF. See Diffusion orientation density function DTI method
(dODF) analysis techniques and approaches, 156
DOT. See Diffusion orientation transform (DOT) axial slice, 156
Double spin-echo (DSE) pulse sequence, 123 brain analysis techniques, 157–158
DRT. See Dentate-rubral-thalamic tract (DRT) brain regional level, 156
DSI. See Diffusion spectrum imaging (DSI) checklist, 170, 171
DT. See Diffusion tensor (DT) data analysis, 168
DTI acquisition, 89–92 data processing, 170
brain scan, 109 data quality, 156, 168
coil elements, 110 DWI, 155
complex number, 92 FA maps, 155
description, 89 factors, 167, 168
EPI, 93–95 guidelines, 167
gradient performance, 111 memory game, 172
K-Space (see K-Space) optimal analysis approach, 166
metal, 109 pipeline, 154
non-diffusion images, 108 population
parallel imaging/multi-shot regimes, 109 characteristics, 167–168
pediatric considerations, 124 composition, 167
phase angles, 93 pathology, 168
PNS, 112 purpose, 156
SENSE/ASSET scan, 110 regional information, 158
signal magnitude, 92 software and hardware, 168
speed, 120 software packages, 170
Stejskal-Tanner diffusion-encoding (see stress, 170
Stejskal-Tanner diffusion-encoding) DTI in TBI, 376
troubleshooting, 107 pathophysiology
DTI data intracellular (cytotoxic), 376
artifacts, 133 traumatic axonal injury, 376
diffusion MRI data, 127 vasogenic (extracellular), 376
diffusion protocols, 134 prognostic predictor, 377
DWIs, 127, 131 Dual spin-echo (DSE) diffusion imaging,
eddy currents, 129 113, 130
FA and MD, 134 DWI. See Diffusion-weighted imaging (DWI)
guidelines, 129 DWIs number of, effect, 73
Index 433

E Fat band, 142


Echo-planar imaging (EPI), 331 Fat suppression method, 109
advantages, 95 Fiber assignment by continuous tracking (FACT),
b-value, 97 231, 297
Cartesian, 99 Fiber count, 78, 223, 224
chemical shift artifacts, 99 Fiber orientation distribution (FOD)
DTI, 93 fiber configurations, 387
sequence, 232 fiber-tracking, 387, 388
source, 104 linear algebra, 387
TE, 98 sphere, 387
Eddy current correction, 123–124 Fiber orientations estimation, 386–389
Eddy currents, 233 anisotropy assumption
Eigendecomposition, 51–52 fiber effects, 388
Eigenvalues and eigenvectors HARDI data, 389
alternatives, 51 partial volume effects, 388
eigendecomposition, 51–52 tensor-derived metrics, 388
in practice, 52–53 multi-tensor fitting (see Multi-tensor sensor
tensor elements, 50–51 approach)
Eloquent WM pathways, 293, 294 spherical deconvolution (see Fibre orientations
categorization, 293 estimation: spherical deconvolution method)
cortical and subcortical structures, 292 Fiber tracking, 231
DTI FT, 295 biopsy, 304
fiber tracking method, 295 CC, 233
fMRI, 292 dependency, 232
functional mapping, 295 fMRI, 292
IFOF and UF, 295 fornix, 260
motor hemisphere dominance, 295
central cerebral peduncle, 293 inferior longitudinal fasciculus, 262
CST, 293 navigation systems, 304
FA maps, 294 non-commutative property of, 211, 213
ISM, 293 principle of, 232
Karnofsky scores, 294 property, 232
MEP, 294 robust, 299
neoplasm, 294 scalar and directional data, 297
SEP, 294 suboptimal/failed, 297
single and multi tensor approach, 293 tractography algorithm, 233
SMA syndrome, 294 Field strength effect, 72–73, 210–211
somatotopic mapping, 293 Field-of-view (FOV), 131
neuronavigation systems, 295 Fornix, 260, 262
OR, 294 Fractional anisotropy (FA), 10, 184, 206, 245, 309,
ROI, 292 382, 383
SLF/AF, 294–295 DTI indices, 76
susceptibility-related artifacts, 294 integrity, 77
tractography, 294, 295 macromolecules, organelles and membranes, 77
Ependymoma (EP), 317 magnitude, anisotropic components, 69
EPI. See Echo planar imaging (EPI) maps of, 56, 57
Epidermoid cysts regional anisotropy, 77
DW trace images, 312 standard deviation, eigenvalues, 56
ectodermal tissue, 312 tissue microstructure, 70
granulomatous meningitis, 312 Free diffusion. See Hindered diffusion
intraparenchymal, 312 Frontotemporal lobar degeneration (FTLD), 349, 350
Extracellular region, hindered diffusion, 25 vs. AD, 351–352
bvFTD, 349–350
DTI vs. pathology and genetic background, 349
F nonfluent/agrammatism, 349
FA. See Fractional anisotropy (FA) PPA
FACT. See Fiber assignment by continuous tracking atrophy, 350
(FACT) logopenic variant, 350
FACT-algorithm, 231 nonfluent and semantic variants, 350
Fast spin-echo (FSE), 105, 116 phenotypes, 349
434 Index

Frontotemporal lobar degeneration (FTLD) (cont.) FA, 383


ROI-based/tractography DTI, 350 N parameters, 385
TBSS, 350 spin propagator
voxel-wise approach, 350 average, 389
FSL BedpostX, 217–218 DSI, 390
FSL flirt, 233 DW signal, 389
Full width at half maximum (FWHM), 195 q-vectors, 390
spin displacement probability density
function, 389
G structural integrity AFD, 399
GAD. See Generalized anxiety disorder (GAD) structural integrity MND, 399
Galantamine, 348 tractography (see Tractography techniques)
Gaussian kernel, 195 High definition fiber tracking method (HDFT), 397
GBM. See Glioblastoma multiforme (GBM) Higher order model tractography, 219, 221
GCC. See Genus of the corpus callosum (GCC) High-field scanners, 211
Generalized anxiety disorder (GAD), 367 High-grade gliomas (HGGs), 311
Generalized fractional anisotropy (GFA), 392 Hindered diffusion
Genus of the corpus callosum (GCC), 43 ADC, 34
GFA. See Generalized fractional anisotropy (GFA) extracellular region, 25
Ghosting, 99 Gaussian dispersion pattern, 34
Gibbs Ringing, 140–142 restricted water inside and around boundaries, 25, 26
Gibbs ringing artifacts, 140, 141 RMS displacement, 34
Glioblastoma multiforme (GBM), 311 water molecules restricted inside cylindrical
vs. lymphoma, 314–315 boundary, 26, 27
perilesional edema, 311 Hindered/restricted diffusion
vs. solitary metastasis, 313–314 biological tissues, 406
Global tracking algorithm, 233 bounding microstructure, 408
Global tractography, 223 Gaussian distribution, 407
Globus pallidus interna (GPi), 296 ventricular system, 406
GM. See Gray matter (GM) Histogram analysis
GPi. See Globus pallidus interna (GPi) parameters, 158
Gray matter (GM), 335, 336 Human resources, 285, 286
Groups of methods operate in either k-space cost, 286
(GRAPPA), 120 support staff, 285
time
acquisition, 285
H analysis/off-line processing, 286
HARDI. See High-angular resolution diffusion imaging outsourcing, 286
(HARDI) preparation, 285
HDFT. See High definition fiber tracking method radiological reporting, 286
(HDFT)
HGGs. See High-grade gliomas (HGGs)
High-angular resolution diffusion imaging (HARDI), 13, I
221, 386–399 IFOF. See Inferior frontal occipital fasciculus (IFOF)
acquisition strategy, 384 ILF. See Inferior longitudinal fasciculus (ILF)
b-value, 384 Image acquisition, DTI parameters, 71–72
connectivity (see Structural connectivity assessment) Image quality
crossing fibers, resolve (see Fiber orientations artifacts, 75–76
estimation) noise, 74–75
data acquisition Image registration techniques, 184, 278
b-value, 392 global, 187
SNR, 393 local deformation, 187
diffusion process characterise rigid-body transformation, 187
dODF, 390–392 Implausible connections, 222
model-free techniques, 389 Inferior frontal-occipital fasciculus (IFOF),
narrow pulse approximation, 391–392 263–265, 301
q-space (see Spin propagator) Inferior longitudinal fasciculus (ILF), 2–265, 301
spin propagator, 389–390 Inhomogeneities, 21
diffusion tensor ellipsoid, 382 Interslice instabilities, 133
DW signal, 385 Intracellular region, restricted diffusion, 25
Index 435

Intraoperative navigation random walk, 24


color FA map, 297 restricted diffusion, 25
fiber tracts, 297 sensitizing, 30
Intraoperative stimulation mapping (ISM), 293 Major depressive disorder (MDD), 6, 36
Inversion time, 143 Manual data quality assurance, 146
ISM. See Intraoperative stimulation mapping (ISM) Mass lesions in brain
Isotropic Gaussian smoothing kernel, 197 abscess, 311
epidermoid and arachnoid cysts, 312
FA values, abscesses, 312
J glioblastoma and metastases, 311
JHU atlas, 177 high- grade gliomas and tumefactive demyelinating,
312–313
necrotic or cystic tumors, 311
K neoplastic cysts, 311
Karnofsky scores, 294 perilesional edema, 311
k-Space, 92 MD. See Mean diffusivity (MD)
k-space trajectory, 137 MDD. See Major depressive disorder (MDD)
Kurtosis parameters Mean diffusivity (MD), 206, 345
axial diffusivity, 410 axial and radial diffusivity, 55
diffusion-weighted MR images, 408 description, 77
eigenvectors and eigenvalues, 410 diagonal tensor elements, 56
fractional anisotropy, 410 diffusivity measures, 68
kurtosis metrics, 410 DTI metrics, stroke, 77
parameter maps, 410, 411 maturation, 78
radial diffusivity, 410 rotationally invariant measures, 55
scatter plots, 410 same rotation invariant MD, 56
water content differences, 78
Medulloblastoma (MB), 317
L Meningiomas, 315
Linear least squares (LLS), 413 factor, 316
ADC values, 59 spindle cells, 316
single-step process, 58 MEP. See Motor evoked potentials (MEP)
sum of squared residuals, 58 Mild cognitive impairment (MCI)
Lipid suppression methods, 113 ACM, 347
Lipid/fat suppression, 113 central nervous system, 346
LLS. See Linear least squares (LLS) cholinesterase inhibition, 347
Longitudinal fasciculus, 215 cortical pathology, 346
Low-grade glioma (LGGs), 315 diagnostic accuracy, 348
Low-intensity voxels, 212 DTI tractography, 346
longitudinal DTI studies, 348
meta-analyses, 346
M MMSE, 346
Magnetic field inhomogeneities, 113 novel strategies, 348–349
Magnetic resonance imaging (MRI), 25–30 Wallerian degeneration, 346
free diffusion, water molecules, 24 WM pathways, 346
hindered diffusion (see Hindered diffusion) Mini-mental state examination (MMSE), 346
isotropic diffusion, 25 MMSE. See Mini-mental state examination (MMSE)
microstructural tissue properties Model-free techniques, 389
astrocytes, 28 Monitoring treatment effects
cellular components, neural tissue, 27, 28 ADC values, 324
cellular structures, 26 contrast-enhancing lesions, 324
grey and white matter contrast, 28, 29 high-grade glioma, 325
microglia, 28 injury and tumor recurrence, 324
myelin sheath, 27 radiochemotherapy, 324
neurites, 27 therapy-induced inflammatory reactions, 324
neurons, building blocks, 27 Mood disorders, DTI
oligodendrocyte cells, 28 BD, 6, 36
optic nerve, healthy rat sliced, 29, 30 depression, 366
physical and mathematical theories, 24 (see also FA reductions, 366
Pulsed gradient spin echo (PGSE) method) MDD, 366
436 Index

Motor evoked potentials (MEP), 294 bulk and physiologic motion, 296
MRI-HARDI-techniques, 292 cardiac gating, 296
MS. See Multiple sclerosis (MS) color FA maps and tractography, 297
Multichannel phased-array head-coils, 211 FACT approach, 297
Multiple sclerosis (MS) fat saturation techniques, 296
CLs, 336 isotropic voxels, 296
CNS, 331 neuroanatomy, 297
demyelinating diseases, 332, 334, 338–339 ROI, 297
DT, 331 intraoperative electrophysiologic mapping, 291
DTI abnormalities, 331 microstructural and macrostructural organization, 291
fiber tracking, 331 misregistration and intraoperative shift, 303
focal WM lesions, 335 multimodal data, 297
GM, 335–336 pathophysiological effects, diffusion properties, 305
MRI, 331 radiation damage, 295–296
NAWM, 335 radiotherapeutic planning, 291
NMO and ADEM, 2, 33 SLF/AF, 300, 301
optical nerve, 336 uncinate fasciculus, 301
spinal cord, 336 white matter tracts and tractography, 304, 305
tractography, 337–338 NICER. See Neuroimaging for coma emergence and
voxel-based method, 338 recovery (NICER)
WM, 331 NIFTI. See Neuroimaging informatics technology
Multi-shell dMRI initiative (NIFTI)
DKI, 70 NLS. See Nonlinear least squares (NLS)
tissue (compartment) model-based approaches, 71 NMO. See Neuromyelitis optica (NMO)
Multi-tensor approach NODDI. See Neurite orientation dispersion and density
CHARMED approach, 386 imaging (NODDI)
nonlinear minimization methods, 386 Nonexisting connections, 223
single tensor model, 386 Nonlinear least squares (NLS), 413
Multi-tensor models, 209 ADCs values, 60
MRI, 61
sum of squared residuals, 60
N Normal-appearing white matter (NAWM), 324
Narrow pulse approximation CIS, 335
DW gradient pulses, 391 DTI metrics, 335
fiber bundles, 392 neuroprotective therapies, 335
tags and untags water molecules, 391 RRMS, 335
NAWM. See Normal appearing white matter (NAWM) T2 lesions, 335
Nerve tracts, 231 Number of signal averages (NSA), 210
Neurite orientation dispersion and density imaging Nyquist ghosting, 140
(NODDI), 71 correction methods, 139
Neurites image Processing Stage, 140
axons, 27 origin, 139
dendrites, 27
Neuroimaging for coma emergence and recovery
(NICER), 377 O
Neuroimaging informatics technology initiative Obsessive-compulsive disorder (OCD), 367
(NIFTI), 282 OCD. See Obsessive-compulsive disorder (OCD)
Neuromyelitis optica (NMO), 332, 338–339 Online tracking result with scanner software, 224, 225
Neurosurgical planning, DTI, 296, 297 Optic nerve, 336
color FA and tractography maps, 292 Optic radiations (OR), 294
cortical and subcortical mapping, 291 Optimal analysis approach, 166
corticospinal (pyramidal) tract, 298, 299 OR. See Optic radiations (OR)
DBS electrode placement, 296 OR/AND/NOT, 212
eloquent WM pathways, 292–295
fiber pathways, 291
HARDI, 291 P
ILF and IFOF, 301 PACS. See Picture archiving and communication system
image acquisition and postprocessing (PACS)
atlas-based segmentation, 297 pAgCC. See Partial agenesis of the corpus callosum
bone and paranasal sinus interfaces, 296 (pAgCC)
Index 437

Partial agenesis of the corpus callosum (pAgCC), 398 multivariate machine learning algorithms, 361
Partial Fourier imaging support vector machines, 361
k-space, 123 white matter abnormalities, 361
TE and TR, 122 clinical research
Partial volume effects (PVE), 178, 278 mood disorders, 360
definition, 82 MRI, 360
DTI analysis, 82 neuroimaging, 360
spatial resolution, 82 schizophrenia, 360
PAS-MRI. See Persistent angular structure MRI T1 scans, 360
(PAS-MRI) comorbidity, 363
Patient group DBS, 362
age DSM-V, 363
developmental phase, 277 heterogeneity, 363, 364
senescent phase, 277–278 illness, 360
effect, brain structure neurobiological models, 360
DTI parameters, 278 psychosurgical procedures, 361, 362
field-map-based techniques, 278 psychotropic medication, 364
frontal and temporal sinuses, 278 PTSD. See Posttraumatic stress disorder (PTSD)
image registration techniques, 278 Pulsation, 134–136
neuroimaging community, 278 Pulsed gradient spin echo (PGSE) method
PVE, 278 b-Value, 31–33
sclerosis, 278 evolution of magnetisation magnetization, 31
effect, patient mobility, 279 evolution of magnetization, 33
patient compliance, 279 Larmor precession, magnetic field gradient, 31, 32
prodromal schizophrenia, 276 pulse sequence, 30, 31
PCNSL. See Primary central nervous system lymphoma signal, 34
(PCNSL) Stejskal-Tanner equation, 30
PDF. See Probability distribution function (PDF) Stejskal-Tanner sequence, 30
Peripheral nerve stimulation (PNS), 103, 111–112 PVE. See Partial volume effects (PVE)
Persistent angular structure MRI (PAS-MRI), 391
PGSE. See Pulsed gradient spin echo (PGSE) method
Picture archiving and communication system Q
(PACS), 282 Q-ball Imaging (QBI)
Pilocytic astrocytoma (PA), 317 CSA, 391
Posttraumatic stress disorder (PTSD), 367 pAgCC, 398
PPA. See Primary progressive aphasia (PPA) tractography, 398
Primary central nervous system lymphoma (PCNSL), QBI. See Q-ball Imaging (QBI)
314, 316 Quality assurance (QA)
Primary progressive aphasia (PPA), 349 and control, 145
Probabilistic tractography tests, 145
automated atlas-based tractography, 219, 220 Quantification in medical imaging, 67–70
distribution, estimated possible orientations, 216, 217 ADC values, 67
mapping connections, 217 advanced and automated tools, 65
multiple tracts, 216, 217 anisotropy measures (see Anisotropy)
propagate/accumulate, 215 diffusivity measures (see Diffusivity measures)
Probability distribution function (PDF), 406 dMRI data, 66
Projection fibres, 205 ellipsoid, 66
PROPELLER technique, 332 example parameter maps, 66, 69 (see also Multi-shell
Protocol dMRI)
data acquisition, 232 radiological diagnosis, 65
data preprocessing, 233
tracking algorithm, 233
Psychiatry, DTI, 360–365 R
application Radial diffusivity (RD), 68, 410. See also Axial
diagnoses, 362, 363 diffusivity (AD)
neuroimaging, 363 Radiofrequency (RF) system, 103
scanner, 364, 365 Random Brownian motion, 222
biomarkers RD. See Radial diffusivity (RD)
learning dataset, 361 Readout-segmented EPI (RS-EPI), 115
machine learning, 361 Reference regions of interest (rROIs), 221
438 Index

Region of interest (ROI), 158–160, 211 multimodal viewing and analysis, 285
ADC, 176 version control, 283–284
axonal damage, 377 Seed points, 206, 212
corpus callosum, 176 Self-diffusion coefficient
definition, 175 description, 37
diffusion images, 178 diffusion weighting, 38
FA and MD value, 178 free diffusion, 38
FA map, 177 minimum requirements, 39
placement, 181 Stejskal-Tanner equation, 38
position and size, 175 SEP. See Somatosensory evoked potential (SEP)
potential myelin, 377 SGP. See Short gradient pulse (SGP)
registration, 179 Shimming, 137
spatial normalization, 180 Short axis PROPELLER (SAP), 115
statistical analysis, 180 Short gradient pulse (SGP), 412
and volume, 292 Signal-to-noise ratio (SNR), 94
voxel-based analysis, 176 Single-seed ROI, 214
white matter, 379 vs. whole-brain tractography, 214
Region-specific analysis techniques Slew rate, 281
diffusion measurement, 158 SLF/AF. See Superior longitudinal/arcuate fasciculus
Relapsing-remitting multiple sclerosis (RRMS), 335 (SLF/AF)
RESTORE. See Robust estimation of tensors by outlier Slice-selection gradient reversal (SSGR)
rejection (RESTORE) method, 143
Restricted diffusion, 25 SMA syndrome, 294
Reverse polarity gradient method, 138 Smoothing, 183
Rigid-body transformation, 187 axial FA slice, 195
Robust estimation of tensors by outlier rejection in DTI, 197
(RESTORE) DTI data sets, 196
data redundancy, 61 FWHM, 195
DTI model, 61 Gaussian kernel, 195
outliers, 61 parameter, 196, 199–200
ROI. See Region of interest (ROI) parametric/nonparametric Statistics, 199
ROI-drawing statistical analysis, 198–199
cingulum bundle, 212, 216 Somatosensory evoked potential (SEP), 294
FA, 208, 209, 214 Spatial normalization, 180
guidelines, 215 Specific absorption rate (SAR), 103
spatial awareness and interpretation, 212 Spherical deconvolution method
tracking procedures, 213 advantages, 387
tract volume, 213, 214 fiber anisotropy, 387
RRMS. See Relapsing-remitting multiple sclerosis FOD, 387
(RRMS) fODF, 387
orientation plots, 387
volume fractions, 387
S Spinal cord, 336
Sagittal slices, 233, 252–259 Spin-echo echo-planar imaging (SE EPI), 127
SBN. See Scanner background noise (SBN) Squared intensity differences (SSD), 188
Scan parameters, 103 Standard templates
Scanner background noise (SBN), 280 advantages, 193, 195
Scanner resources, 280–285 healthy control group, 193
hardware study-specific atlases, 193
gradient system, 281 Stejskal-Tanner diffusion-encoding
magnetic field strength, 280–281 ADC, 90
peripheral equipment, 281 cellular structure, 90
scanners, 281–282 DWI and DTI, 90
software elements, 91
data management, 282–283 HARDI, 90
data security, 282–283 3D sphere of diffusion, 90
data storage, 282 Stejskal-Tanner equation, 30
data transfer, 283 Stejskal-Tanner sequence, 30
features, 284–285 Stimulated echo (STE) diffusion, 113
licensing issues, 283 STN. See Subthalamic nucleus (STN)
Index 439

Structural connectivity assessment ROI-drawing, 212–218


fiber density, 399 ROIs, 211
FRDA pathology, 399 seeding, 207, 211, 212
frontal cortex, 399 Tract-based spatial statistics (TBSS), 38, 350
limitations, 399 Tractography, 206
pAgCC, 398 applications, 206
Subthalamic nucleus (STN), 296 association fibres, 205
Superior fronto-occipital fasciculus (SFOF), 265–269 ATLR, 396
Superior longitudinal/arcuate fasciculus (SLF/AF), 294, automated, 221–222
300, 301 blue voxel, 162
Superior longitudinal fasciculus (SLF), 265–269 brain, 162, 394
Surgical planning, DWI color FA maps, 297
biopsy determination, 323 connecting dots, 161
brain tumor neuroradiology, 309 corpus callosum, 163
tumor boundaries, 321–323 CSD-based method, 394
tumor extension assessment, 323 CST, 94, 2
Susceptibility-induced distortions, 136–139 deterministic (see Deterministic tractography)
DTI, 161, 297
false positives and negatives, 206
T green voxel, 161
TBI classification HDFT, 397
axial and radial diffusivity, 377 higher order model, 219, 221
cerebral lobar white matter, 377 Meyer’s loop, 395
chronic traumatic brain injury, 377 MRI and ROIs, 161
corpus callosum, 377 neoplasm, 305
intra- and extracellular water, 377 neuropsychiatric problems, 296
intracranial lesions, 377 neurosurgical literature, 394
neural pathways, 376 noise and artifacts affect tract, 163
TBSS. See Tract-based spatial statistics (TBSS) OR, 295
TDL. See Tumefactive demyelinating lesions (TDL) probabilistic, 215–219
Teem library, 233 projection fibres, 205
Tensor elements q-ball tractography, 394
DWI and ADC values, 50 region specific analysis, 1, 63
gradient directions, 50 ROIs, 162
off-diagonal elements, 49 routine clinical practice, 224, 225
in practice, 48–49 segmented tract, 162
Tensor ellipsoids, 231 single and multi tensor technique, 394
Tensor fitting methods, 58–61 software packages, 224
LLS (see Linear least squares (LLS)) terminology, 162
NLS (see Nonlinear least squares (NLS)) training session, 214, 216
RESTORE (see Robust estimation of tensors by true anatomic and functional localization, 303
outlier rejection (RESTORE)) VFD, 396
tensor estimation, 58 white matter organisation, 205
WLLS (see Weighted linear least squares (WLLS)) Tractometry, 206
Tensor glyphs, 53–55 Tracts constrained by underlying anatomy (TRACULA),
Tensor-derived measures, 206 220, 222
Termination criterion, 232 Traumatic brain injury (TBI), 376–379
Therapeutic misconception, 280 anticoagulants and platelet aggregation inhibitors, 373
Three-dimensional tract representation challenges (see Challenges in TBI)
anatomy, 233, 260–270 CT scanning, 373
ROI, 233 diagnosis and characterization (see TBI classification)
Through-plane motion, 131 DLPFC, 375
Total slice dropout, 144 primary and secondary brain damage, 374
Total variation (TV) approach, 142 rapid detection, 374
Tracking, 231 role, DTI (see DTI in TBI)
Tracking algorithm, 23 Tumefactive demyelinating lesions (TDL), 312, 313
Track-weighted imaging, 223 Tumor grade
Tract density imaging, 223 ADC, 320
Tract selection angiogenesis, 320
logic combination, 212, 215 DKI, 321
440 Index

Tumor grade (cont.) image registration techniques, 186


GBM, 320 limitations, 165
gliomas, 320 pipeline, 164, 184
HGGs and LGGs, 320 reference image, 186
Two-dimensional tract representation scalar anatomical MRI information, 191
axial slices, 233–242 smoothing, 184 (see also Smoothing)
coronal slices, 233, 243–251 statistical analysis, 184
MITK-Diffusion, 232 and TBSS, 200
sagittal slices, 233, 252–259 tensor information, 191
tensor reorientation, 190
Voxel-based method, 338
U Voxels, 231
Unbiased tractography, 210
Uncinate fasciculus (UF), 210, 212, 262–265, 301, 303
W
Wallerian degeneration, 346
V WB. See Whole brain (WB) tractography
Ventral intermediate nucleus (VIM), 296 Weighted linear least squares (WLLS), 413
VFD. See Visual field deficit (VFD) ADC values, 60
VIM. See Ventral intermediate nucleus (VIM) magnitudes, original data, 59
Visual field deficit (VFD), 396 weights determination, 59
Voxel-based analysis (VBA), 170, 195–197 Westin measures, 68, 70
advantages, 165 White matter anatomy
brain, 163, 187 2D tract representation, 233
Clinical Practice, 201 3D tract representation, 233, 260–269
DTI DTI, 231, 232
data, 189 protocol, White matter atlas, 221, 232
metrics, 183 White matter hyperintensities (WMH), 277
parameter image, 165 Whole-brain tractography, 212–214, 379
FA, 164 Whole-brain analysis methods, 167
and MD Maps, 192 WLLS. See Weighted linear least squares
hybrid analysis methods, 165 (WLLS)
image processing technique, 184 WMH. See White matter hyperintensities (WMH)

Das könnte Ihnen auch gefallen