Sie sind auf Seite 1von 19

Ultrasound Obstet Gynecol 2013; 42: 1533

Published online in Wiley Online Library (wileyonlinelibrary.com). DOI: 10.1002/uog.12513

Non-invasive prenatal testing for aneuploidy: current status


and future prospects
P. BENN*, H. CUCKLE and E. PERGAMENT
*Department of Genetics and Developmental Biology, University of Connecticut Health Center, Farmington, CT, USA; Department of
Obstetrics and Gynecology, Columbia University Medical Center, New York, NY, USA; Northwestern Reproductive Genetics, Chicago,
IL, USA

K E Y W O R D S: amniocentesis; aneuploidy; chorionic villus sampling; Down syndrome; fetal DNA; maternal plasma; screening;
sequencing; trisomy

ABSTRACT 1980s, advanced maternal age, defined in most localities


as over 35 years, was the only means by which the
Non-invasive prenatal testing (NIPT) for aneuploidy
general population was assessed as to risk of a fetal
using cell-free DNA in maternal plasma is revolutionizing
chromosomal abnormality. Fewer than one third of
prenatal screening and diagnosis. We review NIPT in the
Down syndrome pregnancies were diagnosed prenatally
context of established screening and invasive technologies,
and of those undergoing invasive prenatal diagnosis only
the range of cytogenetic abnormalities detectable, cost,
about 2% had fetal karyotype abnormalities1 , a figure
counseling and ethical issues. Current NIPT approaches
comparable to the 0.51% chance of procedure-related
involve whole-genome sequencing, targeted sequencing
fetal loss associated with amniocentesis or chorionic villus
and assessment of single nucleotide polymorphism (SNP)
sampling (CVS)2 . In the late 1980s and early 1990s, the
differences between mother and fetus. Clinical trials
introduction of second-trimester maternal serum markers,
have demonstrated the efficacy of NIPT for Down and
in the form of double, triple and quad marker testing,
Edwards syndromes, and possibly Patau syndrome, in
improved significantly the screening performance for ane-
high-risk women. Universal NIPT is not cost-effective,
uploidy. The proportion of Down syndrome pregnancies
but using NIPT contingently in women found at moderate
diagnosed more than doubled and a chromosomal abnor-
or high risk by conventional screening is cost-effective.
mality was found in as many as 4% of those designated
Positive NIPT results must be confirmed using invasive
as screen-positive3 . In the late 1990s and early 2000s,
techniques. Established screening, fetal ultrasound and
aneuploidy screening shifted to the first trimester with
invasive procedures with microarray testing allow the
the combined test, which uses ultrasound measurement
detection of a broad range of additional abnormalities
of nuchal translucency thickness (NT) together with
not yet detectable by NIPT. NIPT approaches that
maternal serum concentration of placental proteins
take advantage of SNP information potentially allow the
human chorionic gonadotropin (hCG) (free , intact
identification of parent of origin for imbalances, triploidy,
or total) and pregnancy-associated plasma protein-A
uniparental disomy and consanguinity, and separate
(PAPP-A). Currently available screening protocols also
evaluation of dizygotic twins. Fetal fraction enrichment,
incorporate additional ultrasound markers and sequential
improved sequencing and selected analysis of the most
screening using two blood samples, one in the first
informative sequences should result in tests for additional
and one in the second trimester, with or without NT.
chromosomal abnormalities. Providing adequate prenatal
Consequently, screening performance has improved such
counseling poses a substantial challenge given the broad
that more than nine-tenths of Down syndrome cases can
range of prenatal testing options now available. Copyright
be diagnosed prenatally4 and the yield from invasive
2013 ISUOG. Published by John Wiley & Sons Ltd.
testing has risen to about 6%5 .
Recently, analysis of cell-free (cf)DNA in maternal
INTRODUCTION blood for non-invasive prenatal testing (NIPT) has been
shown to be highly accurate in the detection of common
The past quarter century has been witness to a series of fetal autosomal trisomies. In 1997, Lo et al.6 first reported
remarkable advances in the screening of pregnancies for their seminal discovery that plasma from pregnant women
aneuploidy, particularly in the identification of Down contained cfDNA, including a fraction designated fetal,
syndrome (trisomy 21). During the 1970s and early although this is thought to be placental in origin, resulting

Correspondence to: Dr P. Benn, Department of Genetics and Developmental Biology, University of Connecticut Health Center, Farmington,
CT, USA (e-mail: benn@nso1.uchc.edu)
Accepted: 14 May 2013

Copyright 2013 ISUOG. Published by John Wiley & Sons Ltd. REVIEW
16 Benn et al.

from apoptotic trophoblasts. This and other early studies7 Among those undergoing invasive prenatal diagnosis
suggested that the fetal fraction was only 36%, but because of a high risk of Down and Edwards syndromes,
more recent studies have found that it may be closer a large proportion have a different chromosomal abnor-
to 1020%8 . Fetal cfDNA can be detected as early as mality. Alamillo et al.19 reported on 10 years experience
4 weeks gestation9 and exceeds 4% of all the cfDNA in of a combined test, giving risks of Down syndrome
nearly all women from 10 weeks onward. The typical and Edwards or Patau syndromes. The 97 cases with
size of the cfDNA fragments is approximately 150 chromosomal abnormalities included Down syndrome
basepairs10,11 and, importantly, the entire fetal genome (48%), Edwards syndrome (16%), Patau syndrome (6%),
is represented. It has been shown that the half-life of Turner syndrome (9%), other sex chromosomal aneu-
fetal cfDNA is very short12 ; fetal fragments are no longer ploidies (4%), other aneuploidies including mosaics (9%)
detectable very soon after birth13,14 . There is therefore and chromosomal rearrangements (6%). The California
no serious concern that a prenatal cfDNA test could be state-wide screening program20 used a second-trimester
confounded by a prior pregnancy in multigravid women. quad test for Down and Edwards syndromes and detected
Maternal plasma cfRNA, unlike RNA extracted directly a total of 1316 chromosomal abnormalities. Excluding
from cells, appears to be relatively stable and can also balanced translocations, these were Down syndrome
potentially be used in screening15 17 . (48%), Edwards syndrome (14%), Patau syndrome
The commercial introduction of NIPT raises several (2%), Turner syndrome (8%), triploidy (2%), Klinefelter
concerns. One major issue is the optimal integration of syndrome (47,XXY) (1%) and other abnormalities
NIPT into current screening practice18 . Should NIPT and (25%). Some of these were chance findings but many
conventional screening tests be treated as related but were related to maternal age or abnormal marker profiles.
independent measures when assessing aneuploidy risk or Conventional aneuploidy screening modalities also
should they be complementary to one another? How can need to be considered in the context of the substantial
healthcare providers be educated and prospective patients number of non-chromosomal fetal abnormalities and
counseled about all the prenatal testing options now pregnancy complications that may be identified as a result
available? of this testing. Particularly important is the role of first-
In this Review we provide the information needed by trimester ultrasound, which has the potential to identify
clinicians and public health providers when considering a non-chromosomal abnormality in approximately 1%
to whom NIPT will be offered and regarding the of cases5 .
consequences of doing so, practical suggestions on how to
implement this new and powerful technology into routine
clinical practice, and some indications of how we expect ESTABLISHED INVASIVE PRENATAL
this testing to expand. We focus primarily on the detection DIAGNOSIS
of fetal chromosomal abnormalities, recognizing that the Conventional cytogenetics
technology will eventually be used for testing for a broad
range of other genetic disorders. Chromosomal analysis of cultured chorionic villi and
amniotic fluid cells can identify fetal aneuploidy,
structural rearrangements, such as translocations and
ESTABLISHED SCREENING MODALITIES
inversions, and relatively large duplications and deletions
In the current context it is important to distinguish pre- (generally exceeding 5 Mb in size)21 . Chromosomal
natal diagnosis of aneuploidy from antenatal screening. A analysis of amniotic fluid cells is considered to be the
diagnostic test performed on chorionic villi, amniotic fluid gold standard in prenatal testing because error rates are
or fetal blood needs to have very few false negatives (aneu- exceedingly low, probably less than 0.010.02%, and
ploid pregnancies misdiagnosed as euploid) and false pos- mostly appear to be due to maternal cell contamination,
itives (euploid pregnancies misdiagnosed as aneuploid), laboratory error or typographic mistakes22 . Error rates
since the result will inform the decision as to whether to with CVS are higher than those seen with amniocentesis,
terminate the pregnancy. In contrast, antenatal screening and these can be due to confined placental mosaicism
does not aim to be definitive; rather, it is designed to iden- (CPM), maternal cell contamination or lower resolution
tify women who are at sufficiently high risk of common banding of chromosomes23 . In a small proportion of
aneuploidies as to warrant invasive prenatal diagnosis. cases in which CVS karyotyping is carried out there
Since the invasive diagnostic procedures, mainly CVS and are ambiguous results which may require resampling or
amniocentesis, are hazardous and expensive, only a rela- amniocentesis for clarification.
tively small group of women are identified as being at high
risk. Established screening programs determine the risk of Molecular cytogenetics
Down syndrome, many also including Edwards syndrome
(trisomy 18) and some including Patau syndrome (trisomy Because of the time taken to obtain a complete chromo-
13) and Turner syndrome (45,X). The model-predicted somal analysis, the occasional finding of a karyotype of
aneuploidy detection rates (DR) for fixed false-positive minimal or uncertain significance and cost considerations,
rates (FPR) of established prenatal screening protocols some centers perform a rapid aneuploidy test (RAT)
has been reviewed elsewhere4 . using molecular genetic techniques as an adjunct to or

Copyright 2013 ISUOG. Published by John Wiley & Sons Ltd. Ultrasound Obstet Gynecol 2013; 42: 1533.
NIPT for aneuploidy 17

replacement for karyotyping. Methods include the use a particular case of fetal loss following the procedure it
of fluorescence in-situ hybridization (FISH)24,25 , quanti- is possible only rarely to attribute directly the adverse
tative fluorescence polymerase chain reaction (QF-PCR) outcome to the procedure; cases of amnionitis or chronic
testing26 and multiplex ligation-dependent amplification amniotic fluid leakage would be attributable but these
(MLPA)27 . These tests are generally designed to test only a are relatively rare consequences. In a randomized trial of
restricted range of cytogenetic abnormalities28 . Although amniocentesis at a single center31 , the fetal loss rate was
proven to be very accurate, confirmatory testing using 0.8% higher in the amniocentesis group compared with
conventional cytogenetics has been recommended29 . controls. There have been no similar randomized trials
In contrast, relative to conventional karyotyping, other for CVS, but a number of studies have compared late
molecular approaches can now improve substantially on first-trimester CVS with second-trimester amniocentesis.
the detection of clinically significant genetic imbalances. A Cochrane Review32 showed that, when performed by a
In high-risk pregnancies with a normal chromosomal skilled operator, the fetal loss rates of the two procedures
constitution, the increased resolution of array compara- were comparable. An updated review2 concluded that the
tive genome hybridization (aCGH) coupled with single excess miscarriage rate for either procedure is between
nucleotide polymorphism (SNP) array has the potential to 0.5% and 1%. An initial concern that CVS may cause
identify submicroscopic copy number variations (CNVs), fetal limb reduction defects has not been sustained, as
specifically microdeletions and microduplications, as well the reported excesses occurred when CVS was performed
as regions of homozygosity. early in gestation, before 10 weeks32 .
When compared to conventional chromosomal analy-
sis, aCGH and SNP array are purported to have significant
NIPT METHODS
advantages in terms of: (1) faster turnaround time;
(2) higher throughput while being less labor-intensive; The fact that fetal cfDNA and cfRNA are present only
(3) higher resolution independent of the ability of the as minority components of the total cell-free nucleic acids
cells to grow and/or generate good metaphase spreads; in maternal plasma specimens has posed a significant
(4) amenability to automation and quality control; and, technical obstacle in the development of NIPT. A
(5) direct mapping of aberrations to the genome sequence. number of approaches have been suggested that allow
Faster turnaround time is possible because arrays can enrichment of the fetal component. These include taking
be performed on DNA obtained from uncultured villi advantage of size differences between fetal and maternal
and amniotic fluid cells, thus avoiding the need for tis- DNA fragments11 , using formaldehyde in the samples33 ,
sue culture and the associated time delay required for taking advantage of the nucleosome binding property
conventional chromosomal analysis. of the DNA34 and immunoprecipitation of hypo- or
A National Institute of Health (NICHD) multicenter, hypermethylated DNA35 , but none is currently being
prospective, blinded study30 evaluated the application used in clinical practice. Even without enrichment a
of microarrays in over 4000 high-risk pregnancies number of technical approaches have now been proven
undergoing prenatal invasive testing largely because of to be effective. Each approach relies on the extraordinary
advanced maternal age, a positive first-trimester screen advances in molecular biology and sequencing achieved
or the presence of an ultrasound anomaly. The use of in recent years. We briefly review the major methods
microarrays resulted in the identification of clinically below, without discussing in detail all of the possible
relevant CNVs that were unrecognizable by conventional refinements and other developments that are currently
karyotyping in 2.5% of all cases. In those cases referred under consideration.
for testing because of an ultrasound abnormality and
showing a normal karyotype, 6% were found to have
Shotgun massively parallel sequencing (s-MPS)
a clinically relevant CNV. These studies were performed
on low-resolution aCGH platforms; newer arrays have This approach relies on the identification and counting
much higher resolution. The use of SNP arrays has of large numbers of the DNA fragments in the plasma
expanded the ability to detect triploidy and significant specimen. Using massively parallel sequencing (MPS),
regions of homozygosity, thereby detecting uniparental millions of both fetal and maternal DNA fragments can
disomy and potentially identifying disorders associated be sequenced simultaneously and, since the entire human
with consanguinity. The NIH multicenter study also genome sequence is known, each piece that maps to a
revealed the limitations of all aCGH and SNP array discrete locus can be assigned to the chromosome from
platforms, the principal limitation being the identification which it came36,37 . If fetal aneuploidy is present, there
of variants of uncertain significance (VOUS). should be a relative excess or deficit for the chromosome
in question. The difference in counts will be small; for
Hazards of invasive testing example, if fetal trisomy 21 is present and the fetal
fraction is 20%, the relative excess in chromosome
The principal hazard of amniocentesis is miscarriage, but 21 DNA fragments will be (0.8 2) + (0.2 3) = 2.2
the excess risk associated with the procedure is difficult compared with the situation for a euploid fetus
to quantify precisely. Some 34% of mid-trimester (0.8 2) + (0.2 2) = 2, i.e. a relative increase in number
pregnancies will miscarry without amniocentesis and in of chromosome 21 counts of only 10%. To be able to

Copyright 2013 ISUOG. Published by John Wiley & Sons Ltd. Ultrasound Obstet Gynecol 2013; 42: 1533.
18 Benn et al.

detect reliably these differences, large numbers of counts In theory, results could also be combined with other fac-
are necessary, the fetal fraction needs to be appreciable tors, for example results of other screening tests or prior
and the counts need to be compared with the expected history of an aneuploid pregnancy.
counts for euploid cases. The latter can be achieved either
by normalizing counts for the chromosome of interest
Single nucleotide polymorphism (SNP)-based
against other chromosomes that are expected to be
approaches
disomic within the same test run36 or by comparing the
fraction of counts assigned to a particular chromosome The feasibility of a non-invasive aneuploidy test that takes
against the fractions seen for a set of known euploid advantage of DNA polymorphisms was first demonstrated
cases38 . Results can be expressed as a z-score which by Dhallan et al.46 . In their approach, the paternal,
can be converted into a probability that a specific maternal and fetal SNPs present in blood (paternal),
chromosome result departs from the normal diploid buffy coat (maternal) and maternal plasma (maternal
situation. Although it is possible to convert the z-score and fetal) were evaluated as bands on sequencing gels.
to a patient-specific risk, major commercial providers Quantification of band intensities of the uniquely inherited
of this testing currently choose to present their results paternal SNPs in the plasma allowed an estimation of
only in terms of positive or negative based on z-scores the fetal fraction. Comparison of the maternal plus
exceeding predefined thresholds. Another variation of the fetal bands with the unique fetal band intensity also
statistical analysis calculates two Student t-test statistics, allowed estimation of the fetal chromosome 21 dosage.
one based on the hypothesis that the result is from a A similar approach was used by Ghanta et al.47 , who
euploid fetus and the other that it is from an aneuploid identified combinations of paired SNPs to focus on highly
fetus and then calculates a further test statistic, L, informative regions and then used cycling temperature
which is the logarithmic likelihood ratio39 . Refinements capillary electrophoresis for evaluating fetal fraction and
in the statistical analysis also include adjustment for dosage. The method was thought to be useful in situations
guanine-cytosine (GC) base content of the sequences40,41 . in which the fetal fraction was as low as 2%.
The approach is referred to as shotgun because it relies A more complex approach that relies on polymor-
on sequencing and counting all informative chromosome phisms has been proposed by Zimmermann et al.48 . The
regions. Sequences which do not map to a unique locus approach involves a multiplex amplification of 11 000
are uninformative. In some studies, only those fragments SNP sequences in a single PCR reaction carried out on
that have sequences identical to the reference genome the plasma DNA followed by sequencing. Each product is
sequence are accepted in the analysis, while in others evaluated based on the hypothesis that the fetus is mono-
one or two mismatches in the sequence will be accepted. somic, disomic or trisomic. After considering the positions
Because of costs, it is common to run multiple samples of the SNPs on the chromosomes and the possibility
simultaneously (multiplexing) by adding a bar-code that there may have been recombination, a maximum
sequence to each fragment that will allow identification likelihood is calculated that the fetus is either normal,
of the sample origin. The number of cases run together aneuploid (chromosome 21, 18, 13 or sex chromosome)
can be variable and is an important consideration because or triploid, or that uniparental disomy is present.
high-level multiplexing places a limitation on the total In theory, with SNPs there should be advantages over
number of sequence reads that are carried out on each methods based on total sequence counting. SNPs can
case (i.e. the depth of sequencing). The number of provide information about parent of origin of aneuploidy,
fetal sequence reads will also vary according to the recombination and inheritance of mutations. On the
fetal fraction and therefore a minimum fetal fraction, other hand, SNPs account for only 1.6% of the human
typically 4%, is currently required. Most studies have genome and therefore enrichment for fetal DNA, deeper
used Illumina sequence analyzers but there are also studies sequencing or higher levels of high-fidelity amplification
using Solid42,43 and Helicos44 platforms. may be required to identify unambiguously affected
pregnancies with small imbalances49 . When few loci are
Targeted massively parallel sequencing (t-MPS) used in the analysis, greater attention needs to be paid to
the exclusion of regions in which there are rare benign
It is also possible to amplify selectively only those chro- CNVs. The SNP approach could result in the inadvertent
mosomal regions that are of interest (e.g. chromosomes detection of non-paternity or perhaps consanguinity. In
21, 18 and 13) and then to assess whether there is a the case of assisted reproduction with an egg from an
departure from euploidy based on the relative number of unrelated donor, SNP-based approaches would need to
DNA fragment counts for this subset of chromosomes45 . be modified to take into consideration the presence of
This targeted approach has the advantage of requiring additional maternally derived fetal alleles not present in
considerably less sequencing and thereby reduces costs. the surrogate mother.
The major commercial provider of this approach includes The laboratory methods and data analyses used for
an adjustment to allow for the variability in the propor- SNP-based NIPT are substantially different from those
tion of DNA that is fetal and then combines the results of used in shotgun and targeted counting approaches. In this
the laboratory test with maternal age to provide a patient- Review we therefore consider the emerging clinical trial
specific risk for Down, Edwards and Patau syndromes45 . data for SNP-based testing separately.

Copyright 2013 ISUOG. Published by John Wiley & Sons Ltd. Ultrasound Obstet Gynecol 2013; 42: 1533.
NIPT for aneuploidy 19

Methylated DNA-based approaches could be deduced from departures from a normal 1:1
ratio in the relative amount of the inherited alleles60 . This
This approach takes advantage of the fact that there approach to NIPT showed some success in the initial
will be epigenetic differences throughout the genome proof-of-principle studies, but unpublished clinical trial
which will result in the differential expression of genes, data were disappointing. Nevertheless, with the discovery
depending on the tissue type50 . These differences can be of additional RNAs that can be analyzed, this approach
associated with either hyper- or hypomethylation of the may yet play an important role in NIPT61 .
genes. Some genes present in placental cells (specifically,
trophoblasts) will therefore differ from maternal cells
in the methylation pattern. Methylated DNA can be NIPT IN CLINICAL TRIALS:
chemically modified, thus allowing its characterization NON-MOSAIC DOWN, EDWARDS AND
separate from the unmethylated DNA51 . Alternatively, PATAU SYNDROMES
the hypermethylated DNA can be analyzed separately
Proof-of-principle studies demonstrated the feasibility of
following restriction enzyme digest of the hypomethylated
NIPT using small series of maternal plasma samples
DNA52 . If, additionally, there are polymorphic differences
from aneuploid and euploid pregnancies36 38,40 46,62 64 .
in the fetal alleles, disturbance from a 1:1 ratio can be
However, generally, the samples were not tested blind
used to identify trisomy. This has been demonstrated for
to the outcome of pregnancy, ascertainment criteria were
detection of Edwards syndrome but the approach has not
not fully described and some samples were obtained after
been tested in clinical trials51 . Differentially methylated
an invasive diagnostic procedure which could have led
regions may be relatively common53 so the method may
to increased transfer of fetal DNA into the maternal
also be applied to detect other aneuploidies.
circulation. There have now also been several large clinical
It is also possible to use immunoprecipitation with
trials of MPS which have overcome these limitations and
antibodies specific to 5-methylcytidine to enrich for the
potential biases39,65 75 . These have established NIPT as a
methylated DNA. For example, the identification of a set
potentially highly effective screening test but not as a test
of specific DNA sequences on chromosome 21 that are
that could replace current invasive prenatal diagnosis. We
hypermethylated in placental cells allows the selective
therefore prefer the term NIPT or cfDNA screening rather
enrichment of fetal cfDNA from maternal plasma35 .
than non-invasive prenatal diagnosis, which is misleading.
Following quantitative real-time PCR, there may be
measurable differences in the amount of chromosome 21-
derived DNA when fetal trisomy 21 is present, compared High-risk studies
with in euploid fetuses. One proof-of-principle study has
There have been seven cfDNA studies involving
demonstrated the efficacy of such an approach to NIPT54 .
shotgun or targeted sequencing in women who had
A follow-up study using a modified set of differentially
invasive prenatal diagnosis because of high risk for:
methylated chromosome 21 DNA sequences also showed
Down syndrome65,67 , Down syndrome or another
encouraging results55 . This approach is currently the
aneuploidy66,68,69 or any disorder70,71 . In one study, a
subject of further clinical trials. If an approach such
screen-positive screening test was the only indication
as this can be fully developed, it could be substantially
for high risk69 , whilst in the four other studies in which
cheaper than sequencing-based methods.
the reason for invasive testing was high risk for Down
syndrome or another aneuploidy, the indications were
Digital PCR mixed. The reason for invasive testing was a screen-
positive screening test in 77%65 , 43%67 , 30%66 and
Another way in which the costs of sequencing can be 22%68 of cases; advanced maternal age in 68%66 , 37%
avoided is through the use of digital PCR. This technology (only indication in these cases)68 and 33%67 of cases;
is based on the dilution of test materials such that ultrasound in 13%66 , 11%67 and 7%68 of cases; family
individual DNA fragments of interest (e.g. chromosome history in 5%66 , 3%67 and 3%68 of cases; and multiple
21) can be amplified separately and counted56 58 . reasons in 22%68 and 9%67 of cases. Five studies included
Theoretical estimates of the number of amplification results on both Down and Edwards syndromes68 72 and
events have been computed59 and this approach could three also included Patau syndrome68,72,75 .
become particularly attractive if reliable fetal cfDNA The study design details for these trials on high-risk
enrichment methods were developed. women are summarized in Table 1. One study designated
as unclassified results with z-scores of 2.54.0 for
RNA-based testing autosomal chromosomes and excluded them from the
DR and FPR calculations68 . Doing so is not valid76 ,
Identification of genes that are expressed by trophoblasts particularly when no protocol was suggested for the
but not maternal cells should result in the presence of unclassified group, and in this Review we have reclassified
the corresponding cfRNA species in maternal plasma, them as negative. However, the unclassified group had a
free of contaminating maternal RNA with a similar five-fold increased likelihood of aneuploidy (3.5% (5/144)
sequence. If the cfRNA also carries polymorphisms that of trisomies unclassified compared with 0.7% (9/1358) of
distinguish the fetal alleles, the presence of aneuploidy controls) and it could be argued that they should in fact

Copyright 2013 ISUOG. Published by John Wiley & Sons Ltd. Ultrasound Obstet Gynecol 2013; 42: 1533.
20 Benn et al.

Table 1 Eleven cell-free DNA studies of shotgun or targeted sequencing: methodological differences

Trial Design Average GA (weeks) Method Algorithm Cut-off*

High risk
Chiu et al.65 Prospective and casecontrol 13 Shotgun z-score 3.0
Ehrich et al.66 Casecontrol 16 Shotgun z-score 2.5
Palomaki et al.67,72 Casecontrol 15 Shotgun z-score 3.0
Bianchi et al.68 Casecontrol 15 Shotgun z-score 4.0
Sparks et al.70 Casecontrol 18 Targeted Risk 1%
Ashoor et al.69,75 Casecontrol 13 Targeted Risk 1%
Norton et al.71 Prospective 16 Targeted Risk 1%
Other than high risk
Lau et al.73 Prospective 14 Shotgun z-score? ?
Nicolaides et al.74 Retrospective 12 Targeted Risk 1%
Dan et al.39 Prospective 20 Shotgun z-score & L-score 2.5 or 1.0
Gil et al.78 Prospective 10 Targeted Risk 1%

*For autosomal chromosomes. Includes maternal and gestational age. Results with scores 2.54.0 were unclassified. Positive if z < 2.5
but L > 1. GA, gestational age at testing.

be reclassified as positive, so those rates are also given in Down syndrome but depending on her pretest risk it
this Review. may not be extremely high. This can be illustrated from
Table 2 summarizes the Down and Edwards syndromes the combined data in Table 3, by estimating a positive
results for these seven cfDNA studies carried out in high- likelihood ratio (LR) using the DR divided by the FPR,
risk pregnancies. In total these studies compared 594 i.e. 634. For example, a woman with a pre-test risk of
samples from pregnancies with Down syndrome with 1 in 270 following a combined test would have a post-
5745 that did not, and 193 samples from pregnancies test odds of 634:269 or a risk of 2 in 3. This is not
with Edwards syndrome with 5459 that did not, although sufficiently high as to warrant termination of pregnancy
in both cases some had other aneuploidies. The results without a confirmatory invasive diagnostic test. Similarly,
from the various studies, notwithstanding methodological the negative LR, estimated from the false-negative rate
differences, are consistent with each other and their divided by the true negative rate, is 1/148. A woman
combined data represent the best estimates of DR with a pre-NIPT risk of 1 in 10 would have a 1 in 1333
and FPR. For Down syndrome, the overall DR of risk after a negative test, which might be insufficiently
99.3% has a 95% CI of 98.299.8% and FPR of reassuring for some women. These LRs are illustrative
0.16% (95% CI, 0.080.31%). Even with the most and cannot be used to counsel individual women because
extreme CI values this performance exceeds, by far, some of the studies had already incorporated prior risk
anything achievable by current Down syndrome screening based on maternal age into their algorithms. However,
protocols. However, this performance falls short of that it is reasonable to conclude that using NIPT following a
for current diagnostic tests. Furthermore, reclassifying as screen-positive test for any of the established screening
positive the unclassified results in the study of Bianchi protocols followed by confirmatory invasive diagnostic
et al.68 would have increased the overall DR slightly to testing if the NIPT result is positive will probably hardly
99.5%, but almost doubled the FPR to 0.26%. Hence, affect the overall screening DR but will reduce the FPR
we can conclude that based on present evidence cfDNA about 300-fold.
testing is not a diagnostic test of Down syndrome but For Edwards syndrome, the overall DR is 97.4%
rather is a very good secondary screening test. A woman (95% CI, 93.799.0%) and the FPR 0.15% (95% CI,
with a positive cfDNA test will be at high risk of 0.070.31%), with positive LR of 665 and negative

Table 2 Down syndrome and Edwards syndrome results in seven cell-free DNA studies carried out in high-risk pregnancies

Down syndrome Edwards syndrome


Trial DR (n (%)) FPR (n (%)) DR (n (%)) FPR (n (%))

Chiu et al.65 * 86/86 (100) 3/146 (2.1)


Ehrich et al.66 39/39 (100) 1/410 (0.24)
Palomaki et al.67,72 209/212 (98.6) 3/1471 (0.20) 59/59 (100) 5/1688 (0.30)
Bianchi et al.68 89/90 (98.9) 0/410 (0.00) 35/38 (92.1) 0/463 (0.00)
Sparks et al.70 36/36 (100) 1/123 (0.81) 8/8 (100) 1/123 (0.81)
Ashoor et al.69 50/50 (100) 0/297 (0.00) 49/50 (98.0) 0/297 (0.00)
Norton et al.71 81/81 (100) 1/2888 (0.03) 37/38 (97.4) 2/2888 (0.06)
Total 590/594 (99.3) 9/5745 (0.16) 188/193 (97.4) 8/5459 (0.15)

*2-plex and 8-plex data were reported but only the more favorable 2-plex results are included here. DR, detection rate; FPR, false-positive
rate.

Copyright 2013 ISUOG. Published by John Wiley & Sons Ltd. Ultrasound Obstet Gynecol 2013; 42: 1533.
NIPT for aneuploidy 21

Table 3 Failure rates in 11 studies of cell-free DNA testing*

Trial Failure rate (n (%)) Reasons for failure

Chiu et al.65 11/764 (1.4) Low total DNA (n = 2), low DNA library concentration (n = 8), low matched DNA sequence
reads (n = 1)
Ehrich et al.66 18/467 (3.8) Low % fetal DNA (< 4%) (n = 7), low total DNA (< 556 copies) (n = 7), low DNA library
concentration (< 32.3 nM) (n = 15), low number unique DNA sequence counts (< 3
million) (n =11); some failed more than one criteria
Palomaki et al.67 13/1696 (0.8) Low % fetal DNA (< 4%) (n = 6), other QC parameters (n = 7): low DNA library
concentration (< 25 nM) and low matched DNA sequence reads (< 12.5 million)
Bianchi et al.68 16/532 (3.0) No fetal DNA
Sparks et al.70 8/338 (2.4) Low % fetal DNA (< 3%), low DNA sequence counts, evidence from SNPs of non-singleton
pregnancy
Ashoor et al.69 3/400 (0.8) Amplification and sequencing
Norton et al.71 148/3228 (4.6) Low % fetal DNA (< 4%) (n = 57), assay failure (n = 91): inability to measure % fetal, high
variation in DNA counts and failed sequencing
Lau et al.73 0/567 (0.0)
Nicolaides et al.74 100/2049 (4.9) Low % fetal DNA (< 4%) (n = 46), assay failure (n = 54)
Dan et al.39 79/11 184 (0.7) Quality of separation, extraction, sample preparation and sequencing: low peak DNA library
size, low library concentration (< 30 nM) and low matched DNA sequence reads (< 2
million)
Gil et al.78 40/997 (4.0) Low % fetal DNA (< 4%) (n = 23), assay failure (n = 17)

*Excluding tests rejected because of inadequate sample quality.

LR 1/38. Including the unclassified results in the study at 1213 weeks gestation. These women comprised
of Bianchi et al.68 as positive would have increased 179 (32%) who were screen-positive by conventional
the DR to 98.4% and the FPR to 0.20%. Since a testing, 194 (34%) who were screen-negative or awaiting
cfDNA test is unlikely to be aimed at detecting Edwards results and 194 (34%) who were unscreened. There was
syndrome alone, these false-positives should be regarded insufficient follow-up to report DRs reliably, but eight
as additional to those generated by cfDNA testing for cases of Down syndrome and one of Edwards syndrome
Down syndrome. There are no data with which to assess were detected and there were no false positives. Seven of
whether there is any tendency for those testing falsely the detected cases were among the screen-positive women
positive for one type of aneuploidy to be falsely positive (1 in 25) and two, both Down syndrome, were among
for another. Assuming these are uncorrelated, the FPR for the remainder (1 in 194).
both aneuploidies together is 0.30%, or 0.46% if those Nicolaides et al.74 carried out a retrospective study
unclassified by Bianchi et al.68 are included. using stored plasma samples from women who had a
Combining the results from the three studies which combined test at 1113 weeks gestation. After excluding
included Patau syndrome68,72,75 gives a DR of 30/38 non-viable pregnancies, those terminated for reasons
(78.9%; 95% CI, 65.991.9%) and FPR of 17/4112 other than Down or Edwards syndromes and cases lost
(0.41%; 95% CI, 0.220.61%), with a positive LR of to follow-up, 1949 were successfully tested for cfDNA,
191 and a negative LR of 1/4.7. Including the unclassified including eight with Down syndrome and two with
results as positive, the DR increased to 80.0% with no Edwards syndrome. All aneuploidies were detected and,
increase in the FPR. On the basis of these results, an MPS while there were no false-positive Down syndrome cases
test for all three trisomies would have a FPR of 0.70.9%, among 1939 tested, there were two (0.05%) false-positive
depending on the inclusion of the unclassified results. Edwards syndrome cases among 1937 tested. Invasive pre-
Initial prospective clinical experience has been reported natal diagnosis had been carried out in 4.3% (86/2049) of
for a commercial laboratory, Verinata Inc, offering those selected, including all of the cases with aneuploidy.
tests to high-risk women77 . While follow-up information A large prospective study of cfDNA screening was
was incomplete, among 5974 samples there were false carried out in China39 . A total of 11 105 pregnancies
positives for Down, Edwards and Patau syndromes and were tested successfully, including 4522 (41%) that
false negatives for Down and Edwards syndromes, with were screen-positive following conventional screening and
rates compatible with those seen in the trial data. 2770 (25%) with risk factors such as advanced maternal
age, ultrasound abnormalities and family history. All
Non-high risk of the known Down syndrome (n = 140) and Edwards
syndrome (n = 42) cases were detected by the cfDNA
There have been four NIPT studies involving shotgun test. However, approximately one third of the 10 915
or targeted sequencing in which the subjects were not with negative test results were lost to follow-up; 2818
specifically selected because they had invasive prenatal (26%) had invasive testing, a further 70 (< 1%) were
diagnosis. Their methods are summarized in Table 1. terminated, and follow-up was available for 4524 (41%).
Lau et al.73 reported prospective cfDNA screening The protocol included an insurance scheme whereby the
results of 567 patients, 49% of whom were tested family would be compensated in the event of unpredicted

Copyright 2013 ISUOG. Published by John Wiley & Sons Ltd. Ultrasound Obstet Gynecol 2013; 42: 1533.
22 Benn et al.

delivery of either a Down or an Edwards syndrome correlation between fetal fraction and both PAPP-A and
baby. This is likely to be a considerable incentive to free -hCG, which might be related to placental volume,
report false-negative results so the lack of follow-up may although there was no association with gestational age in
not be a major problem. There were two proven false- either study. Since in Down syndrome pregnancies PAPP-
positive cfDNA results, one each for Down and Edwards A is reduced on average and free -hCG is increased, the
syndromes. However, a further eight positive cases did correlations with fetal fraction may cancel each other out.
not have a karyotype following fetal loss or termination In Edwards syndrome pregnancies, both serum analyte
and therefore the possibility of additional false positives markers are reduced, consistent with reduced placental
cannot be excluded. volume80 . There could therefore be a somewhat increased
Gil et al.78 carried out a prospective study of cfDNA chance for cfDNA test failure in affected pregnancies.
screening at 10 weeks gestation in 997 women with On the other hand, those cases with higher PAPP-A and
median age 37 years; combined tests were also carried -hCG which might not be detected by the combined test
out. A total of 15 cases of aneuploidy were detected by may show higher fetal fractions and these may therefore be
cfDNA and confirmed by invasive testing (10 cases of more amenable to detection by a cfDNA-based NIPT. In a
Down syndrome, four of Edwards syndrome and one further published series of those having invasive prenatal
of Patau syndrome) and there was one false positive diagnosis due to high risk generally71 , the mean fetal
(for trisomy 18). One case was positive for trisomy 21 fraction was computed for deciles of maternal age, NT
but miscarried before invasive confirmation and no false and aneuploidy risk81 . There was no significant difference
negatives had emerged at the time of writing. in means between the highest and lowest deciles. The
These studies provide some, albeit limited, evidence highest decile for risk comprised 100 women with risk
to suggest that cfDNA screening may be as effective greater than 1 in 10 and a mean fetal fraction of 11.4%
in the general population as it is in those already compared with 135 women in the lowest decile with risk
scheduled for invasive testing: the FPR is not dissimilar less than 1 in 6500 and a mean fetal fraction of 10.8%.
to that in the high-risk studies; all aneuploidies appear
to have been detected, although most of them were in
Failed results
pregnancies which were already candidates for invasive
testing. Moreover, among high-risk women, performance Several of the MPS studies discussed above specified the
does not appear to be related to the indication for invasive number of pregnancies not considered for cfDNA testing
testing. Palomaki et al.67 found that the DRs and FPRs because of sample quality. Overall, among 13 260 eligible
were very similar according to indication: screen-positive pregnancies there were 814 (6.1%) untested. This rate
99% (96/97) and 0.2% (1/637), respectively; advanced ranged from 0.8% to 9.9% between the studies and may
maternal age or family history 100% (24/24) and 0.3% reflect the stringent standards required when carrying
(2/587); ultrasound 100% (51/51) and 0% (0/130); out a research project. Of more concern is the 2.0% of
and multiple reasons 95% (37/39) and 0% (0/112). pregnancies (436/22 222) in which a cfDNA test was
Nevertheless, until much larger general population studies performed but failed because of insufficient fetal DNA
are published, the limited direct evidence of efficacy needs or other technical reasons (Table 3). The failure rate
to be supplemented by indirect evidence. The separation may be even higher in those resampled after an initial
in the distributions of z-scores or risks between aneuploid failure: it was 32% (13/40) in one study78 . Quality
and euploid pregnancies is necessarily dependent on the control criteria varied between the studies but in those
fetal fraction and it is therefore important to consider that included a minimum acceptable percentage of fetal
whether this might be lower in low-risk women. DNA, about half the failures were due to this not being
Several covariables for fetal fraction have been met. In the prospective laboratory experience reported
investigated, including indication for invasive prenatal by Futch et al.77 , a result was not possible in 2.4% of
diagnosis, maternal age, screening marker level and cases. This is relevant to comparisons with conventional
estimated aneuploidy risk. In one published series of screening, which rarely reports failure to obtain a result.
pregnancies at high risk of Down syndrome or other Three prospective studies of women not specifically
aneuploidy67 , there was no significant difference in mean selected because they had invasive prenatal diagnosis
z-score, in either Down syndrome or euploid pregnancies, provide information on the extent to which a successful
according to indication or correlated with maternal age, test result was achieved following an initial failure. In one
but there was a significant negative correlation with study73 , among 567 successful cfDNA tests, four (0.7%)
maternal weight. The latter effect can be explained by had required a repeat blood sample and no other tests had
there being a fixed mass of fetal DNA, from the placenta, failed during that period (T.K. Lau, pers. comm.). The
mixing with an increasing mass of maternal DNA; such time from the initial blood draw to the cfDNA report was
an effect is also seen in conventional maternal serum greater than 14 days for 16 (2.8%) tests, including those
markers. In another published series of pregnancies at requiring a repeat sample. In another study39 , among
high risk of Down syndrome69 , a multivariate regression 11 105 tests, 97 (0.9%) needed resampling. In this study
analysis was carried out which confirmed that maternal as a whole, the turnaround time was within 10 working
weight was a covariable but maternal age was not79 . In days for 95% of pregnancies and within 15 for 99%;
addition it showed that there was a significant positive those requiring resampling took a further 1015 days to

Copyright 2013 ISUOG. Published by John Wiley & Sons Ltd. Ultrasound Obstet Gynecol 2013; 42: 1533.
NIPT for aneuploidy 23

produce a report. In the third study82 , a request for a the fetoplacental remains of the non-viable fetus could
patient redraw was made in 1.9% of referrals and among interfere with the cfDNA result; there is a relatively high
those who did undergo a redraw there was sufficient fetal chance that the fetal loss would have been associated
cfDNA in the second sample in 56% of cases. The success with aneuploidy and this could lead to a false-positive
rate was strongly dependent on maternal weight. result77 .

SNP studies COST OF NIPT


Following the initial proof-of-concept study using poly- Currently, the cost of a cfDNA test is high, ranging from
morphisms and maximum LRs for test interpretation48 , about $800 to $2000 in the USA, and from $500 to
the results of one small clinical trial have been published83 $1500 elsewhere. Whilst this may not necessarily deter
and additional information has been presented at scien- individuals from being tested, it is likely to determine
tific meetings in the form of an abstract84 and a poster85 . whether public health planners provide the testing and
The study of Nicolaides et al.83 involved 242 singleton will be the basis for whether health insurance schemes
pregnancies with results available for 229 (95%). All 32 reimburse those tested. We have published a sensitivity
aneuploid and 210 euploid cases were identified correctly. analysis90 in which the factors contributing to these
The abstract84 provided information on 407 pregnan- overall costs are elucidated for four cfDNA screening
cies including 44 with aneuploidies and the results were policies: (1) cfDNA screening for those screen-positive on
100% correct among samples that passed quality control. combined test; (2) universal cfDNA screening, replacing
In addition to the maternal plasma, paternal blood or all current screening modalities; (3) contingent cfDNA
buccal mucosa samples were genotyped. Whilst this was screening, for the 1020% of women with the highest
not an absolute requirement (paternally inherited alleles risk based on a combined test; (4) cfDNA screening for
in the fetus can be deduced), the failure rate was lower women of advanced maternal age and younger women
when a paternal sample was available. In the poster85 , with a positive combined test90 .
there were 763 samples, of which 45 (5.9%) failed to pass Restricting cfDNA testing to screen-positive cases leads
quality control, and among the remaining 717 (including to a small reduction in detection, a massive reduction in
47 Down, 15 Edwards, 7 Patau and 12 Turner syndromes) false positives and is the least expensive way in which
samples all except one result, for Turner syndrome, was to utilize cfDNA testing. With this policy the average
correct. cost per Down syndrome birth avoided is dependent only
on the difference in unit cost of a cfDNA test and of
Multiple pregnancies invasive prenatal diagnosis. For a combined test with
FPR of 3%, the average cost would be reduced by using
There is a potential problem with using cfDNA to secondary cfDNA testing, if the unit cost was less than or
screen for aneuploidy in multiple pregnancies. As with equal to three-quarters that of invasive testing. Even at a
biochemical screening, in twins discordant for trisomy the higher unit cost of cfDNA, the average cost per fetal loss
excess in fragments from a specific chromosome in the prevented would be relatively low.
affected twin may be masked by the normal proportion For other policies in which the aim of cfDNA testing
of those fragments in the euploid cotwin. This would also is to increase detection, the most important public health
be the case for higher order multiple pregnancies. This consideration will be not the total or average cost but
problem might be overcome by using SNPs to measure the marginal cost compared with the existing screening
the variations in the fetal fraction between genomic provision. The marginal cost is the average cost per Down
regions86,87 . The method allows determination of zygosity syndrome birth avoided or loss prevented over and beyond
and, in dizygous twins, examination of the distribution of that which would have been avoided by the existing
fetal fragments for each fetus. service. This is shown in Table 4 for fixed unit costs for
At present, only two small multiple-pregnancy cfDNA the combined test and invasive testing and with variable
series have been published, using the same method as unit costs of cfDNA. The table also shows screening
for singletons88,89 . The first series was derived from one performance.
of the high-risk studies67 and included three discordant Universal cfDNA screening yields both a very high DR
twins (two with Down and one Patau syndrome), five and a small FPR but the marginal cost is very high,
concordant Down syndrome twins, 17 euploid twins and exceeding the lifetime cost to the system of a Down
two euploid triplets88 ; there were no false-positive or false- syndrome birth91 94 . With universal screening the cost
negative cfDNA test results. The second series was from per fetal loss prevented when replacing conventional
one of the studies of women not selected because they screening is also very high. Contingent protocols, in which
were at high risk73 and included only one discordant all women receive the combined test and the 1020% with
twin with Down syndrome and 11 euploid twins89 ; the highest risk receive cfDNA testing, are efficient and
all pregnancies were correctly classified by the cfDNA can reduce massively the marginal cost compared with
test. universal screening. The average cost of preventing a fetal
In a singleton pregnancy that had had a vanishing twin, loss is also several times lower than that for universal
it is theoretically possible that apoptosis of cells from cfDNA testing. A policy of cfDNA testing only for older

Copyright 2013 ISUOG. Published by John Wiley & Sons Ltd. Ultrasound Obstet Gynecol 2013; 42: 1533.
24 Benn et al.

Table 4 Four cell-free (cf)DNA policies*: performance, marginal cost per Down syndrome (DS) birth avoided and average cost per fetal loss
prevented, according to unit cost of cfDNA testing

Marginal cost per DS birth Average cost per fetal loss


avoided ($ million) for prevented ($ million) for
Policy Performance unit cost cfDNA of: unit cost cfDNA of:
Combined cfDNA DR (%) FPR (%) OAPR $1500 $1000 $500 $1500 $1000 $500

None All 99.3 0.16 1:1 5.84 3.63 1.42 9.47 5.89 2.31
All Contingent:
10% 90.4 0.016 7:1 1.11 0.65 0.19 0.82 0.48 0.14
15% 92.8 0.024 5:1 1.39 0.86 0.32 1.33 0.82 0.30
20% 94.4 0.032 4:1 1.67 1.05 0.44 1.85 1.16 0.48
< 35 years 35 years 85.0 2.1 1:19 2.90 1.73 0.57 2.35 1.41 0.46
< 35 years 35 years or positive 84.7 0.017 6:1 3.69 2.02 0.37 0.81 0.44 0.08
on combined test

*Three reported in a modeling analysis90 and the fourth, cfDNA restricted to older women, using the same model. Combined test,
false-positive rate (FPR) = 3%, unit cost = $150; invasive prenatal diagnosis, unit cost = $1000, iatrogenic fetal loss rate = 0.5%. DR,
detection rate; OAPR, odds of being affected given a positive result.

women was not substantially more effective than was FUTURE PROSPECTS: NIPT FOR
the combined test and is more costly than contingent ADDITIONAL CYTOGENETIC
screening. A hybrid policy of offering cfDNA testing to ABNORMALITIES
all those of advanced maternal age plus all combined test
screen-positive women was better as it reduced the FPR Other autosomal abnormalities
massively but it was less effective and more expensive With the widely anticipated rapid improvements in
than was a contingent testing policy. sequencing, it may soon be possible to screen for
The detection rate for a contingent policy can be other autosomal trisomies in situations in which the
increased by including additional serum and ultrasound fetal fraction is very low, thereby allowing detection of
markers. Nicolaides et al.95 suggest markers which are aneuploidy even earlier in pregnancy. Early pregnancy
also of value in screening for pre-eclampsia and fetal is associated with very high levels of chromosomal
cardiac abnormalities. Cost-benefit analyses for such abnormality and although nearly all abnormal cases
variants have not yet been carried out. will spontaneously abort, early identification could be
Two other published studies have estimated costs when beneficial to women. First-trimester autosomal trisomy
cfDNA is applied to women with positive conventional can involve any chromosome, but some occur much more
screening results, using the estimated NIPT DR and FPR frequently than others22 . Detection of these through NIPT
from their own single study. Palomaki et al.67 considered will be facilitated by the larger size of the additional
total costs following screening in 100 000 screen-positive chromosome in the non-viable trisomies. Early studies
women, with an average risk of 1 in 33 for a Down suggested that there was greater variability in the fragment
syndrome fetus. They concluded that if the unit cost of counts for some of the larger chromosomes36 , but this
cfDNA was less than 96% of the unit cost of invasive could be overcome by GC correction and selective use of
testing, there could be a net saving (4% of costs need the most consistent regions of chromosomes37,62 .
to be assigned to test failures and confirmatory invasive Screening for a large number of potential abnormalities
testing following cfDNA positive results). The second together does require very high specificity for each
study also considered 100 000 screen-positive women, component so that the aggregate FPR for all chromosomes
modeled as a mixture of combined test or second-trimester combined is low.
serum screening96 . They estimated total costs including
both the conventional screen and subsequent testing and
Sex-chromosome abnormality
concluded that with a unit cost of cfDNA $1200, CVS
$1700 and amniocentesis $1400, there would be a 1% Establishing NIPT for sex-chromosome imbalances is
cost reduction. One other study has estimated costs desirable because some specific sex-chromosomal abnor-
for cfDNA screening in older women and in younger malities are associated with a sufficiently severe phenotype
women with screen-positive conventional screening tests, as to fully justify prenatal diagnosis: for example, many
using DR and FPR estimates from the literature97 . The cases of Turner syndrome, most of which will not sur-
authors claimed that this policy, in the USA, would have vive to full term, gain of multiple X chromosomes and
lower total cost compared with conventional screening. some unbalanced structural rearrangements of the X-
However, this conclusion appears to have been reached chromosome. Collectively, unbalanced sex-chromosomal
following incorrect assumptions about the DR and FPR in abnormalities constitute over a third of the unbalanced
younger women and a very low uptake of invasive testing karyotypes seen in amniocenteses performed in women
for women screen-positive by conventional screening under 35 years of age and nearly a quarter of those in
compared with cfDNA screening90 . older women98 . There is mosaicism in approximately one

Copyright 2013 ISUOG. Published by John Wiley & Sons Ltd. Ultrasound Obstet Gynecol 2013; 42: 1533.
NIPT for aneuploidy 25

third of sex chromosomal aneuploidies detected through The age-related loss of the X-chromosome is thought
amniocentesis98 . to involve preferentially the inactive X-chromosome105 .
One commercial laboratory that provides NIPT, Some women may also show cells that have gained an
Sequenom Inc, was routinely providing information on X-chromosome106 . Clonal populations of maternal cells
the presence or absence of Y-chromosome DNA in their with X-chromosome gain or loss in hematological cells
reporting. Based on a series of 1639 samples, the DR (which are believed to be the source maternal cfDNA in
for a Y-chromosome was 828/833 (99.4%), and the plasma), maternal constitutional mosaicism or even a non-
accuracy for the prediction of a female fetus was 803/806 mosaic X-chromosomal abnormality such as 47,XXX are
(99.6%)99 . More recently, they have been providing also possible107 . The fetal X-chromosomal dosage there-
NIPT for Turner syndrome, Klinefelter syndrome, fore has to be identified against a background of maternal
47,XXX and 47,XYY based on a study by Mazloom X-chromosomal DNA that is likely to be quantitatively
et al.100 , with DRs of 83% (25/30), 85% (11/13), 83% more variable than is seen for the autosomes. These con-
(5/6) and 75% (3/4), respectively, for each of the these siderations suggest that NIPT based on simply counting
abnormalities. The FPR for Turner syndrome was 0.2% the total X- and Y-chromosomal sequences in maternal
(4/1945) and there was one other false positive (47,XXX) plasma and comparing them with the expected normal
in the series100 . Of the normal cases in which gender was karyotype values may be less effective than that which is
interpreted, it was inconsistent with the karyotype result achievable for autosomal gain or loss. Furthermore, it may
in 0.7% (13/1814)100 . Bianchi et al.68 developed a com- be misleading to quote a DR that is based on all sex chro-
plex algorithm for evaluating sex-chromosome-derived mosomal abnormalities combined for genetic counseling.
cfDNA in plasma. Their study derived z-scores for the Approaches based on quantification of SNPs present
X-chromosome (zX ) and Y-chromosome (zY ). A result was in the plasma DNA may be informative for at least some
classified as positive for Turner syndrome if zX < 4.0 and fetal sex chromosomal aneuploidies. Y-chromosome
zY < 2.5; it was classified as 46,XX or 46,XY or 47,XXX gain, most cases of Turner syndrome108 , approximately
or 47,XXY or 47,XYY using different cut-offs, and all half of cases with Klinefelter syndrome109 and some other
others were unclassified. Among the non-mosaic Turner imbalances are attributable to erroneous segregation of
syndrome cases the DR was 75% (15/20) and the FPR was a paternally derived sex chromosome. These cases could
0.2% (1/462), with 49 remaining unclassified (including therefore be identified in a maternal plasma specimen
four affected and 45 controls). Among the mosaic cases through the over- or under-representation of paternal X-
with a 45,X cell line, the DR for the monosomy was and Y-chromosomal SNPs that differ from the mothers
29% (2/7). For the other non-mosaic sex chromosomal SNPs. On the other hand, most cases of 47,XXX110 ,
abnormalities the DRs were: XXX, 75% (3/4); XYY, and the remaining cases of Turner syndrome, Klinefelter
100% (3/3); and Klinefelter syndrome, 67% (2/3). Robust syndrome and others are attributed to erroneous segre-
estimates for the efficacy of sex chromosomal aneuploidy gation of a maternally derived chromosome. The reliable
detection are therefore not provided, although Verinata quantification of cfDNA with these fetal X-chromosomal
Inc. does offer testing on the basis of this study. SNPs, against a background of the identical maternal
There are several factors that could confound the SNPs, is more difficult.
development of highly effective NIPT for X- and Y-
chromosomal imbalances. First, many fetal sex chromo- Mosaicism
somal aneuploidies are mosaic, with either a normal cell
line or an additional abnormal cell line. This includes Two of the NIPT clinical trials have specifically addressed
situations in which one abnormal cell line may mask the ability to detect mosaic cytogenetic abnormalities in
another when cfDNA is analyzed. For example, fetal the fetus68,111 . In one study68 , of 11 cases in which a non-
45,X/46,XXX mosaicism may show approximately nor- complex mosaicism was known to be present based on
mal proportions of X-chromosome cfDNA if the propor- karyotyping, five were called affected and six unaffected.
tions of the two cell types are similar. Second, although In additional studies involving deeper sequencing,
the detection of Y-chromosome-specific DNA is relatively four mosaic abnormalities recognized by conventional
simple and reliable in establishing gender101 , finding suf- chromosomal analysis were not detected by NIPT112 . In
ficient Y-chromosome loci that are informative for copy the second study involving five cases111 , the abnormal line
number quantification may be difficult. The potentially was detected in one case and not detected in four cases.
informative euchromatic part of the Y-chromosome is Approximately 14% of all cytogenetic abnormalities
relatively small, it contains many sequences that show identified through karyotyping of amniotic fluid cells are
polymorphic CNVs, and some segments show partial mosaic98 . For CVS, the proportion showing mosaicism
homology with X-chromosomal sequences102 . A third is much higher; approximately 45%23 . This excludes
difficulty is that normal adult females show an age-related cases in which there is pseudomosaicism (possible cell
loss of X-chromosomes103 . In women at normal reproduc- culture artifact) and includes cases in which there is a
tive age, the proportion of maternal cells with a missing discrepancy between CVS direct or short-term culture
X-chromosome can be comparable to the proportion of preparation results (cytotrophoblasts) and longer term cell
cfDNA in a maternal plasma specimen and there may be culture results (mesenchymal tissue). Discrepancy usually
some women with unusually high levels of 45,X cells104 . involves abnormal cells in cytotrophoblasts with normal

Copyright 2013 ISUOG. Published by John Wiley & Sons Ltd. Ultrasound Obstet Gynecol 2013; 42: 1533.
26 Benn et al.

mesenchyme, but the reverse situation can also be present. cases (four involving chromosome 13, one chromosome
An abnormal cell line can also be present in both direct and 18 and one chromosome 21) interpreted as a false-positive
long-term cultures but be undetectable in the fetus. Longer CPM result and one false-negative result (chromosome
term cell culture is usually considered to be a more reliable 18) among 6560 cases that had analysis of a direct CVS
indicator of the true karyotype of the fetus compared preparation. Similarly, Smith et al.121 reported nine cases
with direct preparations. Overall, approximately 1% of (two involving chromosome 13, four chromosome 18 and
all CVS specimens will show a second cell line that is three chromosome 21) interpreted as a false-positive CPM
not detectable in the fetus, a situation termed confined result and five false-negative results (two chromosome 18,
placental mosaicism (CPM)113 . This may sometimes three chromosome 21) in approximately 18 195 cases
result in spontaneous loss, low birth weight or preterm that had direct preparation analysis. Combining these
delivery, particularly when it involves both trophoblastic data, for chromosome 21, 18 or 13, the incidence of
and mesenchymal cell lineages114 and/or when it involves false positives was 1 in 1650 and the incidence of false
specific karyotype abnormalities such as trisomy 16115 . negatives was 1 in 4126. These estimates are crude because
However, in most pregnancies in which CPM is detected the number of cells analyzed on direct CVS preparation
the pregnancy outcome is normal116 . is often small, some women receiving an abnormal result
The frequency and significance of CPM is relevant to terminated their pregnancy without confirmation, and
NIPT because cfDNA is believed to originate primarily low-level true fetal mosaicism may have gone undetected.
from non-viable trophoblasts. CPM could result in either It should be noted that most cases of CPM do not
false-positive or false-negative NIPT results. It has been involve chromosomes 21, 18 and 13. Therefore, as NIPT
suggested that viable pregnancies with Edwards or Patau tests are expanded to include additional chromosomes or
syndrome may have a substantial euploid cell line in chromosomal regions, the issue of erroneous results due
the trophoblasts117,118 and therefore NIPT could fail to to CPM is likely to become increasingly important.
detect some viable trisomic cases in which there is a For those cases that do show true fetal mosaicism, the
substantial population of abnormal cells in the fetus but phenotype can be highly variable but will generally be less
not in the placenta. This would lead to an apparent false- severe than that present with the non-mosaic abnormal
negative result by NIPT. Conversely, the trophoblasts karyotype22 . Although the proportion of abnormal cells
may be mostly abnormal and the fetal cells normal, in amniotic fluid or chorionic villi can be a poor
leading to an apparent false-positive result. Moreover, if guide to phenotype in individual cases, in general, a
chromosomally abnormal trophoblasts were more likely high proportion of abnormal cells is associated with
to undergo apoptosis, NIPT could preferentially identify adverse pregnancy outcome122 . NIPT tests that only
cases in which the proportion of abnormal cells in the present a result categorically as either positive or
placenta is low. negative obviously cannot provide any information
A number of cases have already been described which about the proportion of abnormal cells or clinical severity.
illustrate how differences in the distribution of normal Furthermore, since some of the clinical trials have
and abnormal cells may lead to test result discrepancies specifically excluded mosaic cases and optimized their
between NIPT and karyotyping. Faas et al.43 described the z-score cut-off on the basis of non-mosaic cases, testing is
identification of Turner syndrome based on NIPT when presumably biased towards not detecting mosaic cases. It
the abnormal cell line was confirmed by cytogenetic anal- is not yet clear whether patients with z-scores close to the
ysis of short-term CVS cultures but the abnormality was cut-off should be offered invasive testing to help rule out
not seen in the long-term CVS cultures or in the newborn mosaic trisomy. To present this problem appropriately
blood. Choi et al.119 described a case involving possible to clinicians, and also to allow better integration of the
trisomy 22 detected by cfDNA testing. The trisomy 22 results with other screening and diagnostic techniques,
cell line was confirmed by analyzing the term placenta we recommend that NIPT reports should be formatted
but the abnormality was not detected in a blood specimen so that they provide patient-specific risks for each non-
from the dysmorphic baby. Hall et al.120 describe a mosaic trisomy, the z-score and the fetal fraction, rather
case with a positive Patau syndrome cfDNA test, mosaic than just a categorical positive or negative finding.
47,XY,+13/46,XY result for CVS, normal result for amni- The possibility of maternal mosaicism also needs to be
otic fluid cells, normal result for cord blood, but trisomy considered. This includes the rare situation in which the
13 mosaicism confirmed in the placenta. These examples abnormal maternal cfDNA is derived from a malignant
can each be interpreted as CPM although the presence of cell population123 . Maternal mosaicism could potentially
low-level true fetal mosaicism cannot be excluded. be recognized using SNP-based NIPT methodology.
Early trials on chromosomal analysis following CVS
provide an indication as to how often CPM might lead to Translocations
an apparent false-positive or false-negative result. Because
milder abnormalities such as sex chromosomal aneuploidy Unbalanced translocations, in theory, should be identi-
or mosaicism involving abnormalities not typically seen fiable in NIPT as a result of the partial trisomy and
in livebirths may not have been fully pursued in follow- monosomy that is present. In a retrospective analysis of
up studies, this analysis needs to be confined to Down, six plasma specimens from pregnant women with known
Edwards and Patau syndrome. Ledbetter et al.23 noted six fetal imbalances, Srinivasan et al.112 were able to identify

Copyright 2013 ISUOG. Published by John Wiley & Sons Ltd. Ultrasound Obstet Gynecol 2013; 42: 1533.
NIPT for aneuploidy 27

the imbalances in maternal plasma in all six cases. In two translocation relies on the identification of a relative excess
of their cases the karyotype showed additional material of of the translocation chromosomal SNPs and relative defi-
unknown origin and the NIPT analysis was able to provide ciency of the normal homolog SNPs in the plasma sample.
a more precise characterization of the extra chromosomal
material. The ability to detect routinely smaller imbal-
Triploidy
ances would require deeper sequencing and improvements
in bioinformatic analysis of sequencing data41,124 126 . Triploid pregnancies can arise as a result of an
For SNP-based approaches to NIPT, detection of small additional set of maternal (digynic) or paternal (diandric)
imbalances requires identification of sufficient informative chromosomes. Most recognized cases show a 69,XXX
polymorphisms in the region of interest. or 69,XXY karyotype; 69,XYY forms are thought to
Conventional chromosomal analysis does allow detec- undergo early first-trimester miscarriage133 . Most cases
tion of most apparently balanced translocations and large of triploidy will be identified by ultrasound abnormality
inversions. Prenatally identified de-novo rearrangements and characteristic first- or second trimester-maternal
are associated with an increased risk for abnormality127 . serum marker levels134 136 . It is exceptional for these
It would therefore be desirable to detect these non- pregnancies to survive into the third trimester.
invasively. Paired-end sequencing of DNA fragments that Since all chromosomes are proportionately represented
span a chromosomal breakpoint can identify at least some in a 69,XXX pregnancy, NIPT based on the counting
translocations through the analysis of cellular DNA. For of all chromosomal sequences would yield a normal
example, Schluth-Bolard et al.128 used a whole-genome result. Pregnancies with either a 69,XXY or a 69,XYY
paired-end protocol to identify the specific sequence dis- karyotype might be identifiable through unexpected sex
ruptions in four cases with apparently balanced transloca- chromosomal sequence ratios. Digynic triploidy might
tions and abnormal phenotypes. Breakpoints were mostly be particularly difficult to detect by NIPT because the
in repeat sequences and most breakpoints were associated placenta is very small and the fetal fraction may therefore
with gain or loss of some basepairs. Talkowski et al.129 be reduced137 . Consistent with this, Bianchi et al.68
used sequencing to refine breakpoints and establish a reported that of nine cases of fetal 69,XXX triploidy, there
diagnosis of CHARGE syndrome based on disruption of was insufficient fetal cfDNA for analysis in three (33%).
the CHD7 gene by analyzing amniotic fluid cells. These Using the approach that takes into consideration the
studies involved the sequencing of larger pieces of DNA SNPs present in the cfDNA, diandric triploidy could be
and were carried out in cases in which the karyotype identified through the presence of two different paternally
information was used to help identify candidate DNA inherited SNPs at some loci. Digynic cases with sufficient
fragment sequences that spanned the breakpoints. fetal cfDNA for analysis might be recognized through
Although the routine detection of de-novo rearrange- the quantitative excess of maternally inherited fetal DNA
ments using cfDNA, without knowledge of where to look, fragments relative to paternally inherited fragments.
would not seem to be technically achievable at this time,
it is potentially possible. The challenge is similar to the CNVs
detection of other mutations130 132 , but is made sim-
pler with the presence of two related reciprocal events. Small duplications and deletions have the potential to be
By sequencing cfDNA it should be possible to construct detected through NIPT. Examples that have been detected
the haplotype sequence in which one end matches per- include a 4-Mb deletion on chromosome 12138 , and two
fectly to a chromosome, the other end matches perfectly cases in which 3-Mb 22q11.2 deletions were present139 .
a second chromosomal location and there are similar In a series of 11 prenatal plasma specimens, Srinivasan
hybrid sequences present that correspond to the recipro- et al.112 identified small gains or losses not reported in
cal translocation product(s). Improved sequencing fidelity, the clinical karyotypes in eight cases, including four with
deeper sequencing and the development of advanced bio- two such variations. However, it is not clear whether they
metric algorithms that specifically look for the hybrid were true or false positives.
sequences would be needed. A variety of strategies have been proposed to detect the
Inherited rearrangements are generally considered small deletions, insertions, inversions, and duplications
benign, but their detection through karyotyping follow- that are common in the human genome140 . Much of
ing CVS or amniocentesis is considered advantageous the analysis is contingent on the ability to identify
because it identifies a risk factor for future pregnancies. departures in the location and orientation of sequenced
When a balanced translocation is already known to be DNA fragments from where they are expected to be found
segregating in a family and the phase of SNPs on the in reference mapped genomes. The ability to do this type
chromosomes in the region of interest can be established, of analysis in a non-invasive test is a long-term goal in
the presence or absence of the breakpoint can be inferred molecular cytogenetics.
from the SNPs in the cfDNA in a plasma specimen. In the
case of a paternally inherited translocation, the presence Overview of future testing
of the translocation chromosomal SNPs and absence of
the normal homolog SNPs identifies the translocation in Currently, only methods based on MPS have been the
the conceptus. In maternal inheritance, detection of the subject of clinical trials and can be viewed as being

Copyright 2013 ISUOG. Published by John Wiley & Sons Ltd. Ultrasound Obstet Gynecol 2013; 42: 1533.
28 Benn et al.

suitable for clinical use. An optimal NIPT for fetal variable expressivity. In addition, the testing could expose
aneuploidy will be accurate, simple, cheap, applicable consanguinity or non-paternity. Given these realities, the
early in pregnancy and fully compatible with existing challenge now facing those providing pretest counseling
prenatal screening methods so that risks developed using cannot be understated. The National Society of Genetic
the different approaches can be combined. Many of the Counselors recognizes that due to limited resources,
methods under consideration can be refined to allow for pretest counseling cannot always be provided by a
the detection of other chromosomal imbalances. MPS genetic counselor and this information will therefore also
refinements that will aid this process include selection need to be communicated by other qualified healthcare
of optimal sequences for analysis, adjustments for base providers141 .
composition and the use of sequencing platforms that The availability of professional guidelines, such
provide higher read accuracy. those from the American College of Obstetricians and
Sequencing also provides opportunities to detect the Gynecologists, together with the Society for Maternal-
presence or absence of single gene disorders130 . The Fetal Medicine142 , the Society of Obstetricians and
methods can even be applied to the construction of an Gynaecologists of Canada143 , the National Society of
entire fetal genotype131,132 . On the other hand, methods Genetic Counselors144 , the American College of Medical
that do not require sequence information from the parents Genetics and Genomics145 and the International Society
or which do not generate unwanted additional genotypic for Prenatal Diagnosis (ISPD)146 , can greatly facilitate
status of the parents (for example resulting in information the process by clarifying which processes are scientifically
about paternity or other disease risks) should also be valid and clinically acceptable standards of care.
considered advantageous. The optimal technologies may
ultimately be those which are most amenable to answering
only those clinical questions that are being asked, rather ETHICAL ISSUES
than those which provide the most information. The promise of NIPT is that it will reduce the number of
women with an indication for invasive testing and only
COUNSELING those women who are at very high risk for defined disor-
ders such as Down syndrome will then need to deal with
The internationally accepted approach to dealing with the ethical challenges associated with procedure-related
patients diverse ethical, religious and cultural values is to loss and pregnancy termination. However, the ease of
provide individual non-directive counseling, testing and providing NIPT will potentially result in substantially
other clinical information at each step in the screening and more prenatal diagnoses and terminations of affected
diagnosis process. This model, which maximizes patient pregnancies18 . The issue is confounded by an inability to
autonomy in reproductive decision making, has been provide adequate counseling for all women who might be
adopted by diverse populations throughout the world. considering NIPT147 . There may also be a shift in patients
Counseling of women who are considering NIPT is and/or healthcare providers views on prenatal diagnosis,
challenging. The range of cytogenetic abnormalities cur- with a greater expectation for pregnancy termination
rently detectable through cfDNA testing is smaller than as the usual response to an abnormal diagnosis. This
that detectable by conventional karyotyping and substan- has been referred to as normalization of testing and
tially less than that achievable through microarray testing. termination148 . Potential consequences could include a
In the absence of public health policies that define testing sharply reduced incidence of Down syndrome neonates,
strategies, each high-risk woman who previously would greater emphasis on prevention and less on support and
have made a decision about amniocentesis or CVS and accommodation of their disabilities, and altered attitudes
karyotyping, will now need to choose between no testing, to individuals with Down syndrome and their parents
NIPT, invasive testing with conventional cytogenetics or who chose not to test or terminate an affected pregnancy.
invasive testing with microarray testing. These women will The currently available NIPT for aneuploidy is the
need to be counseled carefully regarding the complex ben- consequence of a massive investment by private companies
efits, hazards and trade-offs associated with each option. and venture capital, and so far is only provided by private,
In part, these decisions will be determined by the for-profit companies. Aspects of the testing are the subject
indication for testing and perceived risk, but they will also of contested patents and other proprietary ownership.
be determined by the available alternative testing services. Claims of exclusive intellectual property ownership can
For example, in a situation in which routine invasive inflate costs, restrict access to test protocols and limit
testing is based on RAT for a limited set of disorders, the further test development. Although there is currently
choice of NIPT that detects the same range of aneuploidies no direct-to-consumer testing, there are press releases,
might seem preferable. On the other hand, offering array patient information materials and extensive marketing to
technology may provide much more valuable information healthcare providers, many of whom may have a poor
concerning fetal wellbeing than can current NIPT. This understanding of the true performance of the tests.
benefit has to be weighed against the risk of losing a In the future, NIPT may also be encouraged by govern-
normal pregnancy following an invasive procedure or the ments or insurance carriers who view genetic disorders as
heightened anxiety caused by the presence of a CNV of an economic burden149 . The concern is that these financial
unknown significance, poorly defined penetrance and/or pressures will be the dominating factor in determining

Copyright 2013 ISUOG. Published by John Wiley & Sons Ltd. Ultrasound Obstet Gynecol 2013; 42: 1533.
NIPT for aneuploidy 29

who has access to testing, without adequately considering 13. Smid M, Galbiati S, Vassallo A, Gambini D, Ferrari A, Viora
medical need. Changes in financial support for testing, E, Pagliano M, Restagno G, Ferrari M, Cremonesi L. No
evidence of fetal DNA persistence in maternal plasma after
counseling and clinical services can also indirectly
pregnancy. Hum Genet 2003; 112: 617618.
compromise individual patient autonomy when deciding 14. Hui L, Vaughan JI, Nelson M. Effect of labor on postpartum
whether to continue or terminate an affected pregnancy. clearance of cell-free fetal DNA from the maternal circulation.
It is also worth noting that the early history of NIPT Prenat Diagn 2008; 28: 304308.
shows how easily test quality can be compromised when 15. Poon LL, Leung TN, Lau TK, Lo YM. Presence of fetal RNA
in maternal plasma. Clin Chem 2000; 46: 18321834.
development and introduction is left entirely to unregu-
16. Ng EK, Tsui NB, Lam NY, Chiu RW, Yu SC, Wong SC, Lo
lated free market forces150,151 . ISPD, in its latest Position ES, Rainer TH, Johnson PJ, Lo YM. Presence of filterable
Statement on NIPT has strongly emphasized the impor- and nonfilterable mRNA in the plasma of cancer patients and
tance of adoption of laboratory standards for NIPT146 . healthy individuals. Clin Chem 2002; 48: 12121217.
17. Ng EK, Tsui NB, Lau TK, Leung TN, Chiu RW, Panesar
NS, Lit LC, Chan KW, Lo YM. mRNA of placental origin is
DISCLOSURES readily detectable in maternal plasma. Proc Natl Acad Sci U S
A 2003; 100: 47484753.
H. Cuckle is a consultant to PerkinElmer Inc, Ariosa 18. Norton ME, Rose NC, Benn P. Noninvasive prenatal testing
for fetal aneuploidy: clinical assessment and a plea for
Diagnostics Inc, Natera Inc, and director of Genome Ltd;
restraint. Obstet Gynecol 2013; 121: 847850.
E. Pergament is a consultant to PerkinElmer Inc and 19. Alamillo CML, Krantz D, Evans M, Fiddler M, Pergament
Natera Inc, and director of Northwestern Reproductive E. Nearly a third of abnormalities found after first-trimester
Genetics Inc. screening are different than expected: 10-year experience from
a single center. Prenat Diagn 2013; 33: 251256.
20. Kazerouni NN, Currier RJ, Flessel M, Goldman S, Hennigan
C, Hodgkinson C, Lorey F, Malm L, Tempelis C, Roberson M.
REFERENCES
Detection rate of quadruple-marker screening determined by
1. Ferguson-Smith MA, Yates JRW. Maternal age specific clinical follow-up and registry data in the statewide California
rates for chromosome aberrations and factors influencing program, July 2007 to February 2009. Prenat Diagn 2011;
them: report of a collaborative European study on 52965 31: 901906.
amniocenteses. Prenat Diagn 1984; 4 Spec No: 544. 21. Hulten MA, Dhanjal S, Pertl B. Rapid and simple prenatal
2. Tabor A, Alfirevic Z. Update on procedure-related risks for diagnosis of common chromosome disorders: advantages and
prenatal diagnosis techniques. Fetal Diagn Ther 2010; 27: disadvantages of the molecular methods FISH and QF-PCR.
17. Reproduction 2003; 126: 279297.
3. Benn PA, Egan JF, Fang M, Smith-Bindman R. Changes in the 22. Benn PA. Prenatal diagnosis of chromosome abnormalities
utilization of prenatal diagnosis. Obstet Gynecol 2004; 103: through amniocentesis. In Genetic Disorders and the Fetus:
12551260. Diagnosis, Prevention and Treatment (6th edn), Milunsky A,
4. Cuckle H, Benn P. Multianalyte maternal serum screening Milunsky JM (eds). Wiley-Blackwell: Chichester, UK, 2010:
for chromosomal defects. In Genetic Disorders and the Fetus: 771818.
Diagnosis, Prevention and Treatment (6th edn), Milunsky A, 23. Ledbetter DH, Zachary JM, Simpson JL, Golbus MS,
Milunsky JM (eds). Wiley-Blackwell, Chichester, UK, 2010; Pergament E, Jackson L, Mahoney MJ, Desnick RJ, Schulman
771818. J, Copeland KL, Verlinsky Y, Yang-Feng T, Schonberg SA,
5. Syngelaki A, Chelemen T, Dagklis T, Allan L, Nicolaides Babu A, Tharapel A, Dorfmann A, Lubs HA, Rhoads GG,
KH. Challenges in the diagnosis of fetal non-chromosomal Fowler SE, De La Cruz F. Cytogenetic results from the
abnormalities at 1113 weeks. Prenat Diagn 2011; 31: U.S. Collaborative Study on CVS. Prenat Diagn 1992; 12:
90102. 317345.
6. Lo YMD, Corbetta N, Chamberlain PF, Rai V, Sargent IL, 24. Evans MI, Henry GP, Miller WA, Bui TH, Snidjers RJ,
Redman CWG, Wainscoat JS. Presence of fetal DNA in Wapner RJ, Miny P, Johnson MP, Peakman D, Johnson A,
maternal plasma and serum. Lancet 1997; 350: 485487. Nicolaides K, Holzgreve W, Ebrahim SA, Babu R, Jackson
7. Lo YMD, Tein MS, Lau TK, Haines CJ, Leung TN, Poon L. International, collaborative assessment of 146,000 prenatal
PM, Wainscoat JS, Johnson PJ, Chang AM, Hjelm NM. karyotypes: expected limitations if only chromosome-specific
Quantitative analysis of fetal DNA in maternal plasma and probes and fluorescent in-situ hybridization are used. Hum
serum: implications for noninvasive prenatal diagnosis. Am J Reprod 1999; 14: 12131216.
Hum Genet 1998; 62: 768775. 25. Vialard F, Simoni G, Aboura A, De Toffol S, Molina Gomes
8. Lun FM, Chiu RW, Allen Chan KC, Yeung Leung T, Kin Lau D, Marcato L, Serero S, Clement P, Bouhanna P, Rouleau E,
T, Dennis Lo YM. Microfluidics digital PCR reveals a higher Grimi B, Selva J, Gaetani E, Maggi F, Joseph A, Benzacken
than expected fraction of fetal DNA in maternal plasma. Clin B, Grati FR. Prenatal BACs-on-Beads: a new technology for
Chem 2008; 54: 16641672. rapid detection of aneuploidies and microdeletions in prenatal
9. Illanes S, Denbow M, Kailasam C, Finning K, Soothill PW. diagnosis. Prenat Diagn 2011; 31: 500508.
Early detection of cell-free fetal DNA in maternal plasma. 26. Mann K, Hills A, Donaghue C, Thomas H, Ogilvie CM.
Early Hum Dev 2007; 83: 563566. Quantitative fluorescence PCR analysis of > 40,000 prenatal
10. Chan KC, Zhang J, Hui AB, Wong N, Lau TK, Leung TN, Lo samples for the rapid diagnosis of trisomies 13, 18 and 21 and
KW, Huang DW, Lo YM. Size distributions of maternal and monosomy X. Prenat Diagn 2012; 32: 11971204.
fetal DNA in maternal plasma. Clin Chem 2004; 50: 8892. 27. Gerdes T, Kirchhoff M, Lind AM, Vestergaard Larsen G,
11. Li Y, Zimmermann B, Rusterholz C, Kang A, Holzgreve Kjaergaard S. Multiplex ligation-dependent probe amplifica-
W, Hahn S. Size separation of circulatory DNA in maternal tion (MLPA) in prenatal diagnosis-experience of a large series
plasma permits ready detection of fetal DNA polymorphisms. of rapid testing for aneuploidy of chromosomes 13, 18, 21, X,
Clin Chem 2004; 50: 10021011. and Y. Prenat Diagn 2008; 28: 11191125.
12. Lo YMD, Zhang J, Leung TN, Lau TK, Chang AM, Hjelm 28. Caine A, Maltby AE, Parkin CA, Waters JJ, Crolla JA;
NM. Rapid clearance of fetal DNA from maternal plasma. UK Association of Clinical Cytogeneticists (ACC). Prenatal
Am J Hum Genet 1999; 64: 218224. detection of Downs syndrome by rapid aneuploidy testing for

Copyright 2013 ISUOG. Published by John Wiley & Sons Ltd. Ultrasound Obstet Gynecol 2013; 42: 1533.
30 Benn et al.

chromosomes 13, 18, and 21 by FISH or PCR without a full 43. Faas BH, de Ligt J, Janssen I, Eggink AJ, Wijnberger LD,
karyotype: a cytogenetic risk assessment. Lancet 2005; 366: van Vugt JM, Vissers L, Geurts van Kessel A. Non-invasive
123128. prenatal diagnosis of fetal aneuploidies using massively parallel
29. Test and Technology Transfer Committee. Technical and sequencing-by-ligation and evidence that cell-free fetal DNA
clinical assessment of fluorescence in situ hybridization: An in the maternal plasma originates from cytotrophoblastic cells.
ACMG/ASHG position statement. I. Technical considerations. Expert Opin Biol Ther 2012; 12 (Suppl 1): S1926.
Genet Med 2000; 2: 356361. 44. van den Oever JM, Balkassmi S, Verweij EJ, van Iterson M,
30. Wapner RJ, Martin CL, Levy B, Ballif BC, Eng CM, Zachary Adama van Scheltema PN, Oepkes D, van Lith JM, Hoffer
JM, Savage M, Platt LD, Saltzman D, Grobman WA, Klugman MJ, den Dunnen JT, Bakker E, Boon EM. Single molecule
S, Scholl T, Simpson JL, McCall K, Aggarwal VS, Bunke sequencing of free DNA from maternal plasma for noninvasive
B, Nahum O, Patel A, Lamb AN, Thom EA, Beaudet trisomy 21 detection. Clin Chem 2012; 58: 699706.
AL, Ledbetter DH, Shaffer LG, Jackson L. Chromosomal 45. Sparks AB, Wang ET, Struble CA, Barrett W, Stokowski R,
microarray versus karyotyping for prenatal diagnosis. N Engl McBride C, Zahn J, Lee K, Shen N, Doshi J, Sun M, Garrison J,
J Med 2012; 367: 21752184. Sandler J, Hollemon D, Pattee P, Tomita-Mitchell A, Mitchell
31. Tabor A, Madsen M, Obel EB, Philip J, Bang J, Norgaard- M, Stuelpnagel J, Song K, Oliphant A. Selective analysis of cell-
Pedersen B. Randomised controlled trial of genetic amniocen- free DNA in maternal blood for evaluation of fetal trisomy.
tesis in 4606 low-risk women. Lancet 1986; i: 12871292. Prenat Diagn 2012; 32: 39.
32. Alfirevic Z, Mujezinovic F, Sundberg K, Brigham S. 46. Dhallan R, Guo X, Emche S, Damewood M, Bayliss P, Cronin
Amniocentesis and chorionic villus sampling for prenatal M, Barry J, Betz J, Franz K, Gold K, Vallecillo B, Varney J.
diagnosis. Cochrane Database System Rev 2003; Issue 3: A non-invasive test for prenatal diagnosis based on fetal DNA
CD003252. present in maternal blood: a preliminary study. Lancet 2007;
33. Dhallan R, Au WC, Mattagajasingh S, Emche S, Bayliss P, 369: 474481.
Damewood M, Cronin M, Chou V, Mohr M. Methods to 47. Ghanta S, Mitchell ME, Ames M, Hidestrand M, Simpson
increase the percentage of free fetal DNA recovered from the P, Goetsch M, Thilly WG, Struble CA, Tomita-Mitchell A.
maternal circulation. JAMA 2004; 291: 11141119. Non-invasive prenatal detection of trisomy 21 using tandem
34. Go AT, van Vugt JM, Oudejans CB. Non-invasive aneuploidy single nucleotide polymorphisms. PLoS One 2010; 5: e13184.
detection using free fetal DNA and RNA in maternal plasma: 48. Zimmermann B, Hill M, Gemelos G, Demko Z, Banjevic M,
recent progress and future possibilities. Hum Reprod Update Baner J, Ryan A, Sigurjonsson S, Chopra N, Dodd M, Levy
2011; 17: 372382. B, Rabinowitz M. Noninvasive prenatal aneuploidy testing of
35. Papageorgiou EA, Fiegler H, Rakyan V, Beck S, Hulten M,
chromosomes 13, 18, 21, X, and Y, using targeted sequencing
Lamnissou K, Carter NP, Patsalis PC. Sites of differential DNA
of polymorphic loci. Prenat Diagn 2012; 32: 12331241.
methylation between placenta and peripheral blood: molecular
49. Liao GJ, Chan KC, Jiang P, Sun H, Leung TY, Chiu RW,
markers for noninvasive prenatal diagnosis of aneuploidies.
Lo YM. Noninvasive prenatal diagnosis of fetal trisomy
Am J Pathol 2009; 174: 16091618.
21 by allelic ratio analysis using targeted massively parallel
36. Fan HC, Blumenfeld YJ, Chitkara U, Hudgins L, Quake
sequencing of maternal plasma DNA. PLoS One 2012; 7:
SR. Noninvasive diagnosis of fetal aneuploidy by shotgun
e38154.
sequencing DNA from maternal blood. Proc Natl Acad Sci U
50. Poon LLM, Leung TN, Lau TK, Chow KC, Lo YMD.
S A 2008; 105: 1626616271.
Differential DNA methylation between fetus and mother as
37. Chiu RW, Chan KC, Gao Y, Lau VY, Zheng W, Leung TY, Foo
a strategy for detecting fetal DNA in maternal plasma. Clin
CH, Xie B, Tsui NB, Lun FM, Zee BC, Lau TK, Cantor CR,
Chem 2002; 48: 3541.
Lo YM. Noninvasive prenatal diagnosis of fetal chromosomal
51. Tong YK, Ding C, Chiu RW, Gerovassili A, Chim SS, Leung
aneuploidy by massively parallel genomic sequencing of DNA
in maternal plasma. Proc Natl Acad Sci U S A 2008; 105: TY, Leung TN, Lau TK, Nicolaides KH, Lo YM. Noninvasive
2045820463. prenatal detection of fetal trisomy 18 by epigenetic allelic
38. Sehnert AJ, Rhees B, Comstock D, Comstock D, de Feo ratio analysis in maternal plasma: theoretical and empirical
E, Heilek G, Burke J, Rava RP. Optimal detection of considerations. Clin Chem 2006; 52: 21942202.
fetal chromosomal abnormalities by massively parallel DNA 52. Tong YK, Jin S, Chiu RW, Ding C, Chan KC, Leung TY, Yu L,
sequencing of cell-free fetal DNA from maternal blood. Clin Lau TK, Lo YM. Noninvasive prenatal detection of trisomy 21
Chem 2011; 57: 10421049. by an epigenetic-genetic chromosome-dosage approach. Clin
39. Dan S, Wang W, Ren J, Li Y, Hu H, Xu Z, Lau TK, Xie J, Chem 2010; 56: 9098.
Zhao W, Huang H, Xie J, Sun L, Zhang X, Wang W, Liao S, 53. Chim SS, Jin S, Lee TY, Lun FM, Lee WS, Chan LY, Jin Y,
Qiang R, Cao J, Zhang Q, Zhou Y, Zhu H, Zhong M, Guo Yang N, Tong YK, Leung TY, Lau TK, Ding C, Chiu RW,
Y, Lin L, Gao Z, Yao H, Zhang H, Zhao L, Jiang F, Chen F, Lo YM. Systematic search for placental DNA-methylation
Jiang H, Li S, Li Y, Wang J, Wang J, Duan T, Su Y, Zhang markers on chromosome 21: toward a maternal plasma-based
X. Clinical application of massively parallel sequencing-based epigenetic test for fetal trisomy 21. Clin Chem 2008; 54:
prenatal noninvasive fetal trisomy test for trisomies 21 and 18 500511.
in 11,105 pregnancies with mixed risk factors. Prenat Diagn 54. Papageorgiou EA, Karagrigoriou A, Tsaliki E, Velissariou V,
2012; 32: 12251232. Carter NP, Patsalis PC. Fetal-specific DNA methylation ratio
40. Fan HC, Quake SR. Sensitivity of noninvasive prenatal permits noninvasive prenatal diagnosis of trisomy 21. Nat
detection of fetal aneuploidy from maternal plasma using Med 2011; 17: 510513.
shotgun sequencing is limited only by counting statistics. PLoS 55. Tsaliki E, Papageorgiou EA, Spyrou C, Koumbaris G,
One 2010; 5: e10439. Kypri E, Kyriakou S, Sotiriou C, Touvana E, Keravnou A,
41. Liang D, Lv W, Wang H, Xu L, Liu J, Li H, Hu L, Peng Y, Wu Karagrigoriou A, Lamnissou K, Velissariou V, Patsalis PC.
L. Non-invasive prenatal testing of fetal whole chromosome MeDIP real-time qPCR of maternal peripheral blood reliably
aneuploidy by massively parallel sequencing. Prenat Diagn identifies trisomy 21. Prenat Diagn 2012; 32: 9961001.
2013; 33: 409415. 56. Fan HC, Quake SR. Detection of aneuploidy with digital
42. Chiu RW, Sun H, Akolekar R, Clouser C, Lee C, McKernan polymerase chain reaction. Anal Chem 2007; 79: 75767579.
K, Zhou D, Nicolaides KH, Lo YM. Maternal plasma DNA 57. Lo YM, Lun FM, Chan KC, Tsui NB, Chong KC, Lau TK,
analysis with massively parallel sequencing by ligation for Leung TY, Zee BC, Cantor CR, Chiu RW. Digital PCR for the
noninvasive prenatal diagnosis of trisomy 21. Clin Chem molecular detection of fetal chromosomal aneuploidy. Proc
2010; 56: 459463. Natl Acad Sci U S A 2007; 104: 1311613121.

Copyright 2013 ISUOG. Published by John Wiley & Sons Ltd. Ultrasound Obstet Gynecol 2013; 42: 1533.
NIPT for aneuploidy 31

58. Fan HC, Blumenfeld YJ, El-Sayed YY, Chueh J, Quake SR. Lee H, Jacobsson B, Tomlinson MW, Oepkes D, Hollemon D,
Microfluidic digital PCR enables rapid prenatal diagnosis of Sparks AB, Oliphant A, Song K. Non-Invasive Chromosomal
fetal aneuploidy. Am J Obstet Gynecol 2009; 200: 543.e17. Evaluation (NICE) Study: results of a multicenter prospective
59. Evans MI, Wright DA, Pergament E, Cuckle HS, Nicolaides cohort study for detection of fetal trisomy 21 and trisomy 18.
KH. Digital PCR for noninvasive detection of aneuploidy: Am J Obstet Gynecol 2012; 207: 137.e18.
power analysis equations for feasibility. Fetal Diagn Ther 72. Palomaki GE, Deciu C, Kloza EM, Lambert-Messerlian GM,
2012; 31: 244247. Haddow JE, Neveux LM, Ehrich M, van den Boom D,
60. Lo YM, Tsui NB, Chiu RW, Lau TK, Leung TN, Heung Bombard AT, Grody WW, Nelson SF, Canick JA. DNA
MM, Gerovassili A, Jin Y, Nicolaides KH, Cantor CR, Ding sequencing of maternal plasma reliably identifies trisomy 18
C. Plasma placental RNA allelic ratio permits noninvasive and trisomy 13 as well as Down syndrome: an international
prenatal chromosomal aneuploidy detection. Nat Med 2007; collaborative study. Genet Med 2012; 14: 296305.
13: 218223. 73. Lau TK, Chan MK, Salome Lo PS, Chan HY, Chan WS,
61. Go AT, Visser A, van Dijk M, Mulders MA, Eijk P, Ylstra Koo TY, NgHY, Pooh RK. Clinical utility of noninvasive
B, Blankenstein MA, van Vugt JM, Oudejans CB. A novel fetal trisomy (NIFTY) testearly experience. J Matern Fetal
method to identify syncytiotrophoblast-derived RNA products Neonatal Med 2012; 25: 18561859.
representative of trisomy 21 placental RNA in maternal 74. Nicolaides KH, Syngelaki A, Ashoor G, Birdir C, Touzet G.
plasma. Methods Mol Biol 2008; 444: 291302. Noninvasive prenatal testing for fetal trisomies in a routinely
62. Chen EZ, Chiu RW, Sun H, Akolekar R, Chan KC, Leung screened first-trimester population. Am J Obstet Gynecol
TY, Jiang P, Zheng YW, Lun FM, Chan LY, Jin Y, Go AT, 2012; 207: 374.e16.
Lau ET, To WW, Leung WC, Tang RY, Au-Yeung SK, Lam 75. Ashoor G, Syngelaki A, Wang E, Struble C, Oliphant A,
H, Kung YY, Zhang X, van Vugt JM, Minekawa R, Tang Song K, Nicolaides KH. Trisomy 13 detection in the first
MH, Wang J, Oudejans CB, Lau TK, Nicolaides KH, Lo YM. trimester of pregnancy using a chromosome-selective cell-free
Noninvasive prenatal diagnosis of fetal trisomy 18 and trisomy DNA analysis method. Ultrasound Obstet Gynecol 2013; 41:
13 by maternal plasma DNA sequencing. PLoS One 2011; 6: 2125.
e21791. 76. Benn P, Cuckle H, Pergament E. Genome-wide fetal
63. Lau TK, Chen F, Pan X, Pooh RK, Jiang F, Li Y, Jiang H, aneuploidy detection by maternal plasma DNA sequencing.
Li X, Chen S, Zhang X. Noninvasive prenatal diagnosis of Obstet Gynecol 2012; 119: 1270.
common fetal chromosomal aneuploidies by maternal plasma 77. Futch T, Spinosa J, Bhatt S, de Feo E,Rava RP, Sehnert AJ.
DNA sequencing. J Matern Fetal Neonatal Med 2012; 25: Initial clinical laboratory experience in noninvasive prenatal
13701374.
testing for fetal aneuploidy from maternal plasma DNA
64. Guex N, Iseli C, Syngelaki A, Deluen C, Pescia G, Nicolaides
samples. Prenat Diagn 2013; 33: 569574.
KH, Xenarios I, Conrad B. A robust second-generation
78. Gil MM, Quezada MS, Bregant B, Ferraro M, Nicolaides KH.
genome-wide test for fetal aneuploidy based on shotgun
Implementation of maternal blood cell-free DNA testing in
sequencing cell-free DNA in maternal blood. Prenat Diagn
early screening for aneuploidies. Ultrasound Obstet Gynecol
2013. DOI: 10.1002/pd.4130. [Epub ahead of print].
2013; 42: 3440.
65. Chiu RW, Akolekar R, Zheng YW, Leung TY, Sun H, Chan
79. Ashoor G, Poon L, Syngelaki A, Mosimann B, Nicolaides
KC, Lun FM, Go AT, Lau ET, To WW, Leung WC, Tang
KH. Fetal fraction in maternal plasma cell-free DNA at
RY, Au-Yeung SK, Lam H, Kung YY, Zhang X, van Vugt
1113 weeks gestation: effect of maternal and fetal factors.
JM, Minekawa R, Tang MH, Wang J, Oudejans CB, Lau TK,
Fetal Diagn Ther 2012; 31: 237243.
Nicolaides KH, Lo YM. Non-invasive prenatal assessment of
80. Wegrzyn P, Faro C, Falcon O, Peralta CF, Nicolaides KH.
trisomy 21 by multiplexed maternal plasma DNA sequencing:
Placental volume measured by three-dimensional ultrasound
large scale validation study. BMJ 2011; 342: c7401.
66. Ehrich M, Deciu C, Zweifellhofer T, Tynan JA, Cagasan L, at 11 to 13 + 6 weeks of gestation: relation to chromosomal
Tim R, Lu V, McCullough R, McCarthy E, Nygren AO, Dean defects. Ultrasound Obstet Gynecol 2005; 26: 2832.
J, Tang L, Hutchison D, Lu T, Wang H, Angkachatchai 81. Brar H, Wang E, Struble C, Musci T, Norton M. The fetal
V, Oeth P, Cantor CR, Bombard A, van den Boom D. fraction of cell-free DNA in maternal plasma is not affected by
Noninvasive detection of fetal trisomy 21 by sequencing of a priori risk of fetal trisomy. J Met Fetal Neonat Med 2013;
DNA in maternal blood: a study in a clinical setting. Am J 26: 143145.
Obstet Gynecol 2011; 204; 205.e201211. 82. Wang E, Batey A, Struble C, Musci T, Song K, Oliphant
67. Palomaki GE, Kloza EM, Lambert-Messerlian GM, Haddow A. Gestational age and maternal weight effects on fetal cell-
JE, Neveux LM, Ehrich M, van den Boom D, Bombard AT, free DNA in maternal plasma. Prenat Diagn 2013. DOI:
Deciu C, Grody WW, Nelson SF, Canick JA. DNA sequencing 10.1002/pd.4119. [Epub ahead of print].
of maternal plasma to detect Down syndrome: an international 83. Nicolaides KH, Syngelaki A, Gil M, Atanasova V, Markova
clinical validation study. Genet Med 2011; 13: 913920. D. Validation of targeted sequencing of single-nucleotide poly-
68. Bianchi DW, Platt LD, Goldberg JD, Abuhamad AZ, morphisms for non-invasive prenatal detection of aneuploidy
Sehnert AJ, Rava RP, on behalf of the MatErnal BLood IS of chromosomes 13, 18, 21, X, and Y. Prenat Diagn 2013;
Source to Accurately diagnose fetal aneuploidy (MELISSA) 33: 575579.
Study Group. Genome-wide fetal aneuploidy detection by 84. Rabinowitz M, Gemelos G, Hill M, Levy B, McAdoo S, Savage
maternal plasma DNA sequencing. Obstet Gynecol 2012; M, Demko Z. Abstract 3018T. Highly accurate non-invasive
119; 890901. detection of fetal aneuploidy of chromosomes 13, 18, 21, X
69. Ashoor G, Syngelaki A, Wagner M, Birdir C, Nicolaides KH. and Y by targeted sequencing. American Society of Human
Chromosome-selective sequencing of maternal plasma cell-free Genetics Annual Meeting. November 2012. San Francisco.
DNA for first-trimester detection of trisomy 21 and trisomy 85. Rabinowitz M, Hill M, Demko Z, McAdoo S, Zimmermann
18. Am J Obstet Gynecol 2012; 206: 322.e15. B, Levy B. Abstract 591. Using targeted sequencing of SNPs
70. Sparks AB, Struble CA, Wang ET, Song K, Oliphant A. to achieve a highly accurate non-invasive detection of fetal
Noninvasive prenatal detection and selective analysis of cell- aneuploidy of 13, 18, 21 and sex chromosomes. SMFM
free DNA obtained from maternal blood: evaluation for Meeting, 2013.
trisomy 21 and trisomy 18. Am J Obstet Gynecol 2012; 86. Qu JZ, Leung TY, Jiang P, Liao GJ, Cheng YK, Sun H, Chiu
206: 319.e19. RW, Chan KC, Lo YM. Noninvasive prenatal determination
71. Norton ME, Brar H, Weiss J, Karimi A, Laurent LC, Caughey of twin zygosity by maternal plasma DNA analysis. Clin Chem
AB, Rodriguez MH, Williams J 3rd, Mitchell ME, Adair CD, 2013; 59: 427435.

Copyright 2013 ISUOG. Published by John Wiley & Sons Ltd. Ultrasound Obstet Gynecol 2013; 42: 1533.
32 Benn et al.

87. Leung TY, Qu JZZ, Liao GJW, Jiang P, Cheng YKY, Chan 104. Devi AS, Metzger DA, Luciano AA, Benn PA. 45,X/46,XX
KCA, Chiu RWK, Lo YMD. Noninvasive twin zygosity mosaicism in patients with idiopathic premature ovarian
assessment and aneuploidy detection by maternal plasma failure. Fertil Steril 1998; 70: 8993.
DNA sequencing. Prenat Diagn 2013. DOI: 10.1002/pd.4132. 105. Abruzzo MA, Mayer M, Jacobs PA. Aging and aneuploidy:
[Epub ahead of print]. evidence for the preferential involvement of the inactive X
88. Canick JA, Kloza, EM, Lambert-Messerlian GM, Haddow JE, chromosome. Cytogenet Cell Genet 1985; 39: 275278.
Ehrich M, van den Boom D, Bombard AT, Deciu C, Palomaki 106. Holzgreve W, Schonberg RG, Douglas RG, Golbus MS.
GE. DNA sequencing of maternal plasma to identify Down X-chromosome hyperdiploidy in couples with multiple
syndrome and other trisomies in multiple gestations. Prenat spontaneous abortions. Obstet Gynecol 1984; 63: 237240.
Diagn 2012; 32: 15. 107. Yoo H, Zhang L, Zhang H, Jiang F, Hu H, Chen F, Jiang
89. Lau TK, Jiang F, Chan MK, Zhang H, Salome Lo PS, Wang H, Mu F, Zhao L, Liang Z, Wang W. Noninvasive prenatal
W. Non-invasive prenatal screening of fetal Down syndrome genetic testing for fetal aneupliody detects maternal trisomy
by maternal plasma DNA sequencing in twin pregnancies. J X. Prenat Diagn 2012; 32: 11141116.
Matern Fetal Neonatal Med 2013; 26: 434437. 108. Uematsu A, Yorifuji T, Muroi J, Kawai M, Mamada M, Kaji
90. Cuckle H, Benn P, Pergament E. Maternal cfDNA screening for M, Yamanaka C, Momoi T, Nakahata T. Parental origin of
Downs syndromea cost sensitivity analysis. Prenat Diagn normal X chromosomes in Turner syndrome patients with
2013. DOI: 10.1002/pd.4157. [Epub ahead of print]. various karyotypes: implications for the mechanism leading to
91. Waitzman NJ, Romano PS, Scheffler RM. Estimates of the generation of a 45,X karyotype. Am J Med Genet 2002; 111:
economic costs of birth defects. Inquiry 1994; 31: 188205. 134139.
92. United States Bureau of Labor Statistics. CPI Detailed Report, 109. Thomas NS, Hassold TJ. Aberrant recombination and the
Data for January 2011. http: //www.bls.gov/cpi/cpi_dr.htm origin of Klinefelter syndrome. Hum Reprod Update 2003; 9:
[Accessed 8 April 2013]. 309317.
93. Ball RH, Caughey AB, Malone FD, Nyberg DA, Comstock 110. Hassold T, Hunt P. To err (meiotically) is human; the genesis
CH, Saade GR, Berkowitz RL, Gross SJ, Dugoff L, Craigo of human aneuploidy. Nat Rev Genet 2001; 2: 280291.
SD, Timor-Tritsch IE, Carr SR, Wolfe HM, Emig D, DAlton 111. Canick JA, Palomaki GE, Kloza EM, Lambert-Messerlian
ME; First and Second Trimester Evaluation of Risk (FASTER) GM, Haddow JE. The impact of maternal plasma DNA fetal
Research Consortium. First- and second-trimester evaluation fraction on next generation sequencing tests for common fetal
of risk for Down syndrome. Obstet Gynecol 2007; 110: aneuploidies. Prenat Diagn 2013. DOI: 10.1002/pd.4126.
1017. [Epub ahead of print].
94. Caughey AB, Kaimal AJ, Odibo AO. Cost-effectiveness of 112. Srinivasan A, Bianchi DW, Huang H, Sehnert AJ, Rava RP.
Down syndrome screening paradigms. Clin Lab Med 2010; Noninvasive detection of fetal subchromosome abnormalities
30: 629642. via deep sequencing of maternal plasma. Am J Hum Genet
95. Nicolaides KH, Wright D, Poon LC, Syngelaki A, Gil 2013; 92: 167176.
MM. First-trimester contingent screening for trisomy 21 113. Hahnemann JM, Vejerslev LO. Accuracy of cytogenetic
by biomarkers and maternal blood cell-free DNA testing. findings on chorionic villus sampling (CVS)-diagnostic
Ultrasound Obstet Gynecol 2013; 42: 4150. consequences of CVS mosaicism and non-mosaic discrepancy
96. Garfield SS, Armstrong SO. Clinical and cost consequences in centeres contributing to EUCROMIC 19861992. Prenat
of incorporating a novel non-invasive prenatal test into the Diagn 1997; 17: 801820.
diagnostic pathway for fetal trisomies. J Managed Care Med 114. Toutain J, Cabeau-Gauzere C, Barnetche T, Horovitz J, Saura
2012; 15: 3441. R. Confined placental mosaicism and pregnancy outcome: a
97. Song K, Musci T, Caughey AB. Clinical utility and cost of distinction needs to be made between types 2 and 3. Prenat
non-invasive prenatal testing with cfDNA analysis in high risk Diagn 2010; 30: 11551164.
women based on a U.S. population. J Matern Fetal Neonatal 115. Benn PA. Trisomy 16 and trisomy 16 mosaicism: a review.
Med 2013 [Epub ahead of print]. Am J Med Genet 1998; 79: 121133.
98. Forabosco A, Percesepe A, Santucci S. Incidence of non-age- 116. Baffero GM, Somigliana E, Crovetto F, Paffoni A, Persico
dependent chromosomal abnormalities: a population-based N, Guerneri S, Lalatta F, Fogliani R, Fedele L. Confined
study on 88965 amniocenteses. Eur J Human Genet 2009; 17: placental mosaicism at chorionic villous sampling: risk factors
897903. and pregnancy outcome. Prenat Diagn 2012; 32: 11021108.
99. Oeth P, Mazloom A, Wang T, Palomaki GE, Canick 117. Kalousek DK, Barrett IJ, McGillivray BC. Placental mosaicism
JA, Bombard A, van den Boom D, Ehrich M, Deciu C. and interuterine survival of trisomies 13 and 18. Am J Hum
Abstract 3016 T. Non-invasive prenatal sex determination Genet 1989; 44: 338343.
using massively parallel sequencing in samples from a large 118. Schuring-Blom GH, Boer K, Leschot NJ. A placental diploid
clinical validation study. American Society of Human Genetics cell line is not essential for ongoing trisomy 13 or 18
Annual Meeting. November 2012. pregnancies. Europ J Hum Genet 2001; 9: 286290.
100. Mazloom AR, Dzakula Z, Wang H, Oeth P, Jensen T, Tynan 119. Choi H, Lau TK, Jiang FM, Chan MK, Zhang HY, Lo PS, Chen
J, McCullough R,Saldivar J-S, Ehrich M, van den Boom F, Zhang L, Wang W. Fetal aneuploidy screening by maternal
D, Bombard AT, Maeder M, McLennan G, Meschino W, plasma DNA sequencing: False positive due to confined
Palomaki GE, A. Canick JA, Deciu C. Noninvasive prenatal placental mosaicism. Prenat Diagn 2013; 33: 198200.
detection of sex chromosomal aneuploidies by sequencing 120. Hall AL, Drendel HM, Verbrugge JL, Reese AM, Schumacher
circulating cell-free DNA from maternal plasma. Prenat Diagn KL, Griffith GB, Weaver DD, Abernathy MP, Litton CG,
2013; 33: 591597. Vance GH. Positive cell-free fetal DNA testing for trisomy 13
101. Devaney SA, Palomaki GE, Scott JA, Bianchi DW. Noninva- reveals confined placental mosaicism. Genet Med 2013 DOI:
sive fetal sex determination using cell-free fetal DNA. JAMA 10.1038/gim.2013.26. [Epub ahead of print].
2011; 306: 627636. 121. Smith K, Lowther G, Maher E, Hourihan T, Wilkinson
102. Tilford CA, Kuroda-Kawaguchi T, Skaletsky H, Rozen S, T, Wolstenholme J. The predictive value of findings of the
Brown LG, Rosenberg M, McPherson JD, Wylie K, Sekhon common aneuploidies, trisomies 13, 18 and 21, and numerical
M, Kucaba TA, Waterston RH, Page DC. A physical map of sex chromosome abnormalities at CVS: experience from
the human Y-chromosome. Nature 2001; 409: 943945. the ACC U.K. Collaborative Study. Association of Clinical
103. Russell LM, Strike P, Browne CE, Jacobs PA. X chromosome Cytogeneticists Prenatal Diagnosis Working Party. Prenat
loss and aging. Cytogenet Genome Res 2007; 116: 181155. Diagn 1999; 19: 817826.

Copyright 2013 ISUOG. Published by John Wiley & Sons Ltd. Ultrasound Obstet Gynecol 2013; 42: 1533.
NIPT for aneuploidy 33

122. Wallerstein R, Yu M-T, Neu RL, Benn P, Bowen CL, Crandell 135. Spencer K, Liao AW, Skentou H, Cicero S, Nicolaides KH.
B, Disteche CM, Donaghue R, Harrison B, Hershey D, Higgins Screening for triploidy by fetal nuchal translucency and
RR, Jenkins LS, Jackson-Cook C, Keitges E, Khodr GS, Lin maternal serum free beta-hCG and PAPP-A at 1014 weeks
CC, Luthardt FW, Meisner L, Mengden G, Patil S, Rodriguez of gestation. Prenat Diagn 2000; 20: 495499.
M, Sciorra LJ, Shaffer LG, Stetten G, Van Dyke DL, Wang 136. Benn PA, Gainey A, Ingardia CJ, Rodis JF, Egan JF. Second
H, Williams F, Zaslav A-L, Hsu LYF. Common trisomy trimester maternal serum analytes in triploid pregnancies: Cor-
mosaicism diagnosed in amniocytes involving chromosomes relation with phenotype and sex chromosome complement.
13, 18, 20, and 21: karyotype-phenotype correlations. Prenat Prenat Diagn 2001; 21: 680686.
Diagn 2000; 20: 103122. 137. McFadden DE, Kalousek DK. Two different phenotypes of
123. Osborne CM, Hardisty E, Devers P, Kaiser-Rogers K, Hayden fetuses with chromosomal triploidy: correlation with parental
MA, Goodnight W, Vora NL. Discordant noninvasive prenatal origin of the extra haploid set. Am J Med Genet 1991; 38:
testing results in a patient subsequently diagnosed with 535538.
metastatic disease. Prenat Diagn 2013; 33: 609611. 138. Peters D, Chu T, Yatsenko SA, Hendrix N, Hogge WA, Surti
124. Lau TK, Jiang FM, Stevenson RJ, Lo TK, Chan LW, Chan MK, U, Bunce K, Dunkel M, Shaw P, Rajkovic A. Noninvasive
Lo PSS, Wang W, Zhang H-Y, Chen F, Choy KW. Secondary prenatal diagnosis of a fetal microdeletion syndrome. N Engl
findings from non-invasive prenatal testing for common fetal J Med 2011; 365: 18471848.
aneuploidies by whole genome sequencing as a clinical service. 139. Jensen TJ, Dzakula Z, Deciu C, van den Boom D, Ehrich
Prenat Diagn 2013; 33: 602608. M. Detection of microdeletion 22q11.2 in a fetus by next-
125. Yu SCY, Jiang P, Choy KW, Chan KCA, Won H-S, Leung generation sequencing of maternal plasma. Clin Chem 2012;
WC, Lau ET, Tang MHY, Leung TY, Lo YMD. Noninvasive 58: 11481151.
prenatal molecular karyotyping from maternal plasma. PloS 140. Le Scouarnec S, Gribble SM. Charecterising chromosome
One 2013; 8: e60968. rearragements: recent technical advances in molecular cyto-
126. Chen S, Lau TK, Zhang C, Xu C, Xu Z, Hu P, Xu J, Huang H, genetics. Heredity 2012; 108: 7585.
Pan L, Jiang F, Chen F, Pan X, Xie W, Liu P, Li X, Zhang L, Li 141. Devers PL, Cronister A, Ormond KE, Facio F, Brasington CK,
S, Li Y, Xu X, Wang W, Wang W, Jiang H, Zhang X. A method Flodman P. Noninvasive prenatal testing/noninvasive prenatal
for non-invasive detection of fetal large deletions/duplications diagnosis: the position of the National Society of Genetic
by low coverage massively parallel sequencing. Prenat Diagn Counselors. J Genet Couns 2013; 22: 291295.
2013; 33: 584590. 142. Noninvasive prenatal testing for fetal aneuploidy. Committee
127. Warburton D. De novo balanced chromosome rearrangements Opinion No. 545. American College of Obstetricians and
and extra marker chromosomes identified at prenatal Gynecologists. Obstet Gynecol 2012; 120: 15321534.
diagnosis: clinical significance and distribution of breakpoints. 143. Langlois S, Brock J-A. Current status in non-invasive prenatal
Am J Hum Genet 1991; 49: 9951013. detection of Down syndrome, trisomy 18, and trisomy 13
128. Schluth-Bolard C, Labalme A, Cordier MP, Till M, Nadeau using cell-free DNA in maternal plasma. J Obstet Gynaecol
G, Tevissen H, Lesca G, Boutry-Kryza N, Rossignol S, Can 2013; 35: 177181.
Rocas D, Dubruc E, Edery P, Sanlaville D. Breakpoint 144. Wilson KL, Czerwinski JL, Hoskovec JM, Noblin SJ, Sullivan
mapping by next generation sequencing reveals causative CM, Harbison A, Campion MW, Devary K, Devers P,
gene disruption in patients carrying apparently balanced Singletary CN. NSGC practice guideline: prenatal screening
chromosome rearrangements with intellectual deficiency and diagnostic testing options for chromosome aneuploidy. J
and/or congenital malformations. J Med Genet 2013; 50: Genet Couns 2013; 22: 415.
144150. 145. Noninvasive Prenatal Screening Work Group of the American
129. Talkowski ME, Ordulu Z, Pillalamarri V, Benson CB, College of Medical Genetics and Genomics, Gregg AR,
Blumenthal I, Connolly S, Hanscom C, Hussain N, Pereira Gross SJ, Best RG, Monaghan KG, Bajaj K, Skotko BG,
S, Picker J, Rosenfeld JA, Shaffer LG, Wilkins-Haug LE, Thompson BH, Watson MS. ACMG statement on noninvasive
Gusella JF, Morton CC. Clinical diagnosis by whole-genome prenatal screening for fetal aneuploidy. Genet Med 2013; 15:
sequencing of a prenatal sample. N Engl J Med 2012; 367: 395398.
22262232. 146. Benn P, Borell A, Chiu R, Cuckle H, Dugoff L, Faas B, Gross
130. Lo, YM, Chan KC, Sun H, Chen EZ, Jiang P, Lun FM, S, Johnson J, Maymon R, Norton M, Odibo A, Schielen P,
Zheng W, Leung TY, Lau TK, Cantor CR, Chiu RW. Spencer K, Huang T, Wright D, Yaron Y. Position Statement
Maternal plasma sequencing reveals the genome-wide genetic from the Aneuploidy Screening Committee on behalf of the
and mutational profile of the fetus. Sci Trans Med 2010; 2: Board of the International Society for Prenatal Diagnosis.
61ra91. Prenat Diagn 2013. DOI: 10.1002/pd4139. [Epub ahead of
131. Kitzman JO, Snyder MW, Ventura M, Lewis AP, Qiu print].
R, Simmons LE, Gammill HS, Rubens CE, Santillan DA, 147. Benn PA, Chapman A. Ethical challenges in providing
Murray JC, Tabor HK, Bamshad MJ, Eichler EE, Shendure J. nonoinvasive prenatal diagnosis. Curr Opin Obstet Gynecol
Noninvasive whole-genome sequencing of a human fetus. Sci 2010; 22: 128134.
Transl Med 2012; 4: 137ra76. 148. de Jong A, Dondorp WJ, de Die-Smulders CEM, Fritz SGM,
132. Fan HC, Gu W, Wang J, Blumenfeld YJ, El-Sayed YY, Quake de Wert GMWR. Noninvasive prenatal testing: ethical issues
SR. Non-invasive prenatal measurement of the fetal genome. explored. Europ J Hum Genet 2010; 18: 272277.
Nature 2012; 487: 320324. 149. Chachkin CJ. What potent blood: Non-invasive prenatal
133. Jacobs PA, Szulman AE, Funkhouser J, Matsuura JS, Wilson genetic diagnosis and the transformation of modern prenatal
CC. Human triploidy: relationship between parental origin care. Am J Law Med 2007; 33: 953.
of the additional haploid complement and development 150. Bianchi D. At-home fetal DNA gender testing. Obstet Gynecol
of partial hydatidiform mole. Ann Hum Genet 1982; 46: 2006; 107: 216219.
223231. 151. Ray T. Sequenom: Bloodied and Unbowed. Barrons. Sepember
134. Jauniaux E, Campbell S. Ultrasound assessment of placental 29, 2009. http: //online.barrons.com/article/SB12542362487
abnormalities. Am J Obstet Gynecol 1990; 163: 16501658. 5349477.html [Accessed 8 April 2013].

Copyright 2013 ISUOG. Published by John Wiley & Sons Ltd. Ultrasound Obstet Gynecol 2013; 42: 1533.

Das könnte Ihnen auch gefallen