Sie sind auf Seite 1von 35

Materials Science and Engineering R 74 (2013) 35–69

Contents lists available at SciVerse ScienceDirect

Materials Science and Engineering R


journal homepage: www.elsevier.com/locate/mser

Nanoparticles for improving cancer diagnosis


Hongmin Chen a,1, Zipeng Zhen a,1, Trever Todd a, Paul K. Chu b,*, Jin Xie a,**
a
Department of Chemistry and Bio-Imaging Research Center, University of Georgia, 1001Cedar Street, Athens, GA 30602, United States
b
Department of Physics & Materials Science, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China

A R T I C L E I N F O A B S T R A C T

Article history: Despite the progress in developing new therapeutic modalities, cancer remains one of the leading
Available online 2 April 2013 diseases causing human mortality. This is mainly attributed to the inability to diagnose tumors in their
early stage. By the time the tumor is confirmed, the cancer may have already metastasized, thereby
Keywords: making therapies challenging or even impossible. It is therefore crucial to develop new or to improve
Cancer diagnosis existing diagnostic tools to enable diagnosis of cancer in its early or even pre-syndrome stage. The
Biomarkers emergence of nanotechnology has provided such a possibility. Unique physical and physiochemical
Nanotechnology
properties allow nanoparticles to be utilized as tags with excellent sensitivity. When coupled with the
Nanomedicine
Bio-conjugation
appropriate targeting molecules, nanoparticle-based probes can interact with a biological system and
Surface modification sense biological changes on the molecular level with unprecedented accuracy. In the past several years,
Imaging much progress has been made in applying nanotechnology to clinical imaging and diagnostics, and
interdisciplinary efforts have made an impact on clinical cancer management. This article aims to review
the progress in this exciting area with emphases on the preparation and engineering techniques that
have been developed to assemble ‘‘smart’’ nanoprobes.
ß 2013 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 36
2. Tumor biomarkers . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 36
2.1. EGFR and HER2 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 36
2.2. VEGFR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37
2.3. Integrins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37
2.4. Folate receptor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37
2.5. MMPs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37
2.6. PSMA . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38
2.7. Phosphatidylserine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38
3. Engineering of nanoparticles for tumor detection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38
3.1. Magnetic nanoparticles. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38
3.1.1. Synthesis of magnetic nanoparticles. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38
3.1.2. Surface modification of magnetic nanoparticles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 41
3.1.3. Magnetic nanoparticles-based T1 contrast probes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 42
3.1.4. Magnetic nanoparticles for cancer detection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 43
3.2. Quantum dots . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45
3.2.1. Optical properties of quantum dots . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45
3.2.2. Synthesis of quantum dots . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 47
3.2.3. Surface modification of quantum dots . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 48
3.2.4. Toxicity of quantum dots . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 48

* Corresponding author. Tel.: +852 34427724; fax: +852 34420542.


** Corresponding author. Tel.: +1 706 542 1933; fax: +1 706 542 9454.
E-mail addresses: paul.chu@cityu.edu.hk (P.K. Chu), jinxie@uga.edu (J. Xie).
1
Contributed equally to this work.

0927-796X/$ – see front matter ß 2013 Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.mser.2013.03.001
36 H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69

3.2.5. Quantum dots for in vivo imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 50


3.2.6. Quantum dots for immunostaining . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 51
3.3. Upconversion nanoparticles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 52
3.3.1. Construction of upconversion nanoparticles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 52
3.3.2. Synthesis of upconversion nanoparticles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 53
3.3.3. Surface modification of upconversion nanoparticles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 55
3.3.4. Toxicity of upconversion nanoparticles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 55
3.3.5. Upconversion nanoparticles for tumor imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 55
3.4. Gold nanoparticles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 56
3.4.1. Optical properties of gold nanoparticles. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 56
3.4.2. Synthesis of gold nanoparticles. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 57
3.4.3. Surface modification of gold nanoparticles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59
3.4.4. Toxicity of gold nanoparticles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59
3.4.5. Gold nanoparticles for cancer diagnosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 59
4. Conclusion and perspectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 64
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 64
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 64

1. Introduction targeting molecules, followed by a systematic description of the


design and fabrication of magnetic nanoparticle-, quantum dot-,
Cancer accounts for approximately one-fourth of all deaths in upconversion nanoparticle-, and gold nanoparticle-based diagnos-
the United States [1]. Prognosis of cancer patients depends largely tic techniques, as well as their potential applications to cancer
on the time and accuracy of finding the primary and any dormant detection.
metastasis sites. Identification of these abnormal sites in an early
enough stage can significantly reduce the mortality rate. For 2. Tumor biomarkers
example, for breast cancer patients, the 5-year survival rate is 98%
if the tumor can be diagnosed in stages 0 and I and drops to 85% for Tumor biomarkers are substances found in blood, urine, stool, or
stage II and merely 20% for stage IV tumors [2]. Pancreatic cancer, tissues of patients with cancer. They are commonly proteins that are
on the other hand, is usually associated with a high mortality rate; elevated in either cancer cells or other cells in the body in response to
about 75% of pancreatic cancer patients die within 1 year after cancerous conditions. Measuring biomarker levels is one of the most
diagnosis [3]. This high death rate stems from the recessive traits common methods to detect, diagnose, and manage cancer.
exhibited by pancreatic cancer and a lack of an effective early Based on their location, protein biomarkers can be divided into
detection means. Needless to say, there is an urgent need for new three categories: intracellular, extracellular, and those that are on
tools that can detect cancer in the early or even pre-syndrome the cell membrane. In nanoparticle-enabled diagnosis, different
stage. Significant progress has been made recently thanks to biomarkers are targeted for different applications. In imaging, it is
advances in proteomics and genomics. These burgeoning techni- more common to target cell membrane protein biomarkers and
ques have identified various biomarkers whose abnormal regula- regarding in vitro diagnosis, it is more convenient to target proteins
tions are closely associated with tumorigenesis and progression that are soluble and shed into the circulation. Intracellular
[4–6]. In addition, new screening technologies have been proteins, on the other hand, are less targeted given the relatively
established allowing the identification of antibodies, peptide large size of nanoparticles and hence, their inability to pass
sequences, and nucleic acid aptamers with high affinity towards through the cell membrane. Due to a long and ever growing list of
a specific biomarker [7,8]. biomarkers, it is not possible to cover all of them in this article.
Nanotechnology, which is a multidisciplinary science involving Instead, we will focus on several biomarkers that have been well
chemistry, biochemistry, physics, and materials science, has found studied and are commonly targeted in validating nanoparticle-
uses in a wide spectrum of medicine-related applications. These based imaging and diagnosis techniques.
include nanoparticle-based imaging, drug delivery, biosensoring,
and hyperthermia, and in the past decade, a number of these 2.1. EGFR and HER2
nanoparticle-based techniques have been translated into clinical
applications [9]. Materials exhibit unique physical and biochemi- The epidermal growth factor receptor (EGFR) is a 170-kDa
cal properties when their dimensions are reduced to between protein expressed on the membrane of many types of epithelial
several to hundreds of nanometers. By taking advantage of these cancers [10,11]. EGFR has long been regarded as a tumor biomarker
unique characteristics, one can develop nanodevices that can sense because its mutation is often associated with autonomous cell
and monitor biological events with unprecedented efficiency and growth, inhibition of apoptosis, and metastases [12,13]. The
sensitivity. When coupled to the aforementioned targeting ligands, epidermal growth factor (EGF)–EGFR interaction is among the first
one can produce ‘‘smart’’ nanoprobes that can interact with a growth factor ligand–receptors studied [14]. EGFR has been found
biological system and sense changes on the molecular level. This to be a member of a receptor tyrosine kinase (TK) family, the
can take place in vivo, where nanoprobes are systematically human epidermal growth factor receptor (HER) family. The other
administrated, accumulated in tumors through ligand–biomarker three members are HER2 (ErbB2), HER3 (ErbB3), and HER4 (ErbB4)
interaction, and send out signals for sensitive diagnostic imaging. [15]. Hence, EGFR is also referred to as HER1 (ErbB1). Upon ligand
The nanoprobes are also useful to in vitro diagnosis and analysis of binding, the EGFR pairs with either itself or another HER member
biological samples such as saliva, blood, and urine. Both applica- such as HER2 form an active dimer. Dimerization stimulates
tions are invaluable and hold great promise in revolutionizing protein-tyrosine kinase and activates the intracellular protein
cancer management [9]. The objective of this article is to review kinase cascade. Monoclonal antibody-based inhibitors that target
the recent progress in this exciting area. We will begin by EGFR have been developed that take advantage of EGFR’s
discussing common tumor biomarkers and their corresponding important role in cancer development. The first example in this
H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69 37

category is cetuximab, a chimeric monoclonal antibody now used most tumor vasculature and for certain cancer types, also on tumor
clinically to treat patients with head and neck cancer as well as cell membranes. They are important players in tumor angiogenesis
metastatic colorectal cancer. With high avidity, cetuximab binds and metastasis [31]. In particular, integrin avb3 has been widely
with EGFR and sterically blocks its interaction with growth factors studied as a tumor biomarker [27] for ovarian [32], breast [33],
[16]. Other EGFR targeting antibody inhibitors including panitu- lung [34], and prostate [35] cancer detection. Integrin avb3 binds
mumab, zalutumumab, nimotuzumab, and matuzumab, are to arginine-glycine-aspartic acid (RGD), which is a three-amino-
currently in clinical trials. When coupled with nanoparticle-based acid sequence [36]. RGD dimers, tetramers, and octamers have also
probes, these antibodies can guide the probes to tumors to realize been studied for integrin avb3 targeting [37,38]. RGD and its
targeted imaging [17,18]. derivatives have been coupled to the surface of many types of
HER2 has also been intensively studied as a tumor biomarker; in nanoparticles for tumor imaging and site-specific drug delivery
particular, its implications for breast cancer. Deregulation of HER2 is [27]. It is worth mentioning that in many animal models, integrin
found in 20–25% of breast cancer patients, making it an important avb3 is upregulated on both tumor vasculature and tumor cells,
indicator of breast tumor malignancy [19,20]. HER2 usually and targeting occurs at both places when a small molecule-based
dimerizes with EGFR or HER3 [14], although it can also dimerize imaging probe is used. With regard to nanoparticle-based probes,
with itself at high expression levels [21]. Trastuzumab, an anti-HER2 especially those with a relatively large size, extravasation is a
monoclonal antibody, has been used clinically to treat breast cancers limiting factor and targeting typically occurs on the surface of
with HER2 overexpression [14]. It has been coupled with various endothelial cells. In addition to avb3, other members in the
kinds of nanoparticles and studied as a tumor imaging probe in a integrin family have been studied as tumor biomarkers. Examples
preclinical setting [22,23]. Pertuzumab, another HER2 targeting include avb6 in the detection and staging of ovarian carcinomas
monoclonal antibody, has recently received FDA approval for the [32] and integrin a5b1 in early stage lung cancer diagnosis [34].
treatment of HER2-positive metastatic breast cancer.
2.4. Folate receptor
2.2. VEGFR
Folic acid belongs to the vitamin B family. It is a required
The vascular endothelial growth factor (VEGF) is another family nutrient for all living cells and participates in the biosynthesis of
of growth factors that play a key role in angiogenesis, the process of nucleotide bases. Folic acid shows high affinity (KD < 1 nM) to
forming new blood vessels. Angiogenesis is critical to physiological folate receptors (FR) including FR-a and FR-b, both of which are
development and also is frequently implicated in pathological 38 kDa membrane-bound proteins [39]. FRs are expressed at low
processes, including tumor progression [24]. The VEGF family is levels in normal tissues and mostly restricted to epithelial cells
composed of seven members, VEGF-A, VEGF-B, VEGF-C, VEGF-D, [40]. On the other hand, FRs are found overexpressed in several
VEGF-E, VEGF-F, and placenta growth factor [25]. The most tumor types, particularly in ovarian and endometrial cancers [40].
important one is VEGF-A, which is a homodimeric, disulfide-bound Studies have shown that folic acid binds to tumor cells 20 times
glycoprotein existing in several isoforms with different numbers of more than normal epithelial cells or fibroblasts [41], and this
amino acid residues [26]. The most studied isoforms are VEGF165, tumor-associated disregulation makes FR an attractive tumor
VEGF121, and VEGF110. VEGF165 is implicated mostly in angiogene- biomarker.
sis and vascular permeability whereas VEGF121 and VEGF110 are Folic acid can be easily coupled to a peptide or a macromolecule
permeability factors [27]. VEGF-A acts on the vascular endotheli- with insignificant effects to the affinity to FR [42]. Various kinds of
um to stimulate endothelial cell mitogenesis and cell migration nanoparticles have been coupled to folic acid and studied for tumor
[27]. The receptors to VEGF-A are mainly Flt-1 (VEGFR-1) and Flk- imaging [40]. FR is expressed at a relatively high level in the
1/KDR (VEGFR-2). VEGFR-1 is involved in physiological and kidneys [40]. Imaging and therapy with folic acid–small molecule
developmental angiogenesis [25]. VEGFR-2 is a more important conjugates can be impeded by this feature due to high kidney
player in tumor progression and is critical to mitogenic, uptake. However, this issue is of minor concern with nanoparticle-
angiogenic, and permeability enhancement [28]. based probes due to their inaccessibility to these regions.
Overexpression of VEGF/VEGFR is observed from many tumor
types and is indicated as a poor prognostic marker. To inhibit the 2.5. MMPs
interaction, one strategy is to sequester VEGF with an antibody in
order to prevent VEGF from accessing VEGFR. Bevacizumab is a Matrix metalloproteinases (MMPs) are a family of zinc-
human monoclonal antibody against all isoforms of VEGF-A and the dependent endopeptidases. There are at least 20 members in this
first antigenic agent receiving FDA approval for cancer management. protease family and they are capable of degrading multiple
Similarly, ranibizumab, a monoclonal antibody fragment (Fab) of components of the extracellular matrix (ECM) [43]. Based on their
bevacizumab, is used clinically for mascular degeneration treat- substrate specificity, MMPs can be classified as collagenases,
ment. A second approach is to employ an antibody that targets gelatinases, stromelysins, and so on. Another subset is MMPs 14–
VEGFR. Upon binding, VEGFR is sterically blocked from interacting 17 which contain a transmembrane domain and are known as
with VEGF. Several anti-VEGFR-2 antibodies, including IMC-1C11, membrane-associated MMPs (MT-MMPs). In normal tissues,
IMC-1121B, and CDP791, have entered clinical development [29]. MMPs are expressed at a very low level [43], but when active
VEGF, anti-VEGF antibodies, and anti-VEGFR antibodies are all tissue remolding occurs, MMPs are rapidly transcribed, secreted,
possible targeting ligands for tumor imaging in vivo. and activated [44]. Such activation is usually associated with a
pathological process such as cancer. Indeed, high MMP expression
2.3. Integrins levels are found in many types of tumors and are expressed by the
host stromal cells and sometimes by tumor cells [45]. For instance,
Integrins constitute a family of cell adhesion molecules [30]. MMP-2 is highly expressed in breast cancers and an increased ratio
They are always present as heterodimers comprising of one a and of active MMP-2 is usually indicative of a higher malignancy
one b subunit. In mammals, 18 a-subunits and 8 b-subunits have [46,47]. MMPs 1, 3, 9, 14 are also key players in tumor cell growth,
been characterized. These subunits assemble into at least 24 metastasis, and angiogenesis [43].
integrin receptors that mediate the interactions between the cell These implications make MMPs an important class of tumor
and surrounding tissues [27]. Integrins are found upregulated on biomarkers. In the previous decade, efforts were made to prepare
38 H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69

MMP-targeted tumor imaging probes. Targeting can be achieved protein or even non-protein-based signatures that better represent
using a high affinity peptide. For instance, Zhu et al. found that a early stage cancer [63,64]. Therefore, it will not be surprising to see
peptide sequence, HWKHLHNTKTFL, had high MT1-MMP binding in the near future an increased number of reports pertaining to the
affinity and when coupled with a dye molecule, could efficiently development of nanoparticle-based probes/carriers that target
target tumors [48]. Koivunen et al. reported a short target peptide, these novel biomarkers.
CTTHWGFTLC, which could specifically bind MMP-2 and MMP-9
thus inhibiting the migration of human endothelium and cancer 3. Engineering of nanoparticles for tumor detection
cells [49]. An alternative approach is to take advantage of the
enzymatic function of MMPs and develop an MMP-activatable Nanoparticles can be used as probes in in vivo imaging,
imaging probe that displays amplified signals in the tumor region. biosensing, and immunostaining because nanoparticle-based
Gold nanoparticle-based MMP-activatable imaging probes, for probes offer many advantages. First of all, they deliver high
instance, have recently been reported by Lee et al. [50,51]. sensitivity. Many nanoscale materials show unique magnetic,
optical, or acoustic properties and they can be further imparted
2.6. PSMA with other types of imaging functionalities to result in probes with
multimodal abilities. Secondly, their size is appropriate. Most
The prostate-specific membrane antigen (PSMA) is a 100-kDa nanoparticles have a size that is above the threshold of renal
type II transmembrane glycoprotein composed of a short and amino- clearance (<7 nm) allowing them to remain in circulation for a
terminal cytoplasmic tail (19 amino acids), hydrophobic transmem- relatively long time before reaching the desired targets [65,66]. On
brane domain (24 amino acids), and large extracellular domain (707 the other hand, nanoparticles are also small enough to penetrate
amino acids) at the carboxy-terminus [52,53]. PSMA is expressed at many biological barriers such as endothelial barriers, cell mem-
a low or moderate level in most hyperplastic and benign prostates, branes, or even nuclear envelopes to efficiently interact with
but is highly upregulated in malignant tissues [54]. The increased biological systems on the molecular level [67]. Thirdly, they are
PSMA expression is related to tumor aggressiveness, and the highest multivalent. Multiple targeting ligands can be tethered onto one
PSMA levels are usually found from high-grade tumors, metastatic nanoparticle surface. This leads to a larger rate of receptor binding
lesions, and androgen-independent disease [55]. PSMA is also found and smaller rate of dissociation, both contributing to higher tumor
abundantly expressed on neovasculature of non-prostatic solid uptake and longer retention time [68,69], the so-called multivalency
tumors, including lung, colon, breast, renal, liver, and pancreatic effect. Moreover, it is possible to impart more than one type of
carcinomas as well as sarcomas and melanoma [55]. Antibodies targeting ligand [70,71]. Recent studies have confirmed improved
against PSMA such as 7E1, J591 and MDX-070 are now used tumor cell selectivity from such a dually targeting targeted approach
clinically for either imaging or therapeutic purposes [55]. In addition [72–74]. Fourthly, they offer combined therapy and diagnosis. In
to antibodies, other motifs have been discovered for PSMA targeting. addition to imaging functionalities, therapeutics can be loaded onto
Langer’s group, for instance, has been working on aptamer- [56,57] nanoplatforms. The resulting nanoparticles, so-called nano-ther-
and small molecule-based [54,55] ligands with high avidity towards anostics, have both imaging and therapeutic capabilities [75]. Nano-
PSMA. One of the technologies developed by the group, BIND-014, theranostics are appealing for potentially allowing therapy response
recently entered clinical trial for solid tumor therapy. BIND-014, a to be monitored in real-time by imaging methods, an emerging
polylactide (PLA)-based nanoparticle bearing a cytotoxic drug concept in modern personalized medicine [76].
docetaxel, uses S-2-[3-[5-amino-1-carboxypentyl]-ureido]-penta- In the following section, recent progress on nanoparticle-based
nedioic acid, a small molecule with high selectivity to PSMA for probes/sensors for cancer diagnosis is reviewed. Owing to limited
tumor targeting [54,55]. space, we will only describe magnetic nanoparticles, quantum
dots, gold nanoparticles, and upconversion nanoparticles. Readers
2.7. Phosphatidylserine who are interested in other materials such as carbon nanotubes
and graphene are referred to other excellent reviews [77–81].
Cancer cells are essentially cells that grow out of control and
lose the physiological balance of renewal and senescence. Most 3.1. Magnetic nanoparticles
therapeutic approaches work by inducing apoptosis to cancerous
cells. A widely acknowledged apoptosis biomarker is phosphati- Magnetic nanoparticles have been investigated extensively in
dylserine (PS), which in normal cells is fastened on the cytosolic the field of biomedicine and one of their important applications is
side of the cell membrane, but is flipped to the exterior side when contrast probes used in magnetic resonance imaging (MRI).
the cell undergoes apoptosis [58]. Measuring the PS level is a Magnetic nanoparticles create microscopic field gradients in a
proven means to monitor the cell apoptotic level and evaluate the strong magnetic field; causing shortening and diphase of
response of cancerous cells to a treatment [59]. High level of PS has longitudinal (T1) or transverse relaxation times (T2 and T2 ) of
also been found on tumor vascular endothelium [60]. Positive PS nearby nuclei, most commonly protons. This induces hyper- (for
staining was observed on 4–40% tumor vasculature but not on T1) or hypo-intensities (for T2 and T2 ) on MRI maps and by doing so,
normal tissues. Boasting high selectivity toward PS, Annexin-V is highlighting the areas that are concentrated with the particles.
the most commonly used apoptosis probe. Flow cytometry assays Magnetic nanoparticles, particularly iron oxide nanoparticles
with effusions from ovarian carcinoma patients showed good (IONPs), are used in liver imaging, lymph node imaging, and cell
correlation between Annexin-V level and stage, overall survival tracking. Recently, devices with extremely high sensitivity using
(p = 0.005), as well as progression-free survival (p = 0.013) [61]. magnetic nanoparticles as probes have been developed. In this
Annexin-V can be labeled with radio- or optical-tags for tumor section, we will describe the synthesis, surface modification, and
detection or monitoring of therapeutic response [62]. biological applications of magnetic nanoparticles with a focus on
In this section, we summarized some common biomarkers IONPs which are by far the most common magnetic nanoparticles.
targeted for cancer detection. However, it is still debatable
whether some of these proteins can be called early detection 3.1.1. Synthesis of magnetic nanoparticles
biomarkers. Nonetheless, they are the most commonly targeted
from the perspective of nanoparticle-based imaging/delivery 3.1.1.1. Iron oxide nanoparticles made by co-precipitation. Bulk iron
technology. Active research is being conducted to identify other oxide, either magnetite or hematite, is a ferromagnetic material
H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69 39

Table 1
Summary of magnetic nanoparticles of various types.

Particle Synthesis Surface coating r1 (mM1 s1) r2 (mM1 s1)

Fe3O4 Co-precipitation Cross-linked aminated 22.4 [100] 76.7 [100]


dextran [100]
Zn0.4Mn0.6Fe2O4 Thermal decomposition of metal DMSA N/A 860 [113]
chloride (MCl2, M = Zn2+, Mn2+,
and Fe2+) and Fe(acac)3
Fe Thermal decomposition of Fe(CO)5 PEGylated dopamine N/A 220 [118]
MnO Thermal decomposition of Mn–oleate Silica 0.99 [146] N/A
complex
Mn NMOFs [152] Reverse-phase microemulsion using PVP/silica 4.6 [152] 141.2 [152]
MnCl2 and [NMeH3]2(BDC)
Gd-apoferritin Loading GdHPDO3A into apoferritins Apoferritin 80  5 [155] N/A
Metallofullerene Arc-burning of Gd2O3 and graphite Carbon 207 [159] N/A

with a strong magnetic moment. In the ambient environment, 3.1.1.2. Magnetic nanoparticles made by thermal decomposi-
magnetite and hematite have magnetic moments of 90 and tion. Thermal decomposition has become a popular synthetic
76 emu g1, respectively. However, both Fe3O4 and Fe2O3 nano- approach in IONP preparation (Table 1) [101,102]. Instead of using
particles show zero remnant magnetism in the ambient environ- an aqueous solution as the reaction medium, thermal decomposi-
ment. This is because on such a small scales, thermal energy is tion takes place in an organic solvent with a high-boiling point. In a
sufficient to overcome the anisotropy energy of each small magnet typical synthesis, organometallic compounds such as Fe(CO)5,
thereby causing random fluctuations in the magnetization and an Fe(acac)3, and Fe–oleate are heated to a high temperature to induce
overall zero magnetic moment [82]. When an external magnetic nucleation and particle growth, as illustrated in Fig. 1 [102–104].
field is applied, the nanoparticle magnets are easily magnetized Surfactants, such as oleic acid (OA) and oleylamine (OAm), are also
and re-aligned to reach a saturation magnetic moment. However, essential to the process because they dynamically interact with the
when the magnetic field is turned off, the magnets are again growing particle surface and determine the monodispersity and
randomized and the magnetism drops to zero. This phenomenon, morphology of the products. In general, thermal decomposition
termed superparamagnetism, is also observed from other types provides better product quality control over co-precipitation
magnetic nanoparticles [82]. One of the most important reasons methods. By changing/adjusting the surfactant, precursor, precur-
that IONPs are commonly used is the biocompatibility of iron sor concentration, surfactant/precursor ratio, heating rate, and
oxide. Iron oxide has low toxicity and can be degraded by a reaction temperature [105–108], IONPs with single crystallinity,
biological system to become part of the body’s iron stores. Another narrow size distribution, and controllable size can be produced. For
reason is the well-established surface chemistry. In the past instance, Woo et al. prepared monodispersed IONPs by thermal
decade, many surface engineering techniques have been devel- decomposition of Fe(CO)5 in a solution of octyl ether and oleic acid
oped that can modify IONPs, making them stable in physiological [109]. Sun et al. heated Fe(acac)3 with 1,2-hexadecanediol in the
environments and easy to be conjugated with various bio- presence of OA and OAm to make monodispersed IONPs with size
molecular species. of 4–20 nm (Fig. 1a) [110]. An increase in size can be achieved
Traditionally, IONPs are synthesized by co-precipitation using a seed-mediated process using small particles as the seeds.
(Table 1) [83]. In brief, Fe(II) and Fe(III) precursors are dissolved
in an aqueous solution into which an alkaline medium is added to
induce particle formation. The size, shape, and composition of the
magnetic nanoparticles depend on the type of the salts used (e.g.
chlorides, sulfates, nitrates), Fe2+/Fe3+ ratio, reaction temperature,
pH value, and ionic strength [83–85]. This synthetic approach is
straightforward and has a high throughput, but the resulting
particles exhibit low magnetism, typically in the range of 30–
50 emu g1 [86]. Bare IONPs are not stable and tend to aggregate.
To improve their stability, it is common to include stabilizing
agents in the reaction. They are usually polymers such as
polyvinylpyrrolidone (PVP), dendrimers, polyaniline, and dextrans
[87–93] which lower the surface energy and increase electrostatic
repulsion, resulting in improved colloidal stability. Most IONPs
used clinically, including Feridex, Combidex, Resovist, and
Ferumoxytol, are made by the co-precipitation method using
dextran and its derivatives as the coating materials [94,95].
A sugar coating does not favor direct bio-molecule coupling,
and to make the particles conjugation-friendly, a post-synthesis
surface engineering step is usually required. A common approach
is to treat the dextran-coated particles with epichlorohydrin and
ammonia. This treatment produces a more rigid, cross-linked
coating [96] and more importantly, introduces multiple amine
groups on the particle surface that can be used for bio-conjugation
Fig. 1. Schematic illustration of the synthesis of monodisperse magnetic
[97–99]. The treated nanoparticles, termed cross-linked iron oxide, nanoparticles using different precursors: (a) metal acetylacetonates, (b) metal–
can be coupled to antibodies, peptides, aptamers, as well as drug oleate.
molecules for the purpose of tumor imaging and therapy [100]. Reprinted with permission from refs. [110] and [147].
40 H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69

Fig. 2. TEM images of IONPs synthesized by a thermal decomposition route with precise control of their diameters. (a) 6 nm, (b) 7 nm, (c) 8 nm, (d) 9 nm, (e) 10 nm, (f) 11 nm,
(g) 12 nm, (h) 13 nm.
Reproduced with permission from [105].

Park et al. reported that monodispersed magnetic IONPs with a [110]. Other transition metals can be incorporated into the matrix
continuous size between 6 and 13 nm could be synthesized by a as well. Cheon’s group reported the preparation of ZnxMn1  xFe2O4
procedure similar to seed-mediated growth (Fig. 2) [105]. In this nanoparticles. Zn2+ was introduced into the tetrahedral sites, and
process, the metal–surfactant complexes generated in situ are because Zn2+ resides mainly in the Td sites rather than Oh sites,
thermally decomposed to form nanoparticles. A prominent size magnetism is significantly increased in comparison with normal
effect is observed from these pyrolysis-yielded nanoparticles. spinel metal ferrite nanoparticles [113]. In particular,
Generally, particles with a larger diameter possess larger magnetic Zn0.4Mn0.6Fe2O4 nanoparticles with a size of 15 nm have an
moments due to spin disorder and canting on the particle surface extremely high magnetic moment of 175 emu g1 and r2 value of
[111]. For instance, 4, 6, 9, and 12 nm IONPs have magnetic 860 mM1 s1.
moments of 25, 43, 80, and 102 emu g1, respectively (Fig. 3) [112]. Synthesis of metallic nanoparticles such as Fe and Co have been
The difference in magnetism dramatically affects their ability as T2 reported [114]. Bulk Fe and Co have magnetic moments of 218 and
contrast agents in MRI. 162 emu g1, respectively, both of which are significantly larger
The thermal decomposition method can be extended to prepare than iron oxide’s. Metallic Fe nanoparticles have been synthesized
other magnetic nanoparticles with even larger magnetic moments. by thermal decomposition of Fe(CO)5 in the presence of stabilizing
For example, MFe2O4 (M = Co, Mn, Ni) nanoparticles can be surfactants such as polyisobutene and decalin [115,116]. By
prepared by simply replacing one-third of the Fe(acac)3 precursor adjusting the ratio of Fe(CO)5 to surfactants, Fe nanoparticles with
with Co(acac)2/Mn(acac)2/Ni(acac)2 (Fig. 1). The nanoparticles a size of 2–10 nm and narrow size distribution (polydispersity of
produced possess different magnetic properties. MnFe2O4 nano- approximately 10%) have been fabricated. However, these Fe
particles with a size of 15 nm possess a magnetic moment of particles are not stable in the ambient environment. They are
74 emu g1 which is higher than that of IONPs with the same size easily oxidized, leading to reduced magnetic susceptibility. Sun’s
group reported a simple one-pot synthetic approach to make air-
stable Fe nanoparticles [117,118]. Briefly, Fe(CO)5 was decom-
posed in the presence of hexadecylammonium chloride, an
oxidizing agent. This treatment led to the formation of a
crystalline Fe3O4 shell that protected the Fe core from continued
oxidation [119].
Synthesis of Co nanoparticles has been reported by a number of
groups. Alivisatos and co-workers reported the synthesis of cobalt
nanodisks by thermal decomposition of Co2(CO)8 [120,121]. Co
nanoparticles also suffer from the air-sensitivity problem. Bonne-
mann et al. described the synthesis of air-stable monodisperse
colloidal Co nanoparticles by thermolysis of Co2(CO)8 in the
presence of aluminum alkyl compounds [122]. By varying the alkyl
chain length of the organoaluminum compounds, the size of the Co
particles can be tuned in the range of 3–11 nm.
Nanoscale particles of many other magnetic materials have
been made. Hou’s group employed a facile wet-chemical route to
synthesize Hägg iron carbide (Fe5C2) nanoparticles, in which
Fig. 3. Nanoscale size effect of IONPs on MR signals. (a) TEM images of nanoparticles bromide was found to be the key to inducing the conversion of
of 4, 6, 9, and 12 nm. (b) Size-dependent T2-weighted MR images of IONPs at 1.5 T.
Fe(CO)5 to Fe5C2 (Fig. 4) [123]. The Fe5C2 nanoparticles were
(c) Size-dependent changes in color-coded MR images based on T2 values.(For
interpretation of the references to color in this figure legend, the reader is referred 20 nm in diameter (Fig. 4a) and had a core–shell structure
to the web version of the article.) (Fig. 4b). Dai’s group adopted a chemical vapor deposition method
Reprinted with permission from [112]. to synthesize FeCo/single-graphitic-shell nanocrystals [124] and
H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69 41

Fig. 4. Fe5C2 nanopaticles synthesized by thermal decomposition. (a) TEM and (b) HRTEM images of 20 nm Fe5C2 nanopaticles. (c) Schematic illustration of the formation
mechanism of Fe5C2 nanopaticles.
Reprinted with permission from [123].

Kim et al. reported the synthesis of FePt nanoparticles via thermal potential applications of these PEGylated nanoconjugates [132].
decomposition of Pt(acac)2 and Fe(CO)5 in benzyl ether [125]. Xie et al. reported a two-step surface modification approach for
In summary, nanoparticles of iron oxide and other types of IONPs. In this process, the particles were first ligand-exchanged
magnetic materials have been produced mainly by co-precipita- with dopamine and subsequently adsorbed with a layer of human
tion and thermal decomposition. Co-precipitation is straightfor- serum albumin (HSA) as shown in Fig. 5a [133]. Another
ward, repeatable, and has a high throughput. However, the commonly used anchor compound is dimercaptosuccinic acid
particles produced have a relatively broad size distribution and (DMSA), which is able to chelate with surface iron via the two
suboptimal magnetism. Thermal decomposition allows better carboxylic groups. These particles can be further stabilized by
control of the particle size, shape, and composition, but requires disulfide cross-linkages formed among the ligands [23]. Bio-
stricter reaction conditions (high temperature, oxygen and water molecules can be coupled onto the particle surface by utilizing the
free, etc.). Another disadvantage of thermal decomposition is that surface thiol groups. Herceptin, for instance, has been coupled
the synthesized nanoparticles are coated with a thick alkyl coating onto DMSA modified particles [23].
and not water soluble. Subsequent surface modification needs to It is common to observe particle aggregation during ligand
be performed before the particles can be used in biological exchange and phase transition. This is attributed to the dramatic
applications. polarity change on the particle surface which makes the particles

3.1.2. Surface modification of magnetic nanoparticles


The two most commonly used surface modification approaches
are ligand exchange and ligand addition. The former refers to the
strategy of introducing a ligand with higher affinity to the particle
surface. The latter involves the use of an amphiphilic substance
that interacts with the existing hydrophobic alkyl layer via
hydrophobic–hydrophobic interactions while leaving the hydro-
philic section outward to suspend the particles, as shown in Fig. 5.

3.1.2.1. Ligand exchange. Polymers such as poly(pyrrole), poly(-


aniline), poly(alkylcyanoacrylates), poly(methylidene malonate),
poly(lactic acid), poly (glycolic acid), and poly(e-caprolactone)
[126–130] have been studied as ligands to replace the original
alkyl coating layer on the surface of magnetic nanoparticles. These
polymers contain amine, carboxylate, phosphate, or hydroxyl end
groups, and due to their multidentate nature, they can chemically
adsorb onto the nanoparticle surface to form a protective coating.
Alternatively, small molecule-based anchors that chelate with
surface metals can be used as a replacing ligand. For IONPs, one
good example is dopamine and its derivatives. This bidentate
enediol compound can chelate with the under-coordinated
surface Fe and convert it to a bulk-like lattice structure with an
octahedral geometry [131]. This results in strong binding between
the dopamine moiety and surface of the IONPs. This method was
first reported by Xu et al. who successfully anchored nickel
nitrilotriacetic acid (Ni-NTA) on the iron oxide shell of Co or
SmCo5.2 nanoparticles using dopamine as the anchor [131]. Fig. 5. Representative IONPs surface modification methods. (a) Ligand exchange; (b)
Afterwards, Sun’s group reported surface-modifications of IONPs ligand addition; (c) silica coating.
with PEGylated dopamine and evaluated the stability and Reproduced by permission from refs. [133], [138], and [143].
42 H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69

more susceptible to clustering. Moreover, the interaction between (CTAB) as a coating precursor [145]. CTAB works as a surfactant to
the new ligand and particle surface is usually pH sensitive and can transfer IONPs to the aqueous phase where TEOS condenses to form
be labile in a harsh environment, such as that in endosomes/ a silica coating on the IONPs. CTAB is then removed by refluxing
lysosomes. Nonetheless, ligand exchange remains a popular gently in a hydrochloric acid/ethanol solution at 60 8C for 3 h. This
surface modification strategy in the field. produces 2–6 nm pores on the silica coating surface, into which
small molecules can be easily encapsulated via physical interactions
3.1.2.2. Ligand addition. Unlike ligand exchange, the native hydro- [146].
phobic coatings are retained in ligand addition. The new ligands
are usually amphiphilic materials such as poloxamine, poly(lactic- 3.1.3. Magnetic nanoparticles-based T1 contrast probes
co-glycolic acid), poly(maleic anhydride alt-1-tetradecene) The aforementioned magnetic nanoparticles are made of
[134,135], and phospholipids [136]. The hydrophobic segments, ferromagnetic materials. These materials can significantly reduce
mostly hydrocarbons, intercalate/interact with the alkyl tails of the the transverse relaxation time of protons and are primarily used as
native ligands. The hydrophilic segments, usually containing PEG T 2 =T2 contrast agents. On the contrary, nanoparticles made of
and/or multiple charged chemical groups, are pointed outward to paramagnetic materials are used in T1 contrast probes. One
improve water solubility. Ligand addition and phase transition are example is MnO nanoparticles. MnO nanoparticles can be made by
usually achieved by film hydration as shown in Fig. 5b [137,138]. pyrolyzing Mn–oleate in a high-boiling point solvent [22,147]. By
Using phospholipids as an example, the phospholipids and adjusting the heating time and temperature, monodispersed MnO
nanocrystals are first mixed in chloroform to form a homogeneous nanoparticles with size variations of <5% can be synthesized.
solution. The solvent is evaporated with a rotary evaporator to Like IONPs, the synthesized MnO nanoparticles are coated with
produce a film with uniformly distributed components. After- a layer of OA/OAm which isolates the particle surface from the
wards, the film is hydrated and the components self-assemble into surroundings [148]. To become water soluble, both the ligand
water-dispersible nanoparticles. This dispersing step usually addition and ligand exchange routes have been investigated. A
requires energy input from heating and sonication [136]. significant difference in MRI contrast ability has been found from
The coating thickness impacts the relaxation times of the particles surface modified by different approaches. For example, an
magnetic nanoparticles. LaConte et al. used PEGylated phospho- r1 relaxivity of 0.37 mM1 s1 is observed from 20 nm MnO
lipids of PEG chains with various lengths to modify IONPs. As the nanoparticles coated with PEGylated 1,2-distearoyl-sn-glycero-3-
coating thickness increased, R2 decreased and R1 increased [139] phosphoethanolamine (DSPE) (DSPE-PEG) [149]. On the other
due to physical exclusion of protons from the magnetic field and hand, the same size MnO nanoparticles modified by a dopamine-
residence time of protons in the coating zone. It is worth plus-human serum albumin (HSA) approach showed a r1 of
mentioning that PEGylation is a common method in nanoparticle 1.97 mM1 s1 [150]. The difference is due to the dependence of T1
surface modification [132,136–139]. In the blood, nanoparticles reduction on the probe–water interaction. When a DSPE-PEG
can be easily adsorbed with a layer of opsonin proteins and as a coating is used, a hydrophobic inner layer remains on MnO surface
result, recognized by the immune system and removed from that isolates the cores from the surroundings. The dopamine-HSA
circulation. PEGylation has proven to be able to stabilize coating, on the other hand, is more hydrophilic and compact,
nanoparticles, inhibit serum protein absorption, and extend leading to more efficient water interactions on the MnO surface.
particle circulation half-lives [136]. Mesoporous silica-coated hollow manganese oxide (HMnO@m-
Small molecules, such as drug molecules, can be introduced SiO2) nanoparticles have also been studied as a T1 MRI contrast
along with IONPs into polymer/liposome matrices. For instance, agent. The mesoporous shell enables optimal access of water
Hu et al. fabricated core/shell particles containing an IONP core and molecules to the magnetic core and an r1 of 0.99 mM1 s1 is
a thermally responsive polymer shell into which hydrophobic observed at 11.7 T. These nanoparticles have been used to label
drugs were encapsulated [140]. adipose-derived mesenchymal stem cells (MSCs) with electropo-
ration. Intracranial grafting of these HMnO@mSiO2-labeled MSCs
3.1.2.3. Silica coating. In addition to ligand addition and exchange enables serial MR monitoring of the cell transplants over a period
with organic compounds, surface modification can also be of 14 days (Fig. 6) [146].
performed using thick and dense inorganic materials. Compared Paramagnetic nanoparticles made from other materials and
to organic coatings, the rigid structure of an inorganic coating can methods have also been studied. For instance, Lin’s group has
better prevent the particle cores from contacting each other and reported the preparation of paramagnetic nanoscale metal-organic
the environment. The most commonly used inorganic coating is frameworks (NMOFs) [151,152], a new class of isoreticular
silica. For IONPs made by thermal decomposition, the coating materials built from linking metal ions with well-defined
usually occurs in a reverse microemulsion containing hydrophobic coordination geometry using organic bridging ligands. Paramag-
IONPs and tetraethylorthosilicate (TEOS) [141,142]. TEOS hydro- netic transition cations are built into these NMOFs and their
lyzes and condenses onto the particles to form a silica shell. Other potential as T1 contrast probes has been evaluated [151,152]. For
silanes, such as N-(6-aminohexyl)-aminopropyltrimethoxysilane instance, Mn-containing NMOFs with controllable morphologies
(AHAPS) and 3-(triethoxysilyl)propylsuccinic anhydride) (SSA), were synthesized using reverse-phase microemulsion techniques
can be mixed with TEOS as the precursors. The resulting at room temperature and a surfactant-assisted procedure at 120 8C
nanoparticles have surface amine or thiol groups suitable for with microwave heating [152]. The resulting nanoparticles
further bio-conjugation. Alternatively, functional molecules can be showed a modest r1 of 5–8 mM1 s1 on a per Mn basis. Similarly,
pre-conjugated with silanes and encapsulated into the silica Gd-containing NMOFs were prepared and the nanoparticles had an
matrix. Dye molecules such as rhodamine B isothiocyanate (RITC), r1 relaxivity of 1.5 mM1 s1[151]. Luminescent lanthanide ions
for instance, can first be coupled with 3-aminopropyltriethox- such as Eu and Tb can also be doped into the NMOF matrices to
ysilane (APTES) and co-condensed to form a fluorescent silica produce MR and optical dual functional imaging probes [153]. For
coating, as shown in Fig. 5c [143]. To further improve the both Mn- and Gd-containing NMOFs, the nanoparticles can be
physiological stability, PEGylation is usually added to the silica coated with silica and coupled with functional molecules [154]. In
coating [144]. one study, a cyclic RGD peptide and a dye molecule were both
Functionality loading can also be achieved by using a mesopor- covalently coupled onto silica-coated, Mn-containing NMOFs
ous silica coating. This is formed by including cetrimonium bromide [152].
H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69 43

Fig. 6. In vivo MRI tracking of transplanted adipose-derived MSCs bearing HMnO@mSiO2. (a) TEM and (b) HRTEM images of HMnO@mSiO2 nanoparticles. (c) T1 map of
HMnO@mSiO2 nanoparticles of different concentrations at 11.7 T. (d) Plot of R1 versus Mn concentration. In vivo MRI results when (e) unlabeled MSCs and (f) HMnO@mSiO2-
labeled MSCs were injected into small animal models.
Reprinted with permission from [146].

Protein-based nanoparticles can be encapsulated with Gd and along with a NIRF dye IRDye800, were covalently coupled onto the
used as T1 contrast agents. In particular, Gd has been encapsulated particle surface. When the materials were tested on a U87MG
into apoferritin nanocages and the conjugates show a T1 relaxivity subcutaneous model strong hypointensities were observed from the
of about 80  5 mM1 s1 at 20 MHz and 298 K. This value is almost tumor areas and peaked at 4 h post-injection (p.i.) (Fig. 7b).
20 times higher than those of commercial Gd agents [155,156]. Postmortal immunohistological studies revealed nanoparticles in
Yin et al. described a metallofullerene nanoparticle-based MRI both the tumor endothelial cells and tumor cells (Fig. 7c) [160], both
contrast agent [157,158]. These nanoparticles are synthesized by of which overexpressed integrin avb3 [163,164]. Similar targeting
arc-burning of composite rods consisting of Gd2O3 and graphite in profiles were observed from BT-20 (human breast cancer) tumor
a He atmosphere. The Gd cations are encapsulated inside the models with RGD–IONP conjugates [108,109].
fullerene of the nanostructures. Using Gd3N@C80(OH)26(CH2CH2- Other peptide-based targeting molecules have been studied.
COOM)16 (M = Na or H) nanoparticles as an example, r1 is Chlorotoxin (CTX) was conjugated with IONPs and studied on 9L (rat
207 mM1 s1 and 50 times higher than that of commonly used gliosarcoma) xenograft models for tumor imaging [165]. The
Gd3+ complexes, including Omniscan and Magnevist [159]. nanoparticles were found to induce a maximum R2 change of
16 s1 in tumors after 12 h. It was subsequently shown in a
3.1.4. Magnetic nanoparticles for cancer detection transgenic mouse model that the CTX-IONPs could cross the blood–
brain barrier (BBB) and specifically target brain tumors [166] without
3.1.4.1. Magnetic nanoparticle-based probes for tumor imagi- damaging the BBB. Although the exact mechanism is not clear, the
ng. Magnetic nanoparticles made of ferromagnetic materials have BBB penetration is likely associated with CTX on the particle surface.
been coupled with targeting motifs and studied for tumor homing Moreover, A54, CREKA, bombesin, F3, tumorstatin, and LHRH have
and imaging. For instance, RGD has been coupled onto silica [37], been studied as targeting motifs for IONP-based imaging [167–170].
triblock-copolymers [160], polyaspartic acid [161], and dextran Magnetic nanoparticles have been coupled with antibodies
[69,162] coated IONPs and evaluated as MRI contrast probes. [23]. Lee et al. coupled Herceptin with MnFe2O4 nanoparticles and
Coupling of multiple RGD on the particle surface results in a evaluated the MR imaging capability on breast and ovarian tumor
significant increase in the affinity to integrin avb3 due to the models [171]. Hadjipanayis et al. coupled IONPs with a purified
multivalent effect [163]. Taking polyaspartic acid (PASP)-coated antibody that selectively binds to the EGFR deletion mutant
IONPs as an example, the IC50 values of the RGD conjugates are EGFRvIIIAb present in human glioblastoma multiforme cells [172].
34  5 nM, compared to 250  60 nM of free RGD [68]. Chen et al. In vivo MRI revealed hypointensities in or adjacent to intracranial
used a PEGylated triblock copolymer consisting of a polybutylacrylate human xenograft tumors and the IONPs continued producing
segment, polyethylacrylate segment, polymethacrylic acid segment, contrast days later. In both studies, the antibodies had therapeutic
and a hydrophobic hydrocarbon side chain to coat IONPs prepared by abilities and the conjugates were therefore examples of nano-
thermal decomposition (Fig. 7a). A cyclic RGD peptide, c(RGDyK), theranostics.
44 H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69

Fig. 7. Triblock copolymer coated IONPs (TPIONPs) conjugated with RGD peptides and dye molecules for tumor targeting. (a) Schematic illustration of the optical/MR dual-
modal imaging probes. (b) MR imaging of U87MG tumor-bearing mice injected with RGD–TPIONPs or TPIONPs. (c) Prussian blue and CD31/murine b3/F4/80 double staining
with the tumor sections.(For interpretation of the references to color in this figure legend, the reader is referred to the web version of the article.)
Reprinted with permission from ref. [160].

Magnetic nanoparticle–aptamer conjugates have also been Paramagnetic nanoparticle-based T1 contrast probes have been
reported. Hwang et al. conjugated the AS1411 aptamer (MF- studied in tumor imaging. Na et al. coupled MnO nanoparticles
AS1411) onto silica-coated CoFe2O4 nanoparticles [173]. MF- with Herceptin [22]. The conjugates were able to locate breast
AS1411 is a sequence that targets nucleolin, a cellular membrane cancer metastasis in the brain, while those not conjugated with
protein highly expressed on the surface of many cancer types. The Herceptin could not. Geninatti Crich et al. studied Gd-loaded
nanoparticle conjugates labeled with 67Ga were evaluated in vivo apoferritin nanoparticles in tumor angiogenesis imaging using a
on a C6 glioma xenograft model. Accumulation in the tumor areas two-step approach [155]. Firstly, a biotinylated derivative of C3d
was observed by both MRI and single-photon emission computed peptide was injected. With high affinity, C3d bound to the neural
tomography (SPECT) imaging. On the other hand, little tumor cell adhesion molecule (NCAM) that is overexpressed in tumor
accumulation was found when the AS1411 mutant (where core endothelial cell linings of human carcinomas. Secondly, biotiny-
nucleotides G in AS1411 aptamer were substituted with C) was lated Gd-loaded apoferritin (Gd-Apo-Bio) and streptavidin were
coupled onto IONPs. injected. With streptavidin as a cross-linker, Gd-Apo-Bio migrated
H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69 45

to endothelial cells to induce hyperintensities in the T1-weighted and immobilized on the sensor via the biotin–streptavidin
MRI maps. However, in vivo imaging of tumors with nanoparticle- interaction. The attached nanoparticles alter the voltage in the
based T1 contrast agents is relatively scarce due to the relatively magnetoresistive sensor yielding quantitative signals. Initial
less prominent signal change and concerns with particle toxicity. studies show that multiple protein-based biomarkers can be
detected at fM concentration levels with a dynamic range of more
3.1.4.2. Magnetic nanoparticle-based biosensors. Detection and than four orders of magnitude [180]. The detection limit can be
accurate measurement of biomarkers in biological samples are further improved by adopting an amplification step to lower the
invaluable to healthcare. Devices employing magnetic nanostruc- detection limit down to 50 aM, which is 1000 times lower than that
tures have shown marked progress toward this goal. The greatest of ELISA [181]. Gaster et al. used this system to monitor the
advantage of magnetic nanoparticle-based biosensors over tradi- dynamic changes of CEA, VEGF, and EpCAM in a human colorectal
tional diagnostic methods is the near absence of biological cancer xenograft mouse model [181]. A strong correlation between
background interference. This enables the detection schemes to the CEA concentrations and tumor growth was observed. On the
deliver high sensitivity on biological specimens. other hand, ELISA failed to detect the CEA level change as the
Weissleder’s group has recently developed a technology called concentrations were below the detection limit. The GMR-based
diagnostic magnetic resonance (DMR) which exploits magnetic biosensor technology has been improved and a recent publication
nanoparticles as proximity sensors to modulate the spin–spin shows that it can be scaled to over 100,000 sensors per cm2 to
relaxation time of water molecules surrounding the molecularly- measure the binding kinetics of various proteins with high spatial
targeted nanoparticles [174]. When a few magnetic nanoparticles and temporal resolution, as shown in Fig. 8 [182]. Moreover, the
bind to the intended molecular target through affinity ligands, the detection limit has been improved to as low as 20 zeptomoles of
particles form soluble nanoscale clusters causing a decrease in T2. the solute.
Instead of performing analysis on traditional and expensive NMR
equipment, the group has developed miniaturized easy-to-use 3.2. Quantum dots
systems that provide accurate and rapid readouts. With antibody-
conjugated IONPs, they are able to profile mammalian cells using 3.2.1. Optical properties of quantum dots
the expressions of a number of biomarkers such as Her2/neu and Optical fluorescence imaging is used in tumor diagnosis,
EGFR [175]. They have also demonstrated the potential of using especially in preclinical circumstances. Fluorescence microscopy
DMR for multiplexed screening and eight biomarkers can be has advanced the fields of immunology and cellular biology. The
simultaneously analyzed on a 2  4 microcoil array. In the early recent advent of small animal whole-body fluorescence imaging
studies, dextran-coated IONPs with a suboptimal r2 (62 mM1 s1) systems offers a sensitive and cost-effective tool in preclinical
were used. They later employed MnFe2O4 nanoparticles with a cancer studies and has found widespread use in biomarker
much higher r2 (420 mM1 s1) as the probes, and this significantly discovery and drug development. An intrinsic drawback of
improved the sensitivity. As few as two cancer cells in 1-mL fluorescence imaging is limited tissue penetration. On account
volumes of unprocessed fine-needle aspirates of tumors can be of scattering and absorption, light transmission is severely
detected and analyzed for biomarker expression [176]. This attenuated when traveling both in and out of tissues. This leads
sensitivity is comparable to that of clinical methods such as flow to a shallow penetration depth of typically less than 1 cm [183].
cytometry and Western blot, but is performed in a much shorter Using fluorophores with absorption and emission in the near-
time (<15 min) [176]. infrared region can alleviate the energy loss to a certain extent, but
Conjugation chemistry also impacts the sensitivity of DMR. in general, in vivo fluorescence imaging has been limited to small
Instead of using the traditional bio-conjugation techniques, a new animal models with targets close to the skin. Notably, the limited
conjugation technology between 1,2,4,5-tetrazine (Tz) and trans- penetration does not exclude the clinical use of fluorescence
cyclooctene (TCO) has been evaluated [177,178]. Because of the imaging. Fluorescence imaging has found extensive uses in
small size of the coupling reagents, a higher multiplicity of immunostaining for analyzing patient samples. Moreover, exten-
antibodies per nanoparticle is observed [179]. This leads to a sive research is being conducted to develop fluorescence-based
higher number of particles binding to cells and in turn, improves imaging technologies at endoscopic and intraoperative settings,
the detection sensitivity [179]. A clinical study has recently been where tissue penetration is less a concern.
performed using this method [176]. The DMR system was used to Organic dye molecules were overwhelmingly used in fluores-
analyze cells obtained by fine-needle aspirates from suspected cence imaging in the past, but recently, inorganic nanoparticle-
lesions in 50 patients. With a four-protein signature, 96% accuracy based fluorophores have been found to offer advantages over
in cancer diagnosis was accomplished. This is a large improvement traditional organic dyes. One of the most studied fluorescent
over clinical immunohistochemistry which boasted an accuracy nanoparticles is quantum dots (QDs), which are semiconductor
rate of 84%. Further tests conducted on 20 patients yielded an nanoparticles composed of groups II and VI elements (e.g. CdSe and
accuracy rate of 100%. The time required to perform the analysis is CdTe) or groups III and V elements (e.g. InP and InAs). Bulk
reduced from 3 days, for the clinical method, to less than an hour semiconductors have relatively small band gaps (less than 4 eV),
for DMR. This technology holds great clinical promise pertaining to thus behave like insulators at ambient conditions and exhibiting
the analysis of a wide range of targets including whole cells, electrical conductivity only under external stimulation. QDs, on
proteins, DNA/mRNA, metabolites, drugs, viruses, and bacteria the other hand, have a physical size of 2–10 nm in diameter, which
[174]. is smaller than the exciton Bohr radius. This results in a three-
Another magnetic nanoparticle-based biosensor technology dimensional quantum confinement of charge carriers, limiting the
has been developed by Osterfeld et al. based on the magnetoresis- number of possible energy states that an electron can occupy. In
tive (GMR) sensor [180]. Using protein analysis as an example, in a most cases, relaxation of an excited electron in QDs results in the
typical assay, a captured antibody for the desired analyte is release of the band gap energy in the form of light in the visible or
attached to a magnetoresistive sensor. The samples are added to near-infrared (NIR) region [184,185]. This fluorescence mechanism
the sensor but only the targeted analytes remain by interaction gives rise to unique absorption and emission profiles. Unlike
with the captured antibody. A second, biotinylated antibody is organic fluorophores which yield an emission spectrum that is
then introduced to bind to the previously captured analytes. usually a mirror image of the absorption one, QDs exhibit a broad
Finally streptavidin-coated magnetic nanoparticles are applied absorption spectrum but a narrow and usually symmetrical
46 H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69

Fig. 8. Magnetic nanoparticle-based biosensors. (a) Schematic representation of antibody–antigen binding on the GMR sensor surface. (b) Optical micrograph showing the
GMR sensor architecture. Inset: SEM image of one stripe of the GMR sensor that is bound with magnetic nanoparticle tags. (c) Schematic representation of a magnetically
labeled antibody, drawn to scale. (d) Visualization of CEA protein surface concentration at different times using a high-density GMR sensor array.
Reprinted with permission from ref. [182].

emission spectrum. This allows a group of QDs with distinctly illumination. For QD 608–streptavidin, the intensity was 104% of
different emissions to be excited by the same light source, making the initial intensity from the glycerol-mounted specimens and 97%
them ideal tags for multiplex imaging. from the antifade medium–mounted specimens at the end of the
The emission from QDs is size-dependent (Fig. 6a). A smaller illumination. Gao et al. showed that QDs were several thousand
particle size leads to an increased degree of confinement and in
turn, increases the difference in energy between the discrete
ground and excited states [186–189]. Taking CdSe nanoparticles as
an example, a red shift is observed when the QD size increases from
1.5 to 8 nm [190,191]. The emission also depends on the
composition of the QDs. For example, in ZnxHg1  xSe QDs, the
emission can be tuned by adjusting the composition to cover most
of the visible and NIR region [192]. On top of the semiconductor
core, a ZnS or CdS shell is usually deposited. The shell is several
atomic layers thick and passivates the photoactive cores of the QDs
[193,194]. With a wider band gap, the shell efficiently confines the
excitation to the core to reduce the non-radiative relaxation
pathways and increase the quantum yield (QY) [195]. The shell
thickness can have a significant impact on the photo-physical
properties of the QDs. While thinner shells (1–2 monolayers)
usually produce higher QY, thicker shells (4–6 monolayers) protect
the cores more effectively from photooxidation and degradation
[194,196]. A thicker shell also reduces QD ‘‘blinking’’. QD blinking
is an intermittence in light emission associated with charge
trapping and un-trapping at surface defects in a nanomaterial or
due to charge ejection from the QD (Auger ionization) followed by
recombination [197–199]. Reduced blinking is desired in diagnosis
since blinking may cause signal fluctuations in ultrasensitive
detection, loss of distance information when movement of a single
molecule is observed, and spectral jumping (change in the
emission peak position) [200].
In general, QDs have high absorption coefficients and QYs [201].
They can be 10–20 times brighter on an individual basis than
organic dyes [202]. QDs are also much more photostable, as shown
in Fig. 9b. Wu et al. compared the photostability of QD 608–
streptavidin and Alexa 488–streptavidin with specimens mounted Fig. 9. Photo-physical properties of QDs. (a) Size-dependent light emission. (b)
with glycerol or the antifade mounting medium Vectashield [203]. Comparison of photostability between QDs and dye molecules. (c) Capability of
absorbing high-energy (UV-blue) light and emitting fluorescence with a large
Without an antifade medium, the fluorescence intensity of Alexa
Stokes shift enables efficient separation of the QD signal over the fluorescent
488 diminished to 50% of the initial intensity at 10 s, and to 10% at background.(For interpretation of the references to color in this figure legend, the
60 s. With protection by the antifade reagents, Alexa 488 retained reader is referred to the web version of the article)
80% of the initial intensity at 60 s and 55% at the end of 3 min of Reprinted with permission from refs. [200,204].
H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69 47

times more stable against photobleaching than organic dyes (e.g. the CdSe cores. It is noted that Cd(CH3)2 is highly toxic and
Texas red) under the same excitation conditions (Fig. 9b) [204]. unstable, and special equipment and safety measures must be
These observations suggest the advantages of QDs over organic adopted. Alternatively, other Cd precursors (e.g. CdO) have been
dyes, especially for quantitative analysis and longitudinal tracking developed to allow QD preparation under milder conditions [210–
(Fig. 9c) [204]. 212].
Various reaction parameters such as the precursor, precursor
3.2.2. Synthesis of quantum dots concentrations, solvent, temperature, and reaction time can be
Initially, QD synthesis was performed in aqueous solutions tuned to achieve controlled QD synthesis [213–217]. A selective
which yielded particles with relatively poor fluorescence efficien- precipitation step is commonly employed to produce nanoparti-
cies and large size variation [205,206]. Advancements in synthetic cles with the desired size. Dabbousi et al. used selective
procedures and surface chemistry have enabled production of precipitation to prepare CdSe/ZnS QDs with the size of cores
water-soluble QDs with higher QYs (up to 40–50%) and narrower ranging from 2.3 to 5.5 nm [193]. The emission from these QDs
size distributions. For instance, the spectral emission widths are spans most of the visible spectrum from blue to red with QYs of 30–
50 nm from CdTe/CdSe particles and 19 nm from ZnSe QDs made 50%. Alternatively, size control can be achieved by gradually
by water-based approaches [205,207,208]. However, in general, increasing the reaction temperature. Zlateva et al. reported large-
aqueous synthesis is suboptimal in controlling the photo-physical scale synthesis of six colors of CdSe QDs in a single reaction by
and physiochemical properties of QDs. using a slow-increasing temperature gradient [218]. The QD
In 1993, Bawendi and co-workers developed a high-tempera- fractions have a narrow size distribution and produce sharp
ture decomposition procedure to produce QDs, and the procedures photoluminescence spectra with good QY (45–70% in organic
are now widely used [209]. In this process, organometallic solvent) and an overall high productive yield (95%) since every
precursors are pyrolyzed at high temperature to induce nucleation gradient is useful.
and nanocrystal growth. The protecting ligands such as trioctyl The photo-physical properties of QDs depend on the ratio of the
phosphine/trioctyl phosphine oxide (TOP/TOPO) are involved in metals. Zhong et al. prepared a series of alloyed ZnxCd1  xS
the reaction by interacting with the unsaturated surface metal (x = 0.10, 0.25, 0.36, 0.53) nanocrystals by pyrolyzing a mixture of
atoms and preventing the formation of the bulk semiconductor. In CdO– and ZnO–oleic acid complexes with sulfur in octadecene
a typical CdSe nanoparticle synthesis, the liquid precursors, e.g. a [219]. As the Zn content was increased, the photoluminescence
solution of Se and Cd(CH3)2, and capping ligands, e.g. tributylpho- spectra blue-shifted systematically across the visible spectrum
sphine, are injected into an organic solvent comprised of TOPO and from 474 to 391 nm. Qu and Peng found that for CdSe QDs, the
hexadecylamine at high temperature (300 8C). To maximize the initial Cd:Se ratio of the precursors was critical to the emission
photo-physical properties, a layer of ZnS is usually grown on top of wavelength, QY, types of the bright points (sharp or flat), and

Fig. 10. Routes for water-solubilization of hydrophobic QDs. (a–f) Ligand-exchange; (g–h) encapsulation.
Reprinted with permission from ref. [200].
48 H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69

sharpness of the luminescent peak [201]. In particular, a large ligand to displace the native TOPO molecules. Upon addition of
excess of the selenium precursor was found necessary to achieve a base, the silanol groups are hydrolyzed and condense to form a
high QY and narrow emission profile. Weiss group found that the stable and compact (1–5 nm thick) silica/siloxane shell rendering
ratio of Cd to Se (Cd/Se) in colloidal CdSe QDs is a function of the the particles hydrophilic. The QDs can also be encapsulated by
particle radius [199]. A Cd/Se ratio of 1.2:1 was observed from QDs silica using a reverse microemulsion coating method [241].
with a radius larger than 3.3 nm. This number increased to 6.5 However, the resulting coating is usually thicker, typically larger
when the radius was reduced to 1.9 nm, associated with the blue than 25 nm. Copolymers have also been used in QDs coatings. By
shifting of emission wavelength. using the ligand addition approach, the amphiphilic materials are
Much effort has been directed to the preparation of QDs with coated onto the hydrophobic QD surface via hydrophobic–
NIR emission. This is because tissue pigments, most importantly hydrophobic interactions, leaving the hydrophilic section free to
water and hemoglobin, exhibit minimum absorption in the NIR interact with surrounding water molecules to suspend the
region. This makes the NIR region (650–900 nm) a relatively particles. Gao et al. used an amphiphilic triblock copolymer to
transparent spectrum window and ideal for in vivo fluorescence coat QDs [242]. The resulting QDs were very stable in water and
imaging [220]. Liu et al. reported a one-pot synthesis of InAs/CdSe able to be coupled with targeting agents such as antibodies in
QDs with bright, stable, and narrow NIR photoluminescence [192]. tumor imaging. This ligand addition approach usually produces
Kelley’s group prepared NIR PbS QDs with a QY of 26% [221]. QDs with a large hydrodynamic size.
Alloyed QDs emitting in the NIR region such as CuIn5Se8 [222],
CdxHg1  xTe [223,224], and Cu-doped InP [225] have been 3.2.4. Toxicity of quantum dots
synthesized [226–228]. However, Cd-free QDs in general have In biomedical applications, the toxicity of the materials and
poorer stability and inferior photo-physical properties than Cd- their interaction with biological systems must be addressed.
containing ones [200]. Recently, Nie’s group found that lattice Extensive toxicity studies have been performed on QDs both in
strain could be used to adjust the emission wavelength of QDs vitro and in vivo but there is no common consensus, partially due to
[229]. They prepared QDs by epitaxial deposition of a compressive the diversity of QDs. It is important that each type of QD is
shell (ZnSe or CdS) on a soft and small nanocrystalline core (CdTe). characterized individually as not only the constituent metals, but
The lattice mismatch between the core and shell led to dramatic also the QD size, charge, and functional group should be considered
changes in both the conduction and valence band energies and in as contributing factors to QD toxicity [243].
turn, a large spectral shift. These strain-tunable QDs exhibit Protection layers and surface coatings can reduce the toxicity of
narrow light emission with high QY (60%) across a broad range of QDs. This is because QDs deteriorate in the physiological
visible and NIR wavelengths (500–1050 nm). environment and release toxic heavy metals into the surroundings.
The organic coating or inorganic shell can inhibit or retard this
3.2.3. Surface modification of quantum dots process and consequently, reduce the toxicity. Derfus et al. studied
QDs made by thermal decomposition are highly hydrophobic. the cytotoxicity of CdSe QDs and found that without a coating, the
One common surface modification approach is to substitute the cytotoxicity of CdSe cores could be correlated with the liberation of
native TOPO/TOP coating with a bifunctional ligand comprised a free Cd2+ ions [244]. With the appropriate coating, CdSe QDs could
surface-anchoring group (mostly thiols) and a hydrophilic be rendered nontoxic and used to track cell migration and
functional group (Fig. 10). Examples include negatively charged reorganization. Lovric et al. found that CdTe QDs coated with MPA
carboxyl-terminated thiols such as mercaptoacetic (MAA) [230], and cysteamine were much less toxic to rat pheochromocytoma
mercaptopropionic (MPA) acids [231] and thiol-containing zwit- cells (PC12) than uncoated ones [245]. However, the coating itself
terionic molecules such as cysteine [66,232]. In addition to can be the source of toxicity. For instance, Hoshino et al. observed
providing hydrophilicity, these terminal chemical groups can be severe cytotoxicity from EL-4 (mouse lymphoma) cells when they
utilized in conjugation with other biological species such as were treated with a QD capping ligand, MUA at 100 mg/mL [247].
proteins, peptides, or nucleic acids. Despite the simplicity of the They believed that it was MUA, not the QD cores, that was the
procedure, ligand exchange with monodentate surface ligands is major cause of the cytotoxicity but their postulate is debatable
often associated with compromised fluorescence efficiency, [247]. A multidentate coating may help reduce the toxicity. Voura
photochemical stability, and shelf life. Because of limited binding et al. treated B16F10 melanoma cells with DHLA-capped CdSe/ZnS
affinity, monodentate ligands tend to detach from the QD surface, QDs (5 mL/mL) and found no detectable reduction in cell growth
leaving behind surface trap sites and causing nanoparticle [248]. Jaiswal et al. treated HeLa and Dictyostelium discoideum cells
aggregation [233,234]. The stability can be dramatically improved with DHLA-capped CdSe/ZnS QDs (400–600 nM) for 1 week and
by using multidentate ligands. Liu et al. used a di-thiol ligand, observed no adverse effects on the cell morphology and physiology
PEGylated dihydrolipoic acid (DHLA), to modify QDs. The surface [249]. This can be attributed to a more stable coating and reduced
modification process led to aqueous stable nanoparticles with a risk of the capping falling off.
moderate drop in QY (from 65% to 43%) [235]. Sukhanova et al. The particle size also has an impact. Lovric et al. observed that
reported the use of cysteine to modify QDs and further smaller cationic QDs (2.2  0.1 nm) induced more pronounced
stabilization of the particles with poly(allylamine). They observed cytotoxicity than larger cationic QDs (5.2  0.1 nm) [245]. Later
an increase in QY from 40% to 65% [236]. Jiang et al. improved the studies revealed that the variation was associated with the different
stability of a mercaptoundecanoic acid (MUA) shell by covalently subcellular distribution pattern of the two particles. While larger QDs
cross-linking neighboring molecules with lysine [237]. However, are mainly distributed in the cytosol, smaller QDs are localized to the
cross-linking also resulted in a dramatic increase in the nanopar- nuclei which are more susceptible to damage. Toxicity can be caused
ticle size (from 8.7 to 20.3 nm). Recently, Smith and Nie developed by several factors including free Cd ions, free radicals, or QD
a new class of multidentate polymer coatings only 1.5–2 nm thick interaction with intracellular components. To elucidate the mecha-
[238]. Consisting of a poly(acrylic acid) backbone grafted with nism, QDs were co-incubated with N-acetylcysteine (NAC; a known
multiple anchors (thiol and amine groups), this coating renders inhibitor of Cd toxicity) and Trolox (a water-soluble vitamin E). While
CdTe QDs biocompatible and colloidally stable, while keeping the NAC improves the viability, Trolox fails to do so, suggesting that the
final particle size between 5.6 and 9.7 nm. toxicity stems from Cd in lieu of the free radicals. Since it is generally
Silane-based coatings have also been proposed [239,240]. 3- believed that the released heavy metals are the main source of QD
(mercaptopropyl)trimethoxysilane (MPS) has been used as a toxicity, researchers have developed non-Cd formulations such as
H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69 49

InAs/ZnSe [250], InAs/InP/ZnSe [251], and Cu-In-Se/ZnS QDs [252]. DHLA, cysteamine, cysteine, and PEGylated DHLA with hydrody-
Although there are still toxic elements such as indium and arsenic, the namic sizes ranging from 4.4 to 8.7 nm. A hydrodynamic size of
toxicity is significantly mitigated compared to those containing Cd, 5.5 nm was found to be a critical point below which nanoparticles
Hg, and Pb [253]. could be efficiently eliminated from the body through renal
The size of QDs determines their distribution and circulation in a clearance [66]. Gao et al. prepared MPA coated InAs/InP/ZnSe QDs
biological system and therefore affects the toxicity profiles. A with a hydrodynamic size of less than 10 nm [254]. UV/vis
reduced particle size may lead to a longer circulation half-life and absorption and fluorescence emission analyses detected QDs in
lower liver uptake. However, further reducing the particle size to urine samples from the administrated animals, suggesting that QDs
<10 nm will meet the threshold of renal filtration and urinary could be cleared through the urinary excretion.
excretion. This may be desirable in imaging because the unbound The majority of in vivo studies with QDs have hitherto shown no
probes will not stay in the body for a long time but instead be obvious toxicity. Larson et al. observed no noticeable ill effects in
excreted via urine. An important study was conducted by Frangioni mice injected with 20 nM and 1 mM solutions of CdSe/ZnS QDs
et al. who intravenously administered QDs into rodents and studied [255]. Ballou et al. injected amphiphilic polymer coated QDs at a
the size effect. They studied CdSe/ZnS nanoparticles coated with 20 pmol QD/g dose to mice [256]. The injected mice remained

Fig. 11. Histological images from the major organs of the rhesus macaques 3 months after intravenous injection of the QD formulation. In each pair, the left image is from the
control animal and the right image is from a treated animal. Tissues were collected from brain (a), heart (b), liver (c), spleen (d), lung (e), kidney (f), lymph (g), intestine (h) and
skin (i). Images were taken at 40 magnification.
Reprinted with permission from ref. [257].
50 H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69

viable for 133 days and histology studies found no signs of necrosis
at the sites of tissue deposition. More exciting news came recently
from a pilot toxicity study on non-human primates which found no
adverse response to intravenous injection of QDs (Fig. 11) [257].
The researchers injected rhesus macaques with phospholipid
micelle encapsulated CdSe/CdS/ZnS QDs at a dose of 25 mg/kg. The
blood and biochemical markers were found to be in the normal
range and histology of major organs showed no abnormalities after
90 days. Two treated monkeys underwent continued evaluation
and showed no ill effects for 1 year. However, it was observed that
most of the initial cadmium dose remained in the liver, spleen, and
kidneys after 90 days. The ultimate fate of the heavy metals and
their long-term impact on the body require further analysis, but
nonetheless, this encouraging pilot study suggests a potential role
of QDs in clinical applications.

3.2.5. Quantum dots for in vivo imaging


Despite controversies, tumor targeting is believed to benefit
from a long circulation half-life, and for this purpose, a stable and
thick coating offers many advantages [258]. A coating with high
molecular weight PEG has been found to reduce rapid clearance of
QDs by liver and bone marrow [256], and the use of branched PEG
may further improve circulation [258,259]. Amphiphilic triblock
copolymer coated QDs show a circulation half-life of 5–8 h [258].
Gao et al. have coated CdSe/ZnS QDs with a triblock copolymer and
conjugated onto the coating an anti-PSMA antibody [242]. In vivo
imaging conducted on subcutaneous tumor models reveals good
tumor accumulation attributable to the enhanced permeability
and retention effect (EPR) as well as antibody–PSMA interaction
(Fig. 12). Cai et al. conjugated c(RGDyK) onto polymer coated NIR
QDs (705 nm) and studied tumor targeting on a subcutaneous
U87MG human glioblastoma model [260]. The tumor fluorescence
intensity reached a maximum at 6 h p.i. with good tumor-to-
normal-tissue contrast, and the targeting was mainly caused by the
RGD–integrin interaction. Recently, Gao et al. reported a dendron-
coated InP/ZnS core/shell QD with 710 nm emission [261]. After
Fig. 12. In vivo imaging of implanted QD-tagged tumor cells. (a) Bright QD tags
coupling with a RGD dimer, the particles had a hydrodynamic size enable visualization of tumor cells with a non-invasive whole-animal fluorescence
of 12 nm, but unexpectedly, the fluorescent signals were found in imaging, whereas organic dye signal is indistinguishable from autofluorescence. (b)
urinary bladder and urine collected 90 min after administration. A Imaging of subcutaneously implanted QD-loaded microbeads shows the potential
pilot mouse toxicity study confirmed that the particles did not for multiplexed in vivo cell detection and tracking.
Reprinted with permission from ref. [242].
cause significant toxicity at their particular doses. The nanopar-
ticles showed a long circulation half-life that allowed clear
delineation of the tumor up to 28 h p.i. (Fig. 13). Gao et al. also
conjugated anti-HER2 antibody molecules onto phospholipid QDs have been studied with other imaging modalities to assess
coated QDs [262] and the resulting conjugates were able to tumor targeting. Smith et al. used intravital microscopy to study in
specifically target HER2-positive tumors with a peak tumor-to- vivo extravasation and cellular binding with RGD–QD conjugates
background ratio of 4 at the 4 h p.i. time point. [263]. Unlike whole-body fluorescence imaging, intravital micros-
Long circulating QDs can stay intact for months in living subjects copy is used to examine a small area with high spatial (0.5 mm)
and remain fluorescent, making repeated injection and imaging and temporal resolution. It is therefore a prefect technique to study
impossible. Reducing the particle size below the renal clearance the migration of nanoparticles in vessels and their interaction with
threshold may be preferred from the perspective of reduced the surroundings. RGD–QDs do not extravasate and are only bound
background and toxicity. Choi et al. used cysteine to modify QDs as aggregates rather than individually to tumor vasculature, where
and couple them to the terminal carboxyl groups of either GPI (a integrin avb3 is overexpressed. Further studies disclose a similar
small molecule targeting PSMA) or c(RDyK) [66]. A maximum of 5– binding feature in different tumor types [264]. Extravasation, on
10 GPI or cRGD could be conjugated to the QD surface while the the other hand, is highly tumor-type dependent. The QDs used in
hydrodynamic size of the particles was kept within 5.5 nm, the size the study were polymer coated with a hydrodynamic size of 20–
threshold for efficient renal clearance. The QD–GPI conjugates were 25 nm. Whether or not the same conclusion is applicable to other
injected into bilateral subcutaneous tumor models implanted with types of QDs is unknown.
both LNCap (PSMA positive prostate cancer cell line) and PC-3 (PSMA Cai et al. labeled RGD–QDs with 64Cu, a radioisotope for PET
negative prostate cancer cell line). The QD–RGD conjugates were imaging, and studied the particles in subcutaneous U87MG models
injected into animal models where M21 (human melanoma cells, by both PET and fluorescence imaging [265]. Based on PET imaging
integrin avb3 positive) and M21-L (integrin avb3 negative) were results, RGD–QDs showed tumor accumulation rates of 2.2  0.3
inoculated. In both scenarios, tumor accumulation occurred in and 4.0  1.0%ID/g at 5 and 18 h p.i. respectively. Excellent linear
positive tumors but not negative ones, and most of the unbound QDs correlation was achieved from the results measured by in vivo PET
were eliminated through the urine within 4 h, thereby providing a imaging and those measured by ex vivo NIRF imaging and tissue
‘‘clinically realistic’’ imaging window. homogenate fluorescence (r2 = 0.93). Histologic examination revealed
H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69 51

Fig. 13. In vivo NIR fluorescence imaging results with QDs. (a) Structure and synthesis of QD710–Dendron–RGD2 conjugate. (b) Representative organ histology of PBS and
QD710–Dendron treated animals. Scale bar, 100 mm. The dorsal images of SKOV3 tumor-bearing (arrows) mice (L, left side; R, right side) injected with (c) QD710–Dendorn–
RGD2 (200 pmol) and (d) QD710–Dendron (200 pmol) at 0.5, 1, 4, 5, 5.5, 6, 8, 24, and 28 h p.i.
Reprinted with permission from ref. [261].

that the particles mainly targeted the tumor vasculature through QD655, 605, and 565 on fixed breast cancer cells [268]. By
RGD–integrin interaction with little extravasation. analyzing the fluorescence activities and taking into account the
relative intensities of each type of QD, they were able to determine
3.2.6. Quantum dots for immunostaining the relative abundance of the three markers. The results were in
Histological techniques such as immunohistochemistry (IHC) accordance with those from the conventional methods. The same
are used in research and clinics to analyze cell or tissue specimens. group also labeled five biomarkers, HER2, ER, PR, EGFR and mTOR,
Despite the advantages of in vivo imaging, histopathology simultaneously with 525, 565, 605, 655 and 705 nm QDs in breast
continues to be essential to cancer diagnosis and staging. Although cancer cells [268]. The results correlated well with those obtained
organic fluorophores have been widely used, their disadvantages by traditional IHC, Western blotting and FISH, thus corroborating
such as quick photobleaching and inflexible excitation wave- the advantages and potential of QDs in histological studies.
lengths are well documented [266]. QDs, with much better Compared to in vivo applications, there are fewer restrictions on
photostability and brightness, hold great potential in this field. using QDs in a histology setting. The impeding issues in vivo, such
With broad excitation wavelengths as well as narrow and tunable as tissue autofluorescence and toxicity, are less of a concern when
emission wavelengths, QDs allow multiplexed labeling of bio- using QDs in immunostaining. However, with a relatively large
markers in tissue samples. Ghazani et al. showed three-color size, QDs have limited access to intracellular compartments,
staining of EGFR, E-cadherin, and cytokeratin with 655, 605, and making labeling cytoplasmic and nuclear markers challenging.
565 nm QDs on lung carcinoma xenografts [267]. Yezhelyev et al. Another issue is non-specific interaction of particles. To achieve
demonstrated multiplexed labeling of HER2, ER, and PR with highly sensitive and quantitative analysis, it is critical that particles
52 H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69

are immobilized on the specimens through ligand–biomarker anti-Stokes shift is caused by a nonlinear optical process in which
interaction and the background signals are at a minimum. A two or more photons are almost simultaneously absorbed to
majority of the non-specific binding arises from electrostatic provide sufficient energy for the upconversion. UCNPs show a
interactions. To avoid this, a common strategy is to functionalize much lower QY than QDs. Using bulk upconversion materials, the
the QD surface with PEG. This however, leads to a larger particle highest QY reported so far is 4.5% (260 mW cm2) [175,176]. QYs
size. for UCNPs are even smaller and the values are in the range of
Nonetheless, quantitative molecular profiling with QDs has 0.005–0.3, with the highest measured value being 0.3% for 100 nm
recently been studied clinically. Liu et al. used QD-based NaYF4:Yb3+,Er3+ nanoparticles [271]. UCNP-based imaging benifits
immunohistofluorescence (IHF) to analyze samples involving a from a clean background. For in vivo imaging, this may give rise to
confirmed case of Hodgkin’s lymphoma, two suspicious lymphoma an increased signal-to-noise ratio and longer penetration depth.
cases, and two with reactive lymph nodes (but not lymphoma). IHF
was able to accurately detect Hodgkin’s lymphoma and to 3.3.1. Construction of upconversion nanoparticles
differentiate it from lymphoid hyperplasia [269]. Xu et al. UCNPs are comprised of three components: a host matrix,
compared QD-based IHF and IHC on quantifying the aldehyde sensitizer, and activator [272]. The three components play
dehydrogenase 1 (ALDH1A1) expression in primary tumor samples different roles but are all essential to the energy upconversion
taken from 96 HNSCC patients including 50 with disease in the process. The host matrix is usually made of inorganic materials
lymph nodes and 46 without (Fig. 14) [270]. The two methods were with low lattice photon energies to minimize non-radiative loss
found to be comparable from the viewpoint of quantification but and maximize emission [273]. Many fluorides, oxides, phosphates,
QD-IHF was more sensitive and objective than IHC. oxysulfides, and so on have been studied as host matrix materials
to construct UCNPs [274]. Among them, fluorides are the most
3.3. Upconversion nanoparticles widely used due to the small photon energies and high chemical
stability [273]. Sensitizers or activators are usually trivalent
Upconversion nanoparticles (UCNPs) have recently emerged as lanthanides incorporated into the inorganic crystalline host.
a new type of optical nanoparticle. UCNPs absorb NIR light Yb3+ is the most common sensitizer for its large absorption
(commonly 980 nm) and emit with higher frequency. The cross-section in the NIR region [275]. On the other hand, Er3+, Tm3+,

Fig. 14. Correlation between QD-based IHF and conventional IHC. (a) Aldehyde dehydrogenase 1 (ALDH1A1)-positive tumor; (b) ALDH1A1-negative tumor; (c) correlation
between ALDH1A1 expression levels measured by immunohistochemistry (IHC) and by QD-IHF. Calculated using Spearman’s rank correlation. (d) Box-Plot for ALDH1A1 in
non-metastatic and metastatic groups.
Reprinted with permission from ref. [270].
H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69 53

and Ho3+ are the most popular activators because they possess largely minimized surface quenching induced emission losses
multiple metastable states that are beneficial to energy conversion [279].
[275]. Both sensitizers and activators are at a low concentration in The size can also affect the relative emission intensities. For
the overall nanostructure, usually 10 mol% for the sensitizer and Y2O3:Yb3+,Er3+ UCNPs, the relative intensity of the blue and red
<2 mol% for the activator [274]. Upon irradiation, the sensitizers emissions to the green emission increases gradually with
are excited and transfer the energy to nearby activators to emit decreasing particle size [280]. This is ascribed to the highly
light. energetic vibrations of the surface-bound hydroxyl and carbonate
By adjusting the combination of the host, sensitizer and groups that bridge the energetic gaps between different levels
activator, the emission from UCNPs can be tuned to cover a broad thereby enhancing the population of the red emitting levels [281].
spectrum range from violet to NIR [273]. For instance, a Strong size-dependence of the spectral properties was also
combination of NaYF4 nanocrystals with Yb3+ and Tm3+ results observed by Schietinger et al. with NaYF4:Yb3+,Er3+ UCNPs. In
in UCNPs with blue emission. Yb3+/Er3+ co-doped UCNPs display their study however, relatively more efficient green emission was
green emission at 510–570 nm (centered at 525 and 550 nm) and observed when the particle size was decreased. The authors
red emission between 630 and 680 nm (centered at 660 nm) [276]. excluded surface effects in the mechanism and instead attributed
Yb3+/Ho3+ co-doped UCNPs exhibit three main bands centered at this behavior to a change in the phonon-mediated processes in the
541, 647 and 751 nm [277] and multiple dopants can result in emitting ions. They believed that both the confinement effect on
UCNPs with a broader spectrum coverage. Wang and Liu doped the the spatial distribution of phonon modes [282] and reduced
NaYF4 matrix with Yb3+, Tm3+, and Er3+ and by adjusting the spectral density of phonon states contributed to the size effect
concentrations, obtained colloids with over a wide color range: [283].
bluish to whitish (NaYF4:Yb3+,Tm3+,Er3+) and greenish-yellowish The QYs are also affected by the host matrix. In order to
to redish (NaYF4:Yb3+,Er3+), as shown in Fig. 15 [278]. minimize non-radiative losses, fluorides, in particular NaYF4, are
As mentioned above, UCNPs have relatively low QYs. Efforts overwhelmingly used as the host matrix. However, at ambient
have been made to improve the QYs by adjusting the particle pressure, there are two crystal phases of NaYF4, the cubic phase (a-
composition and architecture. For UCNPs with the same composi- phase) and hexagonal phase (b-phase). Although the hexagonal
tion, the QY is size-dependent and enhanced by an increased phase of NaYF4 is thermodynamically more stable, it is the cubic
particle size. This is due to defects at the particle surface boundary phase that is usually obtained during a typical UCNP synthesis.
that are detrimental to luminescence. Smaller particles have larger When converting a-NaYF4:Yb3+,Er3+ UCNPs to b-NaYF4:Yb3+,Er3+
surface-to-volume ratios and are affected more by the effect. The UCNPs by post-synthesis annealing [284], an enhancement in
surface effect was confirmed by Wang et al. who coated fluorescence intensity by 40 fold was observed [285], suggesting b-
NaYF4:Yb3+,Tm3+ UCNPs with an inert thin-shell coating. The NaYF4 as the more favorable phase. However, annealing usually
coating preserved the optical integrity of the nanoparticles and leads to severe aggregation of particles. More practically,
nanoparticles are made by thermal decomposition at a high
temperature and for a long time in order to produce the hexagonal
phase in one step.

3.3.2. Synthesis of upconversion nanoparticles


UCNPs can be synthesized in an aqueous solution by co-
precipitation. Yi et al. utilized a co-precipitation method to
prepare NaYF4:Yb3+,Er3+ UCNPs with the aid of ethylenediamine
tetraacetic acid (EDTA) [285]. The metal–EDTA complex was
quickly injected into a NaF solution under vigorous stirring to
effect nucleation and formation of the cubic phase, a-NaYF4:Y-
b3+,Er3+ UCNPs. The particle size could be controlled from 37 to
166 nm by varying the ratio between EDTA and metals. However,
as aforementioned, a-NaYF4 is suboptimal as a host matrix and
the fluorescent intensity of the resulting UCNPs is weak. An
annealing treatment is necessary to achieve the phase transition
but this is usually accompanied by severe particle aggregation and
coating carbonization, thus imposing limitations on the use of the
products in a biological setting. Instead, b-NaYF4 UCNPs can be
made through one-step thermal decomposition at a high
temperature using a long reaction time. This is accomplished
by a hydrothermal method in which the rare-earth and fluoride
precursors are mixed in an aqueous solution and heated to high
temperature in a sealed autoclave (often Teflon-lined). Li’s group
developed a general hydrothermal protocol to synthesize
nanoparticles [286–289], and a series of rare-earth fluoride
nanocrystals with different hosts, crystal structures, sizes and
shapes were prepared accordingly [290–313]. Wang et al.
demonstrated that NaYF4:Yb3+,Er3+ UCNPs can be tuned in size,
phase (cubic or hexagonal), and upconversion behavior by
Fig. 15. UCNP multicolor fine-tuning with dopants: (a) NaYF4:Yb3+/Er3+ (18/ introducing trivalent lanthanide ions at precisely defined con-
2 mol%), (b) NaYF4:Yb3+/Tm3+ (20/0.2 mol%), (c) NaYF4:Yb3+/Er3+ (25–60/2 mol%),
centrations (Fig. 16) [314]. Zhang’s group prepared pure b-
and (d) NaYF4:Yb3+/Tm3+/Er3+ (20/0.2/0.2–1.5 mol%) (10 mM). Luminescent photos
of (e) NaYF4:Yb/Tm (20/0.2 mol%), (f–j) NaYF4:Yb3+/Tm3+/Er3+ (20/0.2/0.2– NaYF4:Yb3+,Er3+/Tm3+ nanocrystals with controllable shape and
1.5 mol%), and (k–n) NaYF4:Yb3+/Er3+ (18–60/2 mol%). strong emission [315]. Schafer et al. reported the synthesis of b-
Reprinted with permission from ref. [278]. NaYF4:Yb3+,Er3+ particles at ambient temperature by using
54 H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69

Fig. 16. Simultaneous phase and size control of UCNPs through lanthanide doping. (a–c), TEM images of NaYF4:Yb3+/Er3+ (18/2 mol%) products in the absence of Gd3+ dopant
ions. (d) HRTEM image of UCNPs. (e) Selected area electron diffraction pattern of UCNPs in (a). (f–h) TEM images of the NaYF4:Yb3+/Er3+ (18/2 mol%) products. (i) HRTEM
image of UCNPs in (h). (j) DFT calculation. Scale bars are 500 nm in (a–c), 200 nm in (f–h) and 5 nm in (d) and (i).
Reprinted with permission from ref. [314].

Y2(CO3)3, Yb2(CO3)3, Er2(CO3)3, Na2CO3, and NH4F as precursors In the above cases, the UCNPs are prepared in octadecene using
[316]. In addition to NaYF4, LaF3, NaLuF4, and NaYb1  xGdxF4- OA and/or OAm as the capping ligand. Recently, Shan et al.
based UCNPs have also been prepared hydrothermally [317–321]. synthesized b-NaYF4:Yb3+,Er3+/Tm3+/Ho3+ UCNPs using TOPO
UCNPs are more commonly synthesized by thermal decompo- [327] which served as both the high-boiling solvent and capping
sition in organic solvents with a high-boiling point. Zhang et al. reagent. The UCNPs showed good upconversion efficiency and
made triangular LaF3 nanoplates by thermal decomposition of narrow size distribution. More importantly, the energy barrier of
La(CF3COO)3 in octadecene with OA as the capping ligand [322]. the cubic-to-hexagonal phase transition was significantly reduced
This method was later extended to prepare different types of by this procedure which favored the b-phase NaYF4 formation. The
UCNPs. Boyer et al. synthesized a-NaYF4:Yb3+,Er3+/Tm3+ UCNPs same group later reported the synthesis of b-NaYF4:Yb3+,Er3+
using Na(CF3COO) and Re(CF3COO)3 as precursors [323,324]. They UCNPs using octadecene as the solvent and both OA and TOP as
showed that the rate of decomposition and particle formation capping reagents [328]. Based on a series of studies, a two-step
could be controlled by the addition rate of precursors to the growth mechanism for b-NaYF4 UCNPs is proposed. The first step
reaction solution. With a slow addition rate, the nucleation and involves a kinetically controlled precipitation stage in which a-
growth stages of the NPs could be separated, and the resulting NaYF4 UCNPs are formed. The second is a diffusion controlled
products showed a narrow size distribution. Mai et al. further growth stage in which a-NaYF4 are converted into b-NaYF4. Wei
studied the reaction mechanism and demonstrated that NaYF4:Y- et al. prepared NaYF4:Yb3+,Er3+ UCNPs using Re–oleate complexes
b3+,Er3+ UCNPs were formed via a unique delayed nucleation as the precursors and octadecene as the reaction solvent [317]. The
pathway [325]. They found that monodispersed b-NaYF4:Yb3+,Er3+ phase transition could be tuned by controlling the reaction
UCNPs with tunable sizes could be obtained from a-NaYF4:Y- temperature. a-NaYF4:Yb3+,Er3+ UCNPs were obtained by heating
b3+,Er3+ monomers by restricting or enhancing the Ostwald at 210 8C for 6 h whereas b-NaYF4:Yb3+,Er3+ UCNPs were prepared
ripening process in which the cubic-to-hexagonal phase transition by heating at 260 8C for 6 h.
process occurred at a delayed time. This synthetic protocol has Although thermal decomposition is an effective approach to
been developed into a common synthesis technique and many fabricate monodispersed, single-crystalline, well-defined, and
other UCNPs have been synthesized using a similar approach. phase-pure UCNPs, some disadvantages remain, such as requiring
Wang et al. reported the synthesis of Er3+-, Tm3+- and Ho3+-doped rigorous (anhydrous and oxygen-free) and harsh (long reaction
NaYbF4 UCNPs via thermal decomposition [224] and Du et al. time and high reaction temperature) synthesis conditions. In
prepared NaMF3 (M = Mn, Co, Ni, Mg) and LiMAlF6 (M = Ca, Sr) addition, thermal decomposition of metal trifluoroacetates pro-
nanocrystals [326]. duces fluorinated and oxy-fluorinated carbon species that are
H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69 55

considered toxic. Moreover, the capping ligands such as OA, OM,


and TOPO have long hydrocarbon chains that make the synthesized
UCNPs insoluble in water.

3.3.3. Surface modification of upconversion nanoparticles


In this section, we discuss UCNPs produced by thermal
decomposition as this is the most popular method. Similar to
IONPs and QDs, UCNPs made by thermal decomposition are coated
with a hydrophobic layer and not readily applicable to biological
applications. A post-synthesis surface modification step is needed
to make them water soluble. This can be achieved by adding an
amphiphilic coating layer. Pedroni et al. deposited an oleate layer
on OA-coated CaF2:Yb3+,Er3+/Ho3+/Tm3+ nanoparticles, which then
became water soluble [309]. Prud’homme et al. modified OA-
coated NaYF4:Yb3+,Er3+ UCNPs with copolymers including poly(-
ethylene glycol)-block-poly(caprolactone), poly(ethylene glycol)-
block-poly(lactic-coglycolic acid), and poly((ethylene glycol)-
Fig. 17. Cell viability reults from MTT assays with UCNPs. KB cells were cultured in
block-lactic acid). The copolymers could be assembled on the the presence of 100–500 mg/mL UCNPs for 4 and 24 h.
particle surface and transitioned the UCNPs to an aqueous phase Reprinted with permission from ref. [337].
[329]. Surface modification using a biomimetic phospholipid layer
has also been reported [330]. found that even at a high concentration of 250 mg/mL, the cell
UCNPs made from thermal decomposition are frequently viability still remained above 80%. Zhou et al. synthesized citrate-
coated with OAm or OA. The interaction between OAm and coated UCNPs (cit-NPs) and evaluated the impact of cit-NPs on KB
particle surface is relatively weak and can be replaced by ligands cells at various concentrations (100–500 mg/mL, Fig. 17) [337]. No
containing carboxyl or multiple amine groups. PEG-diacid (MW significant difference in cell proliferation was found in the absence
600) [331], polyethylenimine (PEI) [332], and thioglycollic acid or presence of 100–500 mg/mL cit-NPs. Even when the materials
(TGA) [333,334] have been employed to exchange the surface OAm were incubated at the highest dose (500 mg/mL) for 24 h, a cell
of UCNPs. Similarly, NaYF4:Yb3+,Er3+ UCNPs with the coordinating viability of 81% was still observed. Recently, Xiong et al.
solvent N-(2-hydroxyethyl)ethylenediamine (HEEDA) can be intravenously (i.v.) injected UCNPs at 15 mg/kg to mice and
surface modified with ethane-1,1-diphosphonic acid (HEDP) studied the particle distribution and long-term toxicity [342]. They
[335]. On the other hand, OA has stronger interactions with observed no apparent adverse effects for 115 days, suggesting that
lanthanide ions thereby requiring ligands with high affinity and the particles were well-tolerated. Owing to the potential of in the
usually multi-dentation to achieve ligand replacement. Examples field of imaging, there is a growing interest in understanding the
include citrate [336,337], polyacrylic acid (PAA) [329,338–342], toxicity and metabolism of UCNPs in living subjects. So far, it is
DMSA [343], 3-mercaptopropionic acid (3MA) [344,345], poly(- generally believed that UCNPs are more tolerable than QDs as
ethyleneglycol) (PEG)-phosphate [346], hexanedioic acid (HDA) imaging probes in vivo.
[347], 1,10-decanedicarboxylic (DDA) and MUA [348]. OA-coated
NaYF4:Yb3+,Er3+/Tm3+ UCNPs can be modified by PEG-phosphate to 3.3.5. Upconversion nanoparticles for tumor imaging
become water dispersible [346]. Dong et al. reported a generalized Boasting an ultralow background, UCNPs are potentially
ligand exchange strategy using nitrosonium tetrafluoroborate advantageous over QDs for in vivo imaging. Cheng et al. conducted
(NOBF4) to replace the original ligands (OA or OAm) [349]. After the a side-by-side comparison between UCNPs and QDs using small
UCNPs were stabilized in polar and hydrophilic media, they were animals [361]. Despite a high QY, the detection limit of QDs was
further modified by hydrophilic polymers like PVP to achieve severely impeded by tissue autofluorescence and it was not
water stability. possible to detect a concentration lower than 5 nM. On the other
Other ligand modification strategies have also been explored. hand, UCNPs could be detected at a 0.1 nM level.
For example, NaYF4 UCNPs were coated with SiO2 by a common UCNPs have been coupled with targeting peptides and
sol–gel process [285,350]. 3-aminopropyltrimethoxysilane [351] antibodies. However, tumor selectivity by these ligand–UCNP
and N-[3-(trimethoxysilyl)propyl]ethylenediamine (AEAPTMS) conjugates in vivo has seldom been demonstrated. One of the first
[352] are usually co-condensed with TEOS to provide the chemical studies was performed by Xiong et al. who used folic acid as a
functional groups for further conjugation. Chen et al. prepared targeting molecule and coupled it onto 6AA-coated NaYF4:Y-
carboxylic acid functionalized NaYF4 UCNPs by direct oxidation of b3+,Er3+ nanoparticles [362]. The particles were injected into a
the OA coating using the Lemieux–von Rudloff reagent [353]. This HeLa xenograft mouse model and imaging revealed significant
approach is unique because no new ligands are introduced to the difference from the control group to which amine-functionalized
particle surface. UCNPs were injected. The targeting selectivity was also confirmed
by a blocking study in which free folic acid was injected along with
3.3.4. Toxicity of upconversion nanoparticles UCNP probes. The same group also reported RGD conjugated
There have been relatively few studies on the toxicity of UCNPs, UCNPs for tumor imaging [363]. In their investigation, NaYF4:Y-
and based on archival reports, cells and animals appear to have b3+,Er3+,Tm3+ UCNPs were used and imaging was performed on a
good tolerance to UCNPs [354–359]. However, the compatibility of U87MG subcutaneous model. Good tumor to normal tissue
UCNPs depends on the particle composition as well as coating contrast was observed by monitoring the emission at 800 nm at
materials and these issues need to be studied separately. Zhang 24 h p.i. Yu et al. reported CTX conjugated, NaYF4:Yb3+,Er3+/Ce2+
group studied the cytotoxicity of polyethyleneimine (PEI) [360] UCNPs for tumor imaging [364] and good tumor uptake was
and silica-coated [357] UCNPs. On silica-coated UCNPs at 1 mg/mL observed from both in vivo and ex vivo imaging.
for 24 h incubation, they observed cell viability of 93.4% for skeletal There is an interest in developing UCNP-based probes for
cells and 93.2% for bone marrow MSCs [357]. Hu et al. incubated KB multimodality imaging. Zhou et al. evaluated NaGdF4:Y-
cells with elevated concentrations of PEGylated UCNPs [355] and b3+,Er3+,Tm3+ [354] and Gd3+ labeled NaYF4:Yb3+,Er3+,Gd3+ UCNPs
56 H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69

as MRI (T1) and upconversion luminescence (UCL) dual-modal UCL feature. Efforts should be made to develop commercial
imaging probes [337]. These particles can even be labeled with 18F microscopes and scanners dedicated to UCNP-based imaging.
to achieve MRI/UCL/PET tri-modality imaging. Xia et al. developed
NaYF4:Yb3+,Tm3+/FexOy core/shell nanoparticles and applied them 3.4. Gold nanoparticles
to MR (T2) and UCL lymphatic imaging [365]. Xiao et al. coated
UCNPs with a TaO shell and studied the particles in MRI/UCL/CT tri- 3.4.1. Optical properties of gold nanoparticles
modal imaging [366]. In the latter case, the water-soluble The synthesis of GNPs dates back to ancient Roman times but
nanoparticles have an average size of about 30 nm and extraordi- only recently have the biomedical applications of GNPs been
narily large longitudinal and transverse relaxivity values extensively explored. Many studies have focused on surface
(r1 = 11.45 mM1 s1 and r2 = 147.3 mM1 s1). A strong UCL plasmon resonance (SPR) which manifests as a strong absorption
signal can be measured by virtue of the high transparency of in the visible (and NIR) region [367]. This phenomenon is attributed
the TaOx shell (Fig. 18). Despite the promises, in vivo targeting with to the collective oscillation of free electrons on the gold particles’
these multimodal probes has not been reported. surfaces and only occurs when metallic particles have a diameter
Overall, although UCNPs are an emerging class of materials with smaller than the light wavelength. With regard to 5 nm spherical
great potential in optical imaging, more development is needed GNPs, the SPR band is centered at 520 nm [368]. The absorption is
before they can be applied to preclinical studies or clinical red-shifted to 517, 521, 533, and 575 nm when the GNP size
diagnosis. Ongoing studies focus on improving the QY and increases to 9, 22, 48, and 99 nm, as shown in Fig. 19a [271,272]. The
biocompatibility of UCNPs as well as the understanding of their GNP size is found to impact the absorption cross-section. In addition
pharmacokinetics and metabolism in vivo. It is also noted that to absorption, light scattering also occurs when GNPs are exposed to
almost all of the imaging data hitherto reported on UCNPs have electromagnetic radiation. A larger particle size favors particle
been acquired on systems customer-adapted to accommodate the scattering and consequently diminishes absorption [369,370]. The

Fig. 18. In vivo UCL images taken after injection of UCNP solutions into a mouse at (a) 0.5 h, (b) 1 h, and (c) 3 h. (d) In situ UCL imaging results taken 3 h after the particle
injection.
Reprinted with permission from ref. [366].
H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69 57

Fig. 19. SPR absorption spectra of (a) spherical GNPs of different sizes, (b) GNRs of different length-to-diameter ratios, (c) GNSs of different thickness, and (d) GNCs.
Reprinted with permission from ref. [367,375,408].

size effect is important when it comes to picking the right spectrum window. However, recent studies show that spherical
nanostructure for a particular application. In optical coherence GNPs can be excited by radio frequency (RF) radiation, suggesting
tomography (OCT), large GNPs are preferred for stronger scattering an alternative means to circumvent the tissue penetration problem
and better signal-to-noise contrast. In GNP-mediated photothermal [376]. For instance, Raoof et al. showed that nonaggregated GNPs
therapy, relatively small GNPs are used to take advantage of their could serve as mediators to convert the RF energy to heat to cause
higher absorption efficiency. The SPR of spherical GNPs is also highly damage [377].
dependent on the inter-particle distance. When GNPs aggregate,
plasmon–plasmon interaction occurs, resulting in a substantial color 3.4.2. Synthesis of gold nanoparticles
change from red to blue [371]. This unique feature has been
harnessed to develop colorimetric detection methods which will be 3.4.2.1. Synthesis of spherical gold nanoparticles. Spherical GNPs
described later in this review. can be prepared in an aqueous solution by reducing HAuCl4. In the
GNP geometry also has a large impact on the SPR. Using gold protocol introduced by Turkevich et al. in 1951 [378], citrate was
nanorods (GNRs) as an example, two plasmon bands are observed. used as the reducing agent to convert Au3+ into Au0. This method is
The first one is the transverse plasmon band that corresponds to still widely used and spherical GNPs with diameters from 5 to
oscillations along the width of the nanorods usually found around 250 nm can be prepared [379,380]. In addition to citrate, reducing
520 nm. The second one is a longitudinal plasmon band caused agents such as sodium borohydride and hydrazine have been
by the oscillations along the length of the GNPs and its position is employed. Different capping ligands have also been exploited that
determined by the nanoparticle length-to-diameter ratio. For passivate the particle surface and prevent particle aggregation
instance, GNRs with a length-to-diameter ratio of 5.2 (length of [381–383].
48 nm and diameter of 9 nm) show a longitudinal band at 920 nm Ultrasmall GNPs can be made by the Brust–Schiffrin method
[372]. By tuning the length-to-diameter ratio of the GNRs, it is which takes places in a two-phase (water–toluene) solution in
possible to achieve a longitudinal plasmon band that matches which AuCl4 is reduced by sodium borohydride with alkanethiols
commercial lasers at 360, 785, and 1064 nm (Fig. 19b). Red-shifts as the capping ligands [384]. The GNPs produced has diameters
have also been observed with gold nanoshells (GNSs) [373] and between 1 and 3 nm. This method has been replaced by a single-
gold nanocages (GNCs), as shown in Fig. 19c and d [374]. The phase approach to prepare GNPs and other noble metal nanocrystals
absorption is determined by the ratio of the thickness to diameter [292,293]. Briefly, the gold precursors are dissolved in toluene along
of the cavity (Fig. 19c). The smaller the ratio, the further the with fatty acids or aliphatic amines. Tetrabutylammonium borohy-
plasmon absorption shifts to the NIR region [375]. dride (TBAB) or a TBAB and hydrazine mixture is used as the
Non-spherical GNPs with NIR absorption are thought to have reducing reagent. Monodispersed spherical GNPs with a diameter
advantages over spherical GNPs in applications such as hyperther- from 1 to 15 nm can be made, and since weak-binding ligands are
mia-based therapy because tissues are more transparent in the NIR employed, the nanocrystals can be readily surface modified.
58 H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69

Concerning the toxicity of the reducing/capping reagents, determines the final thickness of the GNSs, typically ranging from 5
efforts have been made to employ less toxic materials to prepare to 30 nm. By changing the ratio of the core diameter to shell
GNPs. Yin et al. used lactic acid as a reducing agent to produce thickness, the plasmon resonance peak can be tuned from the
GNPs [385]. The size and shape of the GNPs were modulated by visible to mid-infrared spectrum range [398–400].
varying the ratio of the gold precursors and lactic acid. Dubey et al. Zhang’s group used cobalt nanoparticles as templates to grow
produced GNPs using leaf extracts of Rosa rugosa in an aqueous GNSs [401]. The experiments were performed in an anaerobic
media at room temperature [386]. Roy prepared 198Au nanopar- environment to prevent Co oxidation. Independent of the cores, a
ticles by injecting H198AuCl4 into an aqueous biphasic system critical issue is to form a complete and uniform shell otherwise a
containing 2 M Na2SO4 and a 50% (w/w) solution of PEG4000 [387]. broad absorption spectrum is obtained. This approach is repro-
198
Au3+ was quantitatively extracted by the PEG-rich phase ducible, as demonstrated by several independent studies.
without the addition of an extracting agent. After extraction, Recently, Gao et al. developed a new technique to deposit
Au3+ was reduced to Au0 to form spherical GNPs. Sun’s group ultrathin gold layers onto the surface of lipid vesicles and further,
synthesized GNPs in an organic phase by gently reducing to virtually any discrete nanostructure or continuous surface
HAuCl43H2O in OAm which was used as a mild reducing agent [138,402,403]. Using magnetic nanoparticle coating as an example,
as well as surfactant [388,389]. The prepared core/shell nanopar- IONPs with hydrophobic surface ligands are first solubilized into
ticles were coated with a layer of OAm and dispersed well in aqueous solutions using PEGylated phospholipids terminated with
nonpolar solvents. carboxylic groups. A layer of positively charged peptide is then
electrostatically adsorbed onto the surface, which serves as a
3.4.2.2. Synthesis of gold nanorods. GNRs were initially prepared by template for gold deposition. Afterwards, the gold salts are added
electrochemical and photochemical methods [390]. The use of and reduced to form the gold shell. The size of the shell can be
wet-chemical methods to prepare GNRs was pioneered by tuned by adjusting the thickness of the lipid-PEG layer. This is
Murphy’s group [391,392]. In the process, spherical GNPs are achieved by stacking alternative layers of polyallylamine hydro-
used as seeds and suspended in a solution of gold salt, ascorbic acid chloride and sodium polystyrene sulfonate, as shown in Fig. 20.
(AA, as a mild reducing agent), and CTAB (as a surfactant). CTAB is
critical to the GNR synthesis by forming ‘‘soft’’ micellar templates 3.4.2.4. Synthesis of gold nanocages. The synthesis of GNCs was first
for the nanorod growth. CTAB stabilizes the <1 1 0> faces to a reported by Sun et al. in 2002 [404] based on their previous success
greater extent than the <1 0 0> surfaces of the growing GNRs, on making Ag nanocubes [405]. The GNCs were prepared by
resulting in a faster growth rate on the <1 0 0> surface and incubating Ag nanocubes with HAuCl4 in an aqueous solution
consequently one-dimensional growth [393]. The particle aspect [406–408]. Because of the favorable difference in the electrochem-
ratio can be adjusted from 5 to 20 by changing the ratio of the GNP ical potentials between Ag+/Ag (0.80 V) and AuCl4/Au (1.00 V), a
seed to the gold precursor. Jana et al. used silver ions during the galvanic replacement occurred as follows:
seeded-growth procedure [394] and this method was later
 
improved by Nikoobakht and El-Sayed to become an efficient 3Ag ðsÞ þ AuCl4 ðaqÞ ! Au ðsÞ þ 3Agþ ðaqÞ þ 4Cl ðaqÞ (1)
GNR preparation protocol boasting 100% yield and excellent
monodispersity [393]. The aspect ratios could be tailored by A four-stage morphological change with Ag nanocubes was
changing the quantity of Ag+ ions in the solution [395]. observed [406–408] and is explained by the following: (i) initiation
of Ag dissolution (this usually occurs at a site with poor protection
3.4.2.3. Synthesis of gold nanoshells. The synthesis of GNSs was first such as a defect on the side face of a cube with sharp corners or the
reported by the Halas group using silica nanoparticles as templates truncated corner(s) of a cube); (ii) dissolution of bulk Ag from the
[375,396]. A dielectric spherical silica core is grown by the Stöber interior of the particle through the initial site(s) and concurrent
method by reducing TEOS and APTES in ethanol under basic deposition of Au on the rest of the surface; (iii) formation of a
conditions [397] resulting in silica nanoparticles ranging from 50 nanobox (for a sharp cube) or nanocage (for a truncated cube) with
to 500 nm. Small spherical GNPs (1–2 nm) are then adsorbed onto uniform wall thickness due to alloying between Au and Ag; (iv)
the silica surfaces and serve as nucleation sites. For shell formation, dealloying of Ag to generate pores on the walls of the nanobox
HAuCl4 is reduced to Au0 and deposited onto the particle surface, eventually leading to the formation of GNCs (Fig. 21). This
finally coalescing into a complete gold shell. The amount of HAuCl4 technique allows large-scale synthesis of GNCs with a wall

Fig. 20. Schematic illustration of the formation of gold shells.


Reprinted with permission from ref. [138].
H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69 59

usually occurs at the terminal ends. MUA, 3-animo-5-mercapto-


1,2,4-triazole (AMTAZ), and thiolated PEG have been used in GNR
modification [420–423]. Hu and Gao developed a multilayered
coating that endows remarkable stability on GNRs [424]. They first
coated CTAB-capped GNRs with a silica shell. The resulting
hydrophilic GNRs were subjected to a second silica growth process
with octadecyltrimethoxysilane (OTMS) to be rendered hydro-
phobic. Finally, an amphiphilic polymer such as PEGylated
phospholipid was assembled onto the particle surface to make
the particles water soluble. This technique can be extended to
stabilize other nanostructures that are not in the most thermody-
namically or chemically stable states.

3.4.4. Toxicity of gold nanoparticles


One major advantage of GNPs in tumor imaging or therapy is
their good biocompatibility. Previous studies have suggested that
GNPs are either nontoxic or cause little toxicity to cells or small
animals. Recent studies, however, disclosed size-dependent
toxicity with GNPs [425]. Pan et al. systematically studied the
cytotoxicity of triphenylphosphine coated GNPs with sizes from
Fig. 21. Schematic illustration of the formation of GNCs. 0.8 to 15 nm on four cell lines: connective tissue fibroblasts,
Reprinted with permission from ref. [404]. epithelial cells, macrophages, and melanoma cells. It was found
that 1.4 nm GNPs were 60 time more toxic than 15 nm ones. This
thickness accuracy down to 0.5 nm. By adjusting the wall observation indicates variations in the cellular uptake kinetics and/
thickness, the localized surface plasmon resonance peaks can be or target specificities for GNPs of different sizes, and the difference
fine-tuned in the range of 600–1200 nm [409]. The size of GNCs may lead to a different toxicity profile. It is also possible that the
depends largely on the dimension of the Ag nanocage templates, toxicity may stem from interactions with cell membranes but not
which can be varied from 20 to 500 nm. As mentioned earlier, the necessarily after internalization.
size of the GNCs affects the relative magnitude of absorption or Although the gold cores are inert, the coatings may have an
scattering and consequently the efficacy of the GNCs in imaging adverse impact on the biocompatibility [426]. This is particularly
and therapy. true for GNRs because the synthesis usually involves the use of CTAB
The hollow GNCs can be used to load drug molecules of various which is cytotoxic and difficult to get rid of after the synthesis.
kinds. The loading capacity can be tuned by adjusting the volume Studies with CTAB-capped GNRs on human colon carcinoma cells
of the cavity and the pore size on the walls [410]. Xia et al. (HT-29) show apparent cytotoxicity attributable to the free CTAB
evaluated controlled drug release from the GNCs coated with a released from the particle surface to the solution [427]. Methods
layer of thermally sensitive materials such as poly(N-isopropyla- have been developed to mitigate the toxicity of GNRs [428]. For
crylamide) (pNIPAAm) and its derivatives. These materials display example, CTAB-capped GNRs were coated with a layer of
dramatic conformational changes in response to small variations in polyelectrolyte to retard physical desorption of CTAB molecules
temperature. Upon NIR laser irradiation or high-intensity focused [429,430] and the resulting nanoconjugates showed substantially
ultrasound (HIFU), the local temperature is increased to trigger reduced cytotoxicity. Murphy’s group described an on-rod poly-
polymer collapse and release of pre-loaded therapeutics [411,412]. merization method [431]. They replaced the original CTAB coating
Alternatively, controlled drug release can be achieved by filling the with a CTAB analog, 11-(acryloyloxy) undecyltrimethyl ammonium
hollow interior with a phase-change material (PCM) such as 1- bromide (p-CTAB) with a replacement rate of 77  3%. The new
tetradecanol. Upon heating, PCM in the GNCs is melted to induce ligand, p-CTAB, was polymerized in situ with a cationic free radical
drug release into the surroundings [413]. initiator. The GNRs produced were more stable than the CTAB-capped
ones and less cytotoxic [431]. A similar approach was also applied to
3.4.3. Surface modification of gold nanoparticles coat spherical GNPs with some success [432].
On account of the strong interactions between thiols and gold, More biocompatible molecules such as PEG or phospholipids
surface modification and functionality anchoring have been have been proposed to replace the CTAB coating [433]. Takahashi
performed mostly with thiolated compounds [414]. Many peptides et al. extracted CTAB from an aqueous solution of GNRs with
[415], antibodies [416], and small molecule compounds [417] have chloroform that contained phosphatidylcholine. This two-phase
been tethered onto GNP surfaces by taking advantage of the thiol– ligand replacement did not induce particle aggregation but yielded
gold interaction. With regard to in vivo applications, PEGylation is improved biocompatibility [424]. Achieving complete replacement
usually necessary to reduce the non-specific interaction with the of the CTAB coating, however, is challenging.
biological system and improve the particle circulation half-life. A Owing to their relatively low toxicity, GNPs lead other
long circulation time (t1/2 = 17 h) was observed from GNRs coated nanostructures in the area of clinical translation. Clinical trials on
with 5-kDa methyl-PEG-thiol [418] thus permitting efficient EPR- GNS-based photothermal ablation of recurrent head and neck cancer
mediated tumor uptake. When these GNRs were injected at a dose are currently in progress [434]. Another formulation, recombinant
of 20 mg/kg into MDA-MB-435 xenograft mouse models, tumor human tumor necrosis factor-alpha (rhTNF) bearing GNPs, is in
accumulation of 7%ID/g at 72 h p.i. was observed. In general, phase II clinical trial for advanced-stage cancer therapy [435].
GNPs coated with a long PEG chain (e.g. 5 kDa) are more stable than
those coated with a short PEG chain (e.g. 2 kDa). Furthermore, PEG 3.4.5. Gold nanoparticles for cancer diagnosis
with a multidentate-thiol-anchor such as thioctic acid has better
colloidal stability than monothiolated PEG [415–417,419]. 3.4.5.1. Gold nanoparticles for optical coherence tomography imagi-
Due to a thick layer of CTAB, the longitudinal wall of GNRs is ng. Optical coherence tomography (OCT) provides cross-sectional
nearly inaccessible for thiolated species. In this case, the attack tomographic images of the internal microstructure of biological
60 H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69

systems [332,333]. By measuring the backscattered or back- pulses are delivered to the areas-of-interest and by using the
reflected light, OCT generates three-dimensional images of a appropriate probes, some of the energy is absorbed and converted
subject with micrometer resolution and in real-time [436,437]. to heat, leading to thermoelastic expansion and ultrasonic
With a large scattering cross-section and tunable optical resonance emission. The signals are monitored to produce 2D or 3D images
wavelengths, GNPs are good contrast agents for OCT [335,336], and [443,444]. PAT can detect objects embedded at depths as much as
in particular, GNRs, GNSs, and GNCs that have absorption and 7 cm, which is much deeper than possible by fluorescence imaging
scattering in the NIR region are promising [438]. For instance, [445,446]. Because of strong and tunable NIR absorption, GNCs,
GNCs displaying 800 nm SPR have been studied as OCT contrast GNRs, and GNSs are promising contrast agents in this new imaging
agents [439]. Phantom studies found a scattering cross-section of modality. For instance, Eghtedari et al. used GNRs in PA imaging
8.10  1016/m2 and absorption cross-section of 7.26  1015/m2. [447]. They injected 25 mL GNRs at 1.25 pM into nude mice and
Oldenburg et al. evaluated GNRs with maximum SPR at 775 nm detected the signals using a single-channel acoustic transducer. PA
from the viewpoint of OCT imaging [440]. The spectroscopic imaging could monitor the distribution of 100 mL of subcutane-
absorption profile of the GNRs was incorporated into a depth- ously delivered GNRs at 125 pM. Notably, the irradiation can lead
resolved algorithm to map the relative GNR density in the OCT to a photon-induced shape transition of GNRs, mainly a rod-to-
images. As a proof-of-concept demonstration, they successfully sphere conversion [448]. This transition results in a loss of contrast
imaged GNR distributions in excised human breast carcinomas. at the NIR wavelength of interest.
Oldenburg et al. studied GNSs with a silica core of 119 nm diameter Yang et al. demonstrated PEGylated GNCs as intravascular
and a gold shell of 12 nm as OCT contrast agents [375]. Mie contrast agents in PA imaging of the cerebral cortex in a rat model
scattering theory predicts that the GNSs have approximately 67% [449]. Gradual enhancement of the optical absorption in the
of their extinction due to absorption and 33% due to scattering at cerebral cortex up to 81% was observed over the course of the
800 nm. In vivo imaging was performed after PEGylated GNSs had experiment. The same group described the use of GNCs as probes in
been injected into the subcutaneous tumor models. OCT imaging PA mapping of sentinel lymph nodes (SLN) [450], and the method
was performed 20 h later by directly applying the probe over the was able to detect signals from as deep as 33 mm below the skin
tumor skin. Substantial signal increase was observed from the surface with good contrast. This depth is greater than the mean
tumor area, as shown in Fig. 22 [441]. Further laser irradiation led depth of SLNs in human beings, suggesting good clinical potential.
to complete tumor regression as a result of efficient hyperthermia Recently, Wilson et al. developed a novel PA agent composed of
[441,442]. liquid perfluorocarbon (PFC) nanodroplets with encapsulated
GNRs [451]. These droplets used optical absorption to achieve
3.4.5.2. Gold nanoparticles for photoacoustic imaging. Photoacous- the following purposes: (1) triggering the liquid-to-gas transition
tic imaging (PA) is an emerging imaging modality combining high of a nanoscale PFC droplet; (2) production of strong PA signals by
resolution and good optical contrast. In typical PA imaging, laser vaporization; and (3) prolonged thermal expansion signal from the
GNRs. The gaseous phase of the PFC increases the acoustic
impedance mismatch and ultrasound signal. Both in vitro and in
vivo studies reveal that the nanodroplets are efficient contrast
agents in both PA imaging and ultrasound imaging, as indicated in
Fig. 23.

3.4.5.3. Gold nanoparticles for Raman imaging. Surface-enhanced


Raman scattering (SERS) refers to the enhancement of Raman-
active vibrations of molecules when they are in close proximity to a
metal surface [452]. The enhancement is attributed to both
electromagnetic enhancement through excitation of localized
surface plasmons [453] and chemical enhancement via charge-
transfer [454]. It has been shown that the Raman signals can be
enhanced by a factor of 1014–1015 when molecules adsorb on the
GNP surface [455,456], suggesting GNPs as attractive SERS probes.
The degree of enhancement is influenced by the size, shape, and
inter-particle distance. In addition to the high sensitivity, another
reason that makes SERS an appealing method is the multiplexing
capability. Because every molecule has its own spectral fingerprint,
it is possible to image multiple biomarkers simultaneously, which
could be challenging for other imaging modalities [457].
Lee et al. studied antibody-conjugated Au/Ag core–shell
nanoparticles for targeted tumor imaging with live HEK292 cells
expressing PLGg1 [458]. Zong et al. reported a dual mode cancer
cell targeting probe based on CdTe QDs conjugated, silica-coated
Au/Ag core–shell nanorods. The nanorods could generate both
SERS and fluorescence signals. The nanorods were conjugated with
folic acid and used on both HeLa cells and MRC-5 cells, which were
folate receptor positive and negative, respectively. Both SERS and
fluorescence images confirmed the good targeting selectivity, as
shown in Fig. 24 [459]. Jokerst et al. conjugated antibodies onto a
Fig. 22. Representative OCT images from normal skin and muscle tissue areas of gold–silica nanoparticle and evaluated the conjugates as SERS
mice systemically injected with GNSs (A) or with PBS (B). Representative OCT
images from tumors of mice systemically injected with GNSs (C) or with PBS (D). (E)
probes [460]. The nanoconjugates revealed EGFR-positive A431
Histogram graph of results from (A) to (D). tumors with a signal nearly 35 fold larger than that from EGFR-
Reprinted with permission from ref. [441]. negative MDA-435S (human melanoma) tumors. The low-level
H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69 61

Fig. 23. A novel particle-based contrast probe for PA imaging. (a) Diagram depicting the dual-contrast agent concept. (b) Step-by-step diagram of remote activation of the
particle probes. (c) PA images reconstructed using vaporization-based and thermal expansion-based signals captured at one location. (d) Magnitude of pressure transients.
Reprinted with permission from ref. [451].

Fig. 24. (a) SERS signals obtained from 4MBA-labeled GNRs and Au@Ag NRs; (b) SERS spectra of Au@Ag NR@4MBA@PAH and the targeting probe.
Reprinted with permission from ref. [459].
62 H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69

EGFR expression in adjacent healthy tissue is 7 fold smaller than levels of X-ray attenuation by tissues within the body to produce
that observed from the positive tumors. images [462]. Compared to light- or sound-based imaging
Nie’s group reported SERS-based tumor imaging using PEGy- techniques discussed above, CT imaging offers unlimited tissue
lated GNPs [434]. In the nanoconjugates, small molecule Raman penetration and higher spatial resolution. GNPs have attracted
reporters were found to be stabilized, not displaced, by the PEG much attention in this area because their X-ray absorption
layer and the resulting SERS nanoparticles were considerably coefficient is higher than that of iodine (e.g. Ultravist), which is
brighter than QDs in NIR spectrum window. When conjugated with commonly used to improve the contrast [463,464]. Traditionally,
single-chain variable fragment (ScFv) antibodies targeting EGFR, CT is regarded as an anatomical imaging modality and not ideal for
the particles accumulated in tumors and could be detected by a early stage tumor diagnosis. By conjugating with targeting
Raman detection system. In this particular study, small tumors molecules, GNPs may allow CT-based molecular imaging [465].
(0.03 cm3) at a depth of 1–2 cm could be detected. Gambhir’s group Kim et al. compared GNPs and Ultravist as CT contrast agents.
developed MR-PA-SERS tri-modal imaging nanoparticles [461]; When intravenously injected into rats, PEGylated GNPs showed a
essentially a gold–silica-based SERS probe coated with Gd3+ ions. circulation half-life >4 h, which was significantly longer than that of
The probes showed picomolar sensitivity for all three modalities. Ultravist (<10 min). On account of the improved circulation half-life
When intravenously injected into glioblastoma-bearing mice, the and contrast, CT images acquired from rats using PEG-GNPs showed
probes accumulated in the tumor areas but not in the surrounding clearer delineation of the cardiac ventricles and great vessels. When
healthy tissues and were detected tri-modally. Raman imaging they were tested on a hepatoma-bearing rat model, a high contrast
was used as an intraoperative imaging technique that allowed for (2–6 fold) was obtained between hepatoma and normal liver tissue
guidance of tumor resection. The accuracy of Raman imaging was in the CT images, suggesting that PEGylated GNPs are a potential CT
confirmed histologically, as shown in Fig. 25. contrast agent in blood pool imaging and hepatoma imaging
[418,466]. Recently, Reuveni et al. evaluated anti-EGFR conjugated
3.4.5.4. Gold nanoparticles for computed tomography imaging. GNPs GNPs (30 nm) in tumor CT imaging [467]. The GNRs were
also hold potential in CT imaging which relies on the differential intravenously injected into nude mice implanted with human
squamous cell carcinoma head and neck cancer. It was found that
small tumors not detectable by anatomical CT were visible when
using GNP as contrast agents (Fig. 26). However, a relatively large
amount of GNPs need to be injected to induce sufficient contrast on
CT images, and some researchers believe that GNP-based CT imaging
is not practical in real clinical applications [467].

3.4.5.5. Gold nanoparticles-based biosensors. Owing to their unique


optical and electronic properties, GNPs have been intensively
studied in the detection of biomarkers such as DNA and proteins in
vitro. In particular, many efforts have been made to develop
colorimetric biosensors by making use of the distance-dependent
SPR of GNPs. Mirkin’s group first evaluated this idea by using two
batches of 13-nm gold particles capped with thiolated, non-
complementary DNA oligonucleotides [468,469]. When targeting
strands composed of sticky ends complementary to the two grafted
sequences were introduced, cross-linking among particles oc-
curred, resulting in a color change that could be monitored even by
the naked eye. The sensitivity of the method was found to depend
on the particle size and generally, large GNPs (e.g. 50 nm or
100 nm) delivered better sensitivity. They later reported a biochip
by making use of the sharp melting transitions of GNP-labeled DNA
assemblies. Using a sandwich-like assay involving an oligonucleo-
tide-modified glass slide, a GNP-based probe, and targeted DNA,
this method was able to detect single nucleotide mismatches with
high selectivity [470,471]. A limitation of the colorimetric method
is the inherent inability of multiplexing. Mirkin’s group extended
this technology by developing a Raman-based biosensor [472,473].
The GNPs were labeled with both oligonucleotides and Raman-
active dyes. After a silver treatment, the GNPs enabled SERS-based
detection in a multiplex manner with high sensitivity.
GNP-based sensors have also been utilized to analyze protein
targets. Mirkin and co-workers introduced a GNP-based bio-
barcode assay for quantitative protein analysis [474]. Specifically, a
Fig. 25. (a) Two-dimensional axial MRI, PA and Raman images. The post-injection magnetic microparticle conjugated with a detecting antibody and a
images of all three modalities showed clear tumor visualization (dashed boxes
outline the imaged area). (b) A three-dimensional (3D) rendering of MR images with
GNP conjugated with both polyclonal antibodies for the target
the tumor segmented (red; top), an overlay of the three-dimensional PA images protein and oligonucleotides hybridized to barcode strands were
(green) over the MRI (middle) and an overlay of MRI, the segmented tumor and the used in the assay. The magnetic microparticles were applied to the
PA images (bottom) showing good colocalization of the PA signal with the tumor. (c) analyte and addition of GNPs formed a sandwich-like complex if
Quantification of the signals in the tumor showing a significant increase in the MRI,
the target protein was present in the sample. These complexes
PA, and Raman signals after as compared to before the injection.(For interpretation
of the references to color in this figure legend, the reader is referred to the web were magnetically separated and the barcodes were released by
version of the article.) heating. Subsequently, a silver-amplified sandwich assay similar to
Reprinted with permission from ref. [461]. that described previously was performed to quantify the analytes.
H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69 63

Fig. 26. In vivo CT volume-rendered images of (A) mouse before injection of GNPs, (B) mouse 6 h p.i. of non-specific immunoglobulin G conjugated GNPs as a passive targeting
experiment, and (C) mouse 6 h p.i.
Reprinted with permission from ref. [467].

Fig. 27. (A) Absorption profile variation of multifunctional oval-shaped GNPs due to the addition of different cancerous and noncancerous cells. (B) Photograph showing
colorimetric change upon addition of different cancer cells (104 cells/mL). (C) Photograph demonstrating colorimetric change upon the addition of different numbers of SK-
BR-3 cells.
Reprinted with permission from ref. [476].

Multiple proteins at the mid-femtomolar concentrations could be et al. used GNSs to detect immunoglobulins in saline, serum, and
determined [474]. whole blood [475]. Murphy’s group used biotin–avidin as a model
Although spherical GNPs are used predominately, other gold system to show GNR-based particle aggregation and spectrum
nanostructures have been studied in in vitro biosensing. Hirsch shift [475]. Lu et al. reported a highly sensitive two-photon
64 H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69

scattering assay of cancer cells with an oval-shaped GNP Compared to in vivo imaging, nano-based biosensors are more
conjugated with monoclonal anti-HER2/c-erb-2 antibody and S6 likely to gain immediate regulation approval. Unlike imaging, in
RNA aptamer [476]. When oval GNPs were mixed with the breast which nanoparticles are systematically administrated, nano-
cancer SK-BR-3 cell line, a distinct color change occurred and the biosensors are applied in an in vitro setting and do not invoke
two-photon scattering intensity increased by about 13 times complications arising from toxicity and biodistribution. Many
(Fig. 27). studies have demonstrated the higher sensitivity rendered by
GNP-based Raman particles have also been widely used to nano-biosensors compared to conventional clinical tools such as
detect biomarkers in cancer therapy [477]. Sha et al. reported a ELISA. However, whether or not the high sensitivity can be
SERS-based cell detection assay that allowed rapid and direct translated into capability in practice to identify cancer in an early
enumeration of circulating tumor cells (CTCs) down to 50 tumor or pre-syndrome stage requires further clinical development and
cells/mL in whole blood [478]. Recently, Wang et al. reported an corroboration.
EGF-conjugated, peptide-SERS-encoded GNPs for detecting CTCs in
mouse and human blood samples with a high sensitivity of 5– Acknowledgments
50 cells/mL blood [479].
This work was jointly supported by an NCI/NIH R00 grant
4. Conclusion and perspectives (5R00CA153772), a UGA startup grant, Hong Kong Research Grants
Council (RGC) General Research Funds (GRF) No. CityU 112212 and
This article reviews recent developments in the engineering of City University of Hong Kong Applied Research Grant (ARG) No.
nanoparticles used in cancer imaging and diagnosis. With unique 9667066. T. Todd was partially supported by a Philbrook
physical properties, these nanostructures can revolutionize a scholarship.
number of imaging modalities. For instance, magnetic nanopar-
ticles can serve as contrast probes in MRI, and QDs and UCNPs can References
function as probes in fluorescence imaging. Depending on the
nanostructure, GNPs can find applications in OCT imaging, PA [1] K.D. Kochanek, S.E. Kirmeyer, J.A. Martin, D.M. Strobino, B. Guyer, Pediatrics 129
imaging, or CT imaging. Extensive work conducted on small animal (2012) 338–348.
[2] A. Jemal, R. Siegel, J. Xu, E. Ward, CA: A Cancer Journal for Clinicians 60 (2010)
models confirms the advantages of the nanoparticle-based probe. 277–300.
With regard to multimodality imaging, it is possible to integrate [3] A. Jemal, R. Siegel, E. Ward, Y. Hao, J. Xu, T. Murray, M.J. Thun, CA: A Cancer
more than one imaging functionality into a nanoparticle formula- Journal for Clinicians 58 (2008) 71–96.
[4] Y. Jiang, X. Wang, Journal of Hematology and Oncology 5 (2012) 11.
tion and the resulting conjugates can work as reporters for
[5] S. Zhang, C.C. Liu, W. Li, H. Shen, P.W. Laird, X.J. Zhou, Nucleic Acids Research 40
multiple imaging methods. Multimodality imaging is appealing (2012) 9379–9391.
because existing imaging modalities have their own sets of [6] A.N. Bhatt, R. Mathur, A. Farooque, A. Verma, B.S. Dwarakanath, Indian Journal of
advantages and disadvantages. The combined use of more than one Medical Research 132 (2010) 129–149.
[7] W. James, Current Opinion in Pharmacology 1 (2001) 540–546.
imaging system can potentially circumvent the drawbacks of each [8] C.H. Weng, C.J. Huang, G.B. Lee, Sensors (Basel) 12 (2012) 9514–9529.
modality and reduce the chance of misdiagnosis [480]. Other than [9] M. Ferrari, Nature Reviews. Cancer 5 (2005) 161–171.
imaging functionalities, therapeutics can be loaded onto nanoplat- [10] S. Sebastian, J. Settleman, S.J. Reshkin, A. Azzariti, A. Bellizzi, A. Paradiso,
Biochimica et Biophysica Acta 1766 (2006) 120–139.
forms to achieve nano-theranostics and readers are referred to [11] C. Arteaga, Seminars in Oncology 30 (2003) 3–14.
several reviews on this topic [75,481–484]. [12] J. Schlessinger, Cell 103 (2000) 211–225.
There has been significant progress in our ability to prepare [13] N. Normanno, A. De Luca, C. Bianco, L. Strizzi, M. Mancino, M.R. Maiello, A.
Carotenuto, G. De Feo, F. Caponigro, D.S. Salomon, Gene 366 (2006) 2–16.
nanoparticles with controlled size, composition, morphology, and [14] W. Cai, G. Niu, X. Chen, European Journal of Nuclear Medicine and Molecular
physical properties. Their behavior in the biological environment Imaging 35 (2008) 186–208.
depends on many parameters such as the presence of a surface [15] P. Casalini, M.V. Iorio, E. Galmozzi, S. Menard, Journal of Cellular Physiology 200
(2004) 343–350.
coating, targeting molecules, overall particle size, delivery route, [16] G.K. Dy, A.A. Adjei, Cancer 113 (2008) 1857–1887.
and so on. These issues are sometimes intertwined and the [17] P.C. Li, C.R. Wang, D.B. Shieh, C.W. Wei, C.K. Liao, C. Poe, S. Jhan, A.A. Ding, Y.N.
complication underscores the multidisciplinary nature of nano- Wu, Optics Express 16 (2008) 18605–18615.
[18] J.C. Kah, K.W. Kho, C.G. Lee, C. James, R. Sheppard, Z.X. Shen, K.C. Soo, M.C. Olivo,
biotechnology. To achieve bench-to-bed-side translation, it
International Journal of Nanomedicine 2 (2007) 785–798.
requires combined expertise and efforts from researchers in [19] L. Coussens, T.L. Yang-Feng, Y.C. Liao, E. Chen, A. Gray, J. McGrath, P.H. Seeburg,
chemistry, physics, biology, pharmacology, and engineering. In T.A. Libermann, J. Schlessinger, U. Francke, et al. Science 230 (1985) 1132–1139.
particular, attention should be paid to the safety of the [20] M.A. Olayioye, Breast Cancer Research: BCR 3 (2001) 385–389.
[21] Y. Yarden, M.X. Sliwkowski, Nature Reviews. Molecular Cell Biology 2 (2001)
nanomaterials. As novel materials, there are concerns about 127–137.
possible side effects. So far, only IONPs and GNPs have entered [22] H.B. Na, J.H. Lee, K. An, Y.I. Park, M. Park, I.S. Lee, D.H. Nam, S.T. Kim, S.H. Kim, S.W.
clinics/clinical trials in the attempt to validate their biosafety. With Kim, K.H. Lim, K.S. Kim, S.O. Kim, T. Hyeon, Angewandte Chemie (International
ed. in English) 46 (2007) 5397–5401.
regard to other materials such as QDs and UCNPs, more [23] Y.M. Huh, Y.W. Jun, H.T. Song, S. Kim, J.S. Choi, J.H. Lee, S. Yoon, K.S. Kim, J.S. Shin,
investigations are needed to understand their metabolism in vivo J.S. Suh, J. Cheon, Journal of the American Chemical Society 127 (2005) 12387–
as well as long-term impact on a biological system. 12391.
[24] N. Ferrara, Nature Reviews. Cancer 2 (2002) 795–803.
Most in vivo imaging work described in this review serves as [25] N. Ferrara, Endocrine Reviews 25 (2004) 581–611.
proof-of-concept and is performed mostly on subcutaneous animal [26] W. Cai, X. Chen, Frontiers in Bioscience 12 (2007) 4267–4279.
models with a large tumor volume and known overexpression of [27] W. Cai, X. Chen, Journal of Nuclear Medicine 49 (Suppl. 2) (2008) 113S–128S.
[28] N. Ferrara, EXS 94 (2005) 209–231.
certain biomarkers. Future investigations should focus on more
[29] H. Youssoufian, D.J. Hicklin, E.K. Rowinsky, Clinical Cancer Research 13 (2007)
clinically relevant models. Indeed, we expect that nanoparticle- 5544–5548.
based imaging will improve cancer management by providing the [30] R.O. Hynes, Cell 110 (2002) 673–687.
[31] J.D. Hood, D.A. Cheresh, Nature Reviews. Cancer 2 (2002) 91–100.
following features: firstly as aforementioned, providing good
[32] N. Ahmed, C. Riley, G.E. Rice, M.A. Quinn, M.S. Baker, The Journal of Histochem-
detection sensitivity to permit finding tumors in their early stage of istry and Cytochemistry 50 (2002) 1371–1380.
development; secondly, accurate and dynamic monitoring of [33] M.D. Allen, R. Vaziri, M. Green, C. Chelala, A.R. Brentnall, S. Dreger, S. Vallath, H.
patients’ response to treatment; thirdly, simultaneous measure- Nitch-Smith, J. Hayward, R. Carpenter, D.L. Holliday, R.A. Walker, I.R. Hart, J.L.
Jones, The Journal of Pathology 223 (2011) 646–658.
ment of the expression levels of multiple biomarkers in vivo and [34] A.M. Dingemans, V. van den Boogaart, B.A. Vosse, R.J. van Suylen, A.W. Griffioen,
non-invasively. So far, we are still far from achieving these goals. V.L. Thijssen, Molecular Cancer 9 (2010) 152.
H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69 65

[35] N.P. McCabe, S. De, A. Vasanji, J. Brainard, T.V. Byzova, Oncogene 26 (2007) 6238– [74] Y. Li, H. He, X. Jia, W.L. Lu, J. Lou, Y. Wei, Biomaterials 33 (2012) 3899–3908.
6243. [75] S. Mura, P. Couvreur, Advanced Drug Delivery Reviews 64 (2012) 1394–
[36] J.P. Xiong, T. Stehle, R. Zhang, A. Joachimiak, M. Frech, S.L. Goodman, M.A. 1416.
Arnaout, Science 296 (2002) 151–155. [76] R.R. Sawant, A.M. Jhaveri, V.P. Torchilin, Advanced Drug Delivery Reviews 64
[37] C.F. Zhang, M. Jugold, E.C. Woenne, T. Lammers, B. Morgenstern, M.M. Mueller, H. (2012) 1436–1446.
Zentgraf, M. Bock, M. Eisenhut, W. Semmler, F. Kiessling, Cancer Research 67 [77] Z. Liu, S. Tabakman, K. Welsher, H. Dai, Nano Research 2 (2009) 85–120.
(2007) 1555–1562. [78] K. Kostarelos, A. Bianco, M. Prato, Nature Nanotechnology 4 (2009) 627–633.
[38] Z. Wu, Z.B. Li, K. Chen, W. Cai, L. He, F.T. Chin, F. Li, X. Chen, Journal of Nuclear [79] Y. Huang, S. He, W. Cao, K. Cai, X.J. Liang, Nanoscale 4 (2012) 6135–6149.
Medicine 48 (2007) 1536–1544. [80] C.M. Santos, J. Mangadlao, F. Ahmed, A. Leon, R.C. Advincula, D.F. Rodrigues,
[39] H. Elnakat, M. Ratnam, Advanced Drug Delivery Reviews 56 (2004) 1067–1084. Nanotechnology 23 (2012) 395101.
[40] S. Jaracz, J. Chen, L.V. Kuznetsova, I. Ojima, Bioorganic & Medicinal Chemistry 13 [81] H. Shen, L. Zhang, M. Liu, Z. Zhang, Theranostics 2 (2012) 283–294.
(2005) 5043–5054. [82] J. Xie, S. Lee, X. Chen, Advanced Drug Delivery Reviews 62 (2010) 1064–1079.
[41] S.D. Weitman, R.H. Lark, L.R. Coney, D.W. Fort, V. Frasca, V.R. Zurawski Jr., B.A. [83] R. Massart, IEEE Transactions on Magnetics 17 (1981) 1247–1248.
Kamen, Cancer Research 52 (1992) 3396–3401. [84] A. Bee, R. Massart, S. Neveu, Journal of Magnetism and Magnetic Materials 149
[42] P.S. Low, W.A. Henne, D.D. Doorneweerd, Accounts of Chemical Research 41 (1995) 6–9.
(2008) 120–129. [85] L. Babes, B. Denizot, G. Tanguy, J.J. Le Jeune, P. Jallet, Journal of Colloid and
[43] R.L. Scherer, J.O. McIntyre, L.M. Matrisian, Cancer Metastasis Reviews 27 (2008) Interface Science 212 (1999) 474–482.
679–690. [86] A.H. Lu, E.L. Salabas, F. Schuth, Angewandte Chemie (International ed. in English)
[44] M.D. Sternlicht, Z. Werb, Annual Review of Cell and Developmental Biology 17 46 (2007) 1222–1244.
(2001) 463–516. [87] F. Mahtab, Y. Yu, J.W.Y. Lam, J.Z. Liu, B. Zhang, P. Lu, X.X. Zhang, B.Z. Tang,
[45] J.H. McKerrow, V. Bhargava, E. Hansell, S. Huling, T. Kuwahara, M. Matley, L. Advanced Functional Materials 21 (2011) 1733–1740.
Coussens, R. Warren, Molecular Medicine 6 (2000) 450–460. [88] M.S. Sadjadi, F. Fathi, N. Farhadyar, K. Zare, Journal of Nano Research 16 (2011)
[46] B. Davies, J. Waxman, H. Wasan, P. Abel, G. Williams, T. Krausz, D. Neal, D. 43–48.
Thomas, A. Hanby, F. Balkwill, Cancer Research 53 (1993) 5365–5369. [89] B.F. Pan, D.X. Cui, Y. Sheng, C.G. Ozkan, F. Gao, R. He, Q. Li, P. Xu, T. Huang, Cancer
[47] A. Vaisanen, H. Tuominen, M. Kallioinen, T. Turpeenniemi-Hujanen, The Journal Research 67 (2007) 8156–8163.
of Pathology 180 (1996) 283–289. [90] E. Tahmasebi, Y. Yamini, A. Mehdinia, F. Rouhi, Journal of Separation Science 35
[48] L. Zhu, H. Wang, L. Wang, Y. Wang, K. Jiang, C. Li, Q. Ma, S. Gao, W. Li, M. Cai, G. (2012) 2256–2265.
Niu, S. Lee, W. Yang, X. Fang, X. Chen, Journal of Controlled Release 150 (2011) [91] C. Corot, P. Robert, J.M. Idee, M. Port, Advanced Drug Delivery Reviews 58 (2006)
248–255. 1471–1504.
[49] E. Koivunen, W. Arap, H. Valtanen, A. Rainisalo, O.P. Medina, P. Heikkila, C. [92] H.Y. Lee, S.H. Lee, C.J. Xu, J. Xie, J.H. Lee, B. Wu, A.L. Koh, X.Y. Wang, R. Sinclair, S.
Kantor, C.G. Gahmberg, T. Salo, Y.T. Konttinen, T. Sorsa, E. Ruoslahti, R. Pasqua- Xwang, D.G. Nishimura, S. Biswal, S.H. Sun, S.H. Cho, X.Y. Chen, Nanotechnology
lini, Nature Biotechnology 17 (1999) 768–774. 19 (2008) 165101.
[50] S. Lee, E.J. Cha, K. Park, S.Y. Lee, J.K. Hong, I.C. Sun, S.Y. Kim, K. Choi, I.C. Kwon, K. [93] Z. Li, Q. Sun, M. Gao, Angewandte Chemie (International ed. in English) 44 (2004)
Kim, C.H. Ahn, Angewandte Chemie (International ed. in English) 47 (2008) 123–126.
2804–2807. [94] M.G. Harisinghani, K.S. Jhaveri, R. Weissleder, W. Schima, S. Saini, P.F. Hahn, P.R.
[51] J. Xie, F. Zhang, M. Aronova, L. Zhu, X. Lin, Q. Quan, G. Liu, G. Zhang, K.Y. Choi, K. Mueller, Clinical Radiology 56 (2001) 714–725.
Kim, X. Sun, S. Lee, S. Sun, R. Leapman, X. Chen, ACS Nano 5 (2011) 3043–3051. [95] L.H. Deddens, G.A. Van Tilborg, W.J. Mulder, H.E. De Vries, R.M. Dijkhuizen,
[52] R.S. Israeli, C.T. Powell, W.R. Fair, W.D. Heston, Cancer Research 53 (1993) 227– Cerebrovascular Diseases (Basel, Switzerland) 33 (2012) 392–402.
230. [96] H.W. Kang, L. Josephson, A. Petrovsky, R. Weissleder, A. Bogdanov Jr., Bioconju-
[53] D.S. O’Keefe, S.L. Su, D.J. Bacich, Y. Horiguchi, Y. Luo, C.T. Powell, D. Zandvliet, P.J. gate Chemistry 13 (2002) 122–127.
Russell, P.L. Molloy, N.J. Nowak, T.B. Shows, C. Mullins, R.A. Vonder Haar, W.R. [97] S.R. Banerjee, Abstracts of Papers of the American Chemical Society 228 (2004)
Fair, W.D. Heston, Biochimica et Biophysica Acta 1443 (1998) 113–127. U31.
[54] J.S. Horoszewicz, E. Kawinski, G.P. Murphy, Anticancer Research 7 (1987) 927– [98] A.N. Glazer, Nature 381 (1996) 290.
935. [99] M.N. Gupta, Abstracts of Papers of the American Chemical Society 202 (1991)
[55] W.C. Olson, W.D. Heston, A.K. Rajasekaran, Reviews on Recent Clinical Trials 2 220–230.
(2007) 182–190. [100] C. Tassa, S.Y. Shaw, R. Weissleder, Accounts of Chemical Research 44 (2011) 842–
[56] O.C. Farokhzad, S. Jon, A. Khademhosseini, T.N. Tran, D.A. Lavan, R. Langer, Cancer 852.
Research 64 (2004) 7668–7672. [101] R. Hao, R. Xing, Z. Xu, Y. Hou, S. Gao, S. Sun, Advanced Materials 22 (2010) 2729–
[57] O.C. Farokhzad, J. Cheng, B.A. Teply, I. Sherifi, S. Jon, P.W. Kantoff, J.P. Richie, R. 2742.
Langer, Proceedings of the National Academy of Sciences of the United States of [102] T. Hyeon, Chemical Communications (8) (2003) 927–934.
America 103 (2006) 6315–6320. [103] M. Green, Chemical Communications (24) (2005) 3002–3011.
[58] B. Verhoven, R.A. Schlegel, P. Williamson, The Journal of Experimental Medicine [104] U. Jeong, X.W. Teng, Y. Wang, H. Yang, Y.N. Xia, Advanced Materials 19 (2007)
182 (1995) 1597–1601. 33–60.
[59] D. Schilling, M. Gehrmann, C. Steinem, A. De Maio, A.G. Pockley, M. Abend, M. [105] J. Park, E. Lee, N.M. Hwang, M. Kang, S.C. Kim, Y. Hwang, J.G. Park, H.J. Noh, J.Y.
Molls, G. Multhoff, The FASEB Journal 23 (2009) 2467–2477. Kim, J.H. Park, T. Hyeon, Angewandte Chemie (International ed. in English) 44
[60] S. Ran, P.E. Thorpe, International Journal of Radiation Oncology, Biology, Physics (2005) 2872–2877.
54 (2002) 1479–1484. [106] T. Hyeon, S.S. Lee, J. Park, Y. Chung, H. Bin Na, Journal of the American Chemical
[61] H.P. Dong, A. Holth, L. Kleinberg, M.G. Ruud, M.B. Elstrand, C.G. Trope, B. Society 123 (2001) 12798–12801.
Davidson, B. Risberg, American Journal of Clinical Pathology 132 (2009) 756– [107] J. Park, K.J. An, Y.S. Hwang, J.G. Park, H.J. Noh, J.Y. Kim, J.H. Park, N.M. Hwang, T.
762. Hyeon, Nature Materials 3 (2004) 891–895.
[62] H. Kenis, C. Reutelingsperger, Current Pharmaceutical Design 15 (2009) 2719– [108] N.R. Jana, Y.F. Chen, X.G. Peng, Chemistry of Materials 16 (2004) 3931–3935.
2723. [109] K. Woo, J. Hong, J.P. Ahn, Journal of Magnetism and Magnetic Materials 293
[63] G. Malik, M.D. Ward, S.K. Gupta, M.W. Trosset, W.E. Grizzle, B.L. Adam, J.I. Diaz, (2005) 177–181.
O.J. Semmes, Clinical Cancer Research 11 (2005) 1073–1085. [110] S. Sun, H. Zeng, D.B. Robinson, S. Raoux, P.M. Rice, S.X. Wang, G. Li, Journal of the
[64] K. Ueda, Y. Fukase, T. Katagiri, N. Ishikawa, S. Irie, T.A. Sato, H. Ito, H. Nakayama, Y. American Chemical Society 126 (2004) 273–279.
Miyagi, E. Tsuchiya, N. Kohno, M. Shiwa, Y. Nakamura, Y. Daigo, Proteomics 9 [111] H. Khurshid, W.F. Li, M.H. Phan, P. Mukherjee, G.C. Hadjipanayis, H. Srikanth,
(2009) 2182–2192. Applied Physics Letters 101 (2012) 232405.
[65] C. Zhou, M. Long, Y. Qin, X. Sun, J. Zheng, Angewandte Chemie (International ed. [112] Y.W. Jun, Y.M. Huh, J.S. Choi, J.H. Lee, H.T. Song, S. Kim, S. Yoon, K.S. Kim, J.S. Shin,
in English) 50 (2011) 3168–3172. J.S. Suh, J. Cheon, Journal of the American Chemical Society 127 (2005) 5732–
[66] H.S. Choi, W. Liu, P. Misra, E. Tanaka, J.P. Zimmer, B. Itty Ipe, M.G. Bawendi, J.V. 5733.
Frangioni, Nature Biotechnology 25 (2007) 1165–1170. [113] J.T. Jang, H. Nah, J.H. Lee, S.H. Moon, M.G. Kim, J. Cheon, Angewandte Chemie
[67] M.M. Cheng, G. Cuda, Y.L. Bunimovich, M. Gaspari, J.R. Heath, H.D. Hill, C.A. (International ed. in English) 48 (2009) 1234–1238.
Mirkin, A.J. Nijdam, R. Terracciano, T. Thundat, M. Ferrari, Current Opinion in [114] C.B. Murray, S.H. Sun, H. Doyle, T. Betley, MRS Bulletin 26 (2001) 985–991.
Chemical Biology 10 (2006) 11–19. [115] K. Butter, K. Kassapidou, G.J. Vroege, A.P. Philipse, Journal of Colloid and Interface
[68] H.Y. Lee, Z. Li, K. Chen, A.R. Hsu, C. Xu, J. Xie, S. Sun, X. Chen, Journal of Nuclear Science 287 (2005) 485–495.
Medicine 49 (2008) 1371–1379. [116] K. Butter, A.P. Philipse, G.J. Vroege, Journal of Magnetism and Magnetic Materials
[69] X. Montet, K. Montet-Abou, F. Reynolds, R. Weissleder, L. Josephson, Neoplasia 8 252 (2002) 1–3.
(2006) 214–222. [117] S. Peng, C. Wang, J. Xie, S. Sun, Journal of the American Chemical Society 128
[70] J.H. Lee, K. Lee, S.H. Moon, Y. Lee, T.G. Park, J. Cheon, Angewandte Chemie (2006) 10676–10677.
(International ed. in English) 48 (2009) 4174–4179. [118] L.M. Lacroix, N.F. Huls, D. Ho, X. Sun, K. Cheng, S. Sun, Nano Letters 11 (2011)
[71] J. Cheon, J.H. Lee, Accounts of Chemical Research 41 (2008) 1630–1640. 1641–1645.
[72] J. Du, W.L. Lu, X. Ying, Y. Liu, P. Du, W. Tian, Y. Men, J. Guo, Y. Zhang, R.J. Li, J. Zhou, J.N. [119] Q. Song, Z.J. Zhang, Journal of the American Chemical Society 126 (2004) 6164–
Lou, J.C. Wang, X. Zhang, Q. Zhang, Molecular Pharmaceutics 6 (2009) 905–917. 6168.
[73] X. Ying, H. Wen, W.L. Lu, J. Du, J. Guo, W. Tian, Y. Men, Y. Zhang, R.J. Li, T.Y. Yang, [120] V.F. Puntes, D. Zanchet, C.K. Erdonmez, A.P. Alivisatos, Journal of the American
D.W. Shang, J.N. Lou, L.R. Zhang, Q. Zhang, Journal of Controlled Release 141 Chemical Society 124 (2002) 12874–12880.
(2010) 183–192. [121] V.F. Puntes, K.M. Krishnan, A.P. Alivisatos, Science 291 (2001) 2115–2117.
66 H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69

[122] H. Bonnemann, W. Brijoux, R. Brinkmann, N. Matoussevitch, N. Waldofner, N. [166] O. Veiseh, C. Sun, C. Fang, N. Bhattarai, J. Gunn, F. Kievit, K. Du, B. Pullar, D. Lee,
Palina, H. Modrow, Inorganic Chimica Acta 350 (2003) 617–624. R.G. Ellenbogen, J. Olson, M. Zhang, Cancer Research 69 (2009) 6200–6207.
[123] C. Yang, H. Zhao, Y. Hou, D. Ma, Journal of the American Chemical Society 134 [167] O. Veiseh, C. Sun, J. Gunn, N. Kohler, P. Gabikian, D. Lee, N. Bhattarai, R.
(2012) 15814–15821. Ellenbogen, R. Sze, A. Hallahan, J. Olson, M.Q. Zhang, Nano Letters 5 (2005)
[124] W.S. Seo, J.H. Lee, X.M. Sun, Y. Suzuki, D. Mann, Z. Liu, M. Terashima, P.C. Yang, 1003–1008.
M.V. McConnell, D.G. Nishimura, H.J. Dai, Nature Materials 5 (2006) 971–976. [168] C. Leuschner, C.S. Kumar, W. Hansel, W. Soboyejo, J. Zhou, J. Hormes, Breast
[125] J. Kim, Y. Lee, S.H. Sun, Journal of the American Chemical Society 132 (2010) Cancer Research and Treatment 99 (2006) 163–176.
4996–4997. [169] G.R. Reddy, M.S. Bhojani, P. McConville, J. Moody, B.A. Moffat, D.E. Hall, G. Kim,
[126] M.X. Wan, J.C. Li, Journal of Polymer Science Part A: Polymer Chemistry 36 Y.E. Koo, M.J. Woolliscroft, J.V. Sugai, T.D. Johnson, M.A. Philbert, R. Kopelman, A.
(1998) 2799–2805. Rehemtulla, B.D. Ross, Clinical Cancer Research 12 (2006) 6677–6686.
[127] M.D. Butterworth, S.A. Bell, S.P. Armes, A.W. Simpson, Journal of Colloid and [170] S. Lee, J. Xie, X. Chen, Chemical Reviews 110 (2010) 3087–3111.
Interface Science 183 (1996) 91–99. [171] J.H. Lee, Y.M. Huh, Y.W. Jun, J.W. Seo, J.T. Jang, H.T. Song, S. Kim, E.J. Cho, H.G.
[128] P. Tartaj, M.P. Morales, T. Gonzalez-Carreno, S. Veintemillas-Verdaguer, C.J. Yoon, J.S. Suh, J. Cheon, Nature Medicine 13 (2007) 95–99.
Serna, Journal of Magnetism and Magnetic Materials 290 (2005) 28–34. [172] C.G. Hadjipanayis, R. Machaidze, M. Kaluzova, L. Wang, A.J. Schuette, H. Chen, X.
[129] T.R. Zhang, J.P. Ge, Y.P. Hu, Y.D. Yin, Nano Letters 7 (2007) 3203–3207. Wu, H. Mao, Cancer Research 70 (2010) 6303–6312.
[130] G. Huang, C.F. Zhang, S.Z. Li, C. Khemtong, S.G. Yang, R.H. Tian, J.D. Minna, K.C. [173] W. Hwang do, H.Y. Ko, J.H. Lee, H. Kang, S.H. Ryu, I.C. Song, D.S. Lee, S. Kim,
Brown, J.M. Gao, Journal of Material Chemistry 19 (2009) 6367–6372. Journal of Nuclear Medicine 51 (2010) 98–105.
[131] C.J. Xu, K.M. Xu, H.W. Gu, R.K. Zheng, H. Liu, X.X. Zhang, Z.H. Guo, B. Xu, Journal of [174] H. Shao, C. Min, D. Issadore, M. Liong, T.J. Yoon, R. Weissleder, H. Lee, Ther-
the American Chemical Society 126 (2004) 9938–9939. anostics 2 (2012) 55–65.
[132] J. Xie, K. Chen, H.Y. Lee, C.J. Xu, A.R. Hsu, S. Peng, X.Y. Chen, S.H. Sun, Journal of the [175] H. Lee, E. Sun, D. Ham, R. Weissleder, Nature Medicine 14 (2008) 869–874.
American Chemical Society 130 (2008) 7542–7543. [176] J.B. Haun, C.M. Castro, R. Wang, V.M. Peterson, B.S. Marinelli, H. Lee, R. Weissle-
[133] J. Xie, J.H. Wang, G. Niu, J. Huang, K. Chen, X.G. Li, X.Y. Chen, Chemical der, Science Translational Medicine 3 (2011) 71ra16.
Communications 46 (2010) 433–435. [177] C. Tassa, M. Liong, S. Hilderbrand, J.E. Sandler, T. Reiner, E.J. Keliher, R. Weissle-
[134] V.P. Torchilin, V.S. Trubetskoy, Advanced Drug Delivery Reviews 16 (1995) 141– der, S.Y. Shaw, Lab on a Chip 12 (2012) 3103–3110.
155. [178] N.K. Devaraj, S. Hilderbrand, R. Upadhyay, R. Mazitschek, R. Weissleder, Ange-
[135] T. Pellegrino, L. Manna, S. Kudera, T. Liedl, D. Koktysh, A.L. Rogach, S. Keller, J. wandte Chemie (International ed. in English) 49 (2010) 2869–2872.
Radler, G. Natile, W.J. Parak, Nano Letters 4 (2004) 703–707. [179] J.B. Haun, N.K. Devaraj, S.A. Hilderbrand, H. Lee, R. Weissleder, Nature Nano-
[136] K.L. Hultman, A.J. Raffo, A.L. Grzenda, P.E. Harris, T.R. Brown, S. O’Brien, ACS Nano technology 5 (2010) 660–665.
2 (2008) 477–484. [180] S.J. Osterfeld, H. Yu, R.S. Gaster, S. Caramuta, L. Xu, S.J. Han, D.A. Hall, R.J. Wilson,
[137] S. Tong, S.J. Hou, B.B. Ren, Z.L. Zheng, G. Bao, Nano. Lett. 11 (2011) 3720–3726. S. Sun, R.L. White, R.W. Davis, N. Pourmand, S.X. Wang, Proceedings of the
[138] Y.D. Jin, C.X. Jia, S.W. Huang, M. O’Donnell, X.H. Gao, Nature Communications 1 National Academy of Sciences of the United States of America 105 (2008)
(2010) 41. 20637–20640.
[139] L.E. LaConte, N. Nitin, O. Zurkiya, D. Caruntu, C.J. O’Connor, X. Hu, G. Bao, Journal [181] R.S. Gaster, D.A. Hall, C.H. Nielsen, S.J. Osterfeld, H. Yu, K.E. Mach, R.J. Wilson, B.
of Magnetic Resonance Imaging 26 (2007) 1634–1641. Murmann, J.C. Liao, S.S. Gambhir, S.X. Wang, Nature Medicine 15 (2009) 1327–
[140] S.H. Hu, Y.Y. Chen, T.C. Liu, T.H. Tung, D.M. Liu, S.Y. Chen, Chemical 1332.
Communications 47 (2011) 1776–1778. [182] R.S. Gaster, L. Xu, S.J. Han, R.J. Wilson, D.A. Hall, S.J. Osterfeld, H. Yu, S.X. Wang,
[141] D.K. Yi, S.S. Lee, G.C. Papaefthymiou, J.Y. Ying, Chemistry of Materials 18 (2006) Nature Nanotechnology 6 (2011) 314–320.
614–619. [183] J.L. Kovar, M.A. Simpson, A. Schutz-Geschwender, D.M. Olive, Analytical Bio-
[142] D.K. Yi, S.T. Selvan, S.S. Lee, G.C. Papaefthymiou, D. Kundaliya, J.Y. Ying, Journal of chemistry 367 (2007) 1–12.
the American Chemical Society 127 (2005) 4990–4991. [184] A.P. Alivisatos, The Journal of Physical Chemistry 100 (1996) 13226–13239.
[143] J. Kim, H.S. Kim, N. Lee, T. Kim, H. Kim, T. Yu, I.C. Song, W.K. Moon, T. Hyeon, [185] R. Rossetti, S. Nakahara, L.E. Brus, The Journal of Chemical Physics 79 (1983)
Angewandte Chemie (International ed. in English) 47 (2008) 8438–8441. 1086–1088.
[144] R. De Palma, S. Peeters, M.J. Van Bael, H. Van den Rul, K. Bonroy, W. Laureyn, J. [186] S. Baskoutas, A.F. Terzis, Journal of Applied Physics 99 (2006) 013708.
Mullens, G. Borghs, G. Maes, Chemistry of Materials 19 (2007) 1821–1831. [187] S. Neeleshwar, C.L. Chen, C.B. Tsai, Y.Y. Chen, C.C. Chen, S.G. Shyu, M.S. Seehra,
[145] J.E. Lee, N. Lee, T. Kim, J. Kim, T. Hyeon, Accounts of Chemical Research 44 (2011) Physical Review B 71 (2005) 201307.
893–902. [188] M. Sharma, A.B. Sharma, N. Mishra, R.K. Pandey, Materials Research Bulletin 46
[146] T. Kim, E. Momin, J. Choi, K. Yuan, H. Zaidi, J. Kim, M. Park, N. Lee, M.T. McMahon, (2011) 453–459.
A. Quinones-Hinojosa, J.W.M. Bulte, T. Hyeon, A.A. Gilad, Journal of the American [189] S. Mathew, A.D. Saran, B.S. Bhardwaj, S.A. Joseph, P. Radhakrishnan, V.P.N.
Chemical Society 133 (2011) 2955–2961. Nampoori, C.P.G. Vallabhan, J.R. Bellare, Journal of Applied Physics 111
[147] J. Park, K. An, Y. Hwang, J.G. Park, H.J. Noh, J.Y. Kim, J.H. Park, N.M. Hwang, T. (2012) 013708.
Hyeon, Nature Materials 3 (2004) 891–895. [190] D.J. Norris, M.G. Bawendi, Physical Review B 53 (1996) 16338–16346.
[148] Z.P. Zhen, J. Xie, Theranostics 2 (2012) 45–54. [191] J. Jasieniak, L. Smith, J. van Embden, P. Mulvaney, M. Califano, The Journal of
[149] J.M. Shin, R.M. Anisur, M.K. Ko, G.H. Im, J.H. Lee, I.S. Lee, Angewandte Chemie Physical Chemistry. C 113 (2009) 19468–19474.
(International ed. in English) 48 (2009) 321–324. [192] F.C. Liu, Y.M. Chen, J.H. Lin, W.L. Tseng, Journal of Colloid and Interface Science
[150] J. Huang, J. Xie, K. Chen, L. Bu, S. Lee, Z. Cheng, X. Li, X. Chen, Chemical 337 (2009) 414–419.
Communications 46 (2010) 6684–6686. [193] B.O. Dabbousi, J. RodriguezViejo, F.V. Mikulec, J.R. Heine, H. Mattoussi, R. Ober,
[151] K.M.L. Taylor, A. Jin, W.B. Lin, Angewandte Chemie (International ed. in English) K.F. Jensen, M.G. Bawendi, The Journal of Physical Chemistry. B 101 (1997)
47 (2008) 7722–7725. 9463–9475.
[152] K.M.L. Taylor, W.J. Rieter, W.B. Lin, Journal of the American Chemical Society 130 [194] X.G. Peng, M.C. Schlamp, A.V. Kadavanich, A.P. Alivisatos, Journal of the Ameri-
(2008) 14358–14359. can Chemical Society 119 (1997) 7019–7029.
[153] W.J. Rieter, K.M. Taylor, W. Lin, Journal of the American Chemical Society 129 [195] W.C. Chan, D.J. Maxwell, X. Gao, R.E. Bailey, M. Han, S. Nie, Current Opinion in
(2007) 9852–9853. Biotechnology 13 (2002) 40–46.
[154] P. Horcajada, T. Chalati, C. Serre, B. Gillet, C. Sebrie, T. Baati, J.F. Eubank, D. [196] I.L. Medintz, H.T. Uyeda, E.R. Goldman, H. Mattoussi, Nature Materials 4 (2005)
Heurtaux, P. Clayette, C. Kreuz, J.S. Chang, Y.K. Hwang, V. Marsaud, P.N. Bories, L. 435–446.
Cynober, S. Gil, G. Ferey, P. Couvreur, R. Gref, Nature Materials 9 (2010) 172–178. [197] A. Franceschetti, A. Zunger, Physical Review Letters 78 (1997) 915–918.
[155] S. Geninatti Crich, B. Bussolati, L. Tei, C. Grange, G. Esposito, S. Lanzardo, G. [198] M. Nirmal, B.O. Dabbousi, M.G. Bawendi, J.J. Macklin, J.K. Trautman, T.D. Harris,
Camussi, S. Aime, Cancer Research 66 (2006) 9196–9201. L.E. Brus, Nature 383 (1996) 802–804.
[156] S. Aime, L. Frullano, S. Geninatti Crich, Angewandte Chemie (International ed. in [199] F. Pinaud, X. Michalet, L.A. Bentolila, J.M. Tsay, S. Doose, J.J. Li, G. Iyer, S. Weiss,
English) 41 (2002) 1017–1019. Biomaterials 27 (2006) 1679–1687.
[157] J.J. Yin, F. Lao, P.P. Fu, W.G. Wamer, Y. Zhao, P.C. Wang, Y. Qiu, B. Sun, G. Xing, J. [200] P. Zrazhevskiy, M. Sena, X. Gao, Chemical Society Reviews 39 (2010) 4326–4354.
Dong, X.J. Liang, C. Chen, Biomaterials 30 (2009) 611–621. [201] L. Qu, X. Peng, Journal of the American Chemical Society 124 (2002) 2049–2055.
[158] J.J. Yin, F. Lao, J. Meng, P.P. Fu, Y. Zhao, G. Xing, X. Gao, B. Sun, P.C. Wang, C. Chen, [202] U. Resch-Genger, M. Grabolle, S. Cavaliere-Jaricot, R. Nitschke, T. Nann, Nature
X.J. Liang, Molecular Pharmacology 74 (2008) 1132–1140. Methods 5 (2008) 763–775.
[159] C.Y. Shu, F.D. Corwin, J.F. Zhang, Z.J. Chen, J.E. Reid, M.H. Sun, W. Xu, J.H. Sim, C.R. [203] X. Wu, H. Liu, J. Liu, K.N. Haley, J.A. Treadway, J.P. Larson, N. Ge, F. Peale, M.P.
Wang, P.P. Fatouros, A.R. Esker, H.W. Gibson, H.C. Dorn, Bioconjugate Chemistry Bruchez, Nature Biotechnology 21 (2003) 41–46.
20 (2009) 1186–1193. [204] X.H. Gao, L.L. Yang, J.A. Petros, F.F. Marshal, J.W. Simons, S.M. Nie, Current
[160] K. Chen, J. Xie, H.Y. Xu, D. Behera, M.H. Michalski, S. Biswal, A. Wang, X.Y. Chen, Opinion in Biotechnology 16 (2005) 63–72.
Biomaterials 30 (2009) 6912–6919. [205] R. Zeng, T. Zhang, J. Liu, S. Hu, Q. Wan, X. Liu, Z. Peng, B. Zou, Nanotechnology 20
[161] K. Chen, Z.B. Li, H. Wang, W.B. Cai, X.Y. Chen, European Journal of Nuclear (2009) 095102.
Medicine and Molecular Imaging 35 (2008) 2235–2244. [206] V. Lesnyak, A. Dubavik, A. Plotnikov, N. Gaponik, A. Eychmuller, Chemical
[162] X. Montet, R. Weissleder, L. Josephson, Bioconjugate Chemistry 17 (2006) 905– Communications 46 (2010) 886–888.
911. [207] Y.G. Zheng, Z.C. Yang, J.Y. Ying, Advanced Materials 19 (2007) 1475–1479.
[163] G. Niu, X.Y. Chen, Theranostics 1 (2011) 30–47. [208] W.C. Law, K.T. Yong, I. Roy, H. Ding, R. Hu, W.W. Zhao, P.N. Prasad, Small 5 (2009)
[164] X.Y. Chen, Theranostics 1 (2011) 28–29. 1302–1310.
[165] C. Sun, O. Veiseh, J. Gunn, C. Fang, S. Hansen, D. Lee, R. Sze, R.G. Ellenbogen, J. [209] C.B. Murray, D.J. Norris, M.G. Bawendi, Journal of the American Chemical Society
Olson, M. Zhang, Small 4 (2008) 372–379. 115 (1993) 8706–8715.
H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69 67

[210] Z.A. Peng, X. Peng, Journal of the American Chemical Society 123 (2001) 183– [260] W. Cai, D.W. Shin, K. Chen, O. Gheysens, Q. Cao, S.X. Wang, S.S. Gambhir, X. Chen,
184. Nano Letters 6 (2006) 669–676.
[211] X.G. Peng, Advanced Materials 15 (2003) 459–463. [261] J. Gao, K. Chen, R. Luong, D.M. Bouley, H. Mao, T. Qiao, S.S. Gambhir, Z. Cheng,
[212] Z.A. Peng, X.G. Peng, Journal of the American Chemical Society 123 (2001) 1389– Nano Letters 12 (2012) 281–286.
1395. [262] J. Gao, K. Chen, Z. Miao, G. Ren, X. Chen, S.S. Gambhir, Z. Cheng, Biomaterials 32
[213] A.M. Smith, S.M. Nie, Accounts of Chemical Research 43 (2010) 190–200. (2011) 2141–2148.
[214] A.P. Alivisatos, Science 271 (1996) 933–937. [263] B.R. Smith, Z. Cheng, A. De, A.L. Koh, R. Sinclair, S.S. Gambhir, Nano Letters 8
[215] S. Kim, B. Fisher, H.J. Eisler, M. Bawendi, Journal of the American Chemical (2008) 2599–2606.
Society 125 (2003) 11466–11467. [264] B.R. Smith, Z. Cheng, A. De, J. Rosenberg, S.S. Gambhir, Small 6 (2010) 2222–
[216] J.M. Pietryga, R.D. Schaller, D. Werder, M.H. Stewart, V.I. Klimov, J.A. Hollings- 2229.
worth, Journal of the American Chemical Society 126 (2004) 11752–11753. [265] W. Cai, K. Chen, Z.B. Li, S.S. Gambhir, X. Chen, Journal of Nuclear Medicine 48
[217] A.M. Smith, X. Gao, S. Nie, Photochemistry and Photobiology 80 (2004) 377–385. (2007) 1862–1870.
[218] G. Zlateva, Z. Zhelev, R. Bakalova, I. Kanno, Inorganic Chemistry 46 (2007) 6212– [266] J. Probst, S. Dembski, M. Milde, S. Rupp, Expert Review of Molecular Diagnostics
6214. 12 (2012) 49–64.
[219] X. Zhong, Y. Feng, W. Knoll, M. Han, Journal of the American Chemical Society [267] A.A. Ghazani, J.A. Lee, J. Klostranec, Q. Xiang, R.S. Dacosta, B.C. Wilson, M.S. Tsao,
125 (2003) 13559–13563. W.C. Chan, Nano Letters 6 (2006) 2881–2886.
[220] Q. Ma, X. Su, Analyst 135 (2010) 1867–1877. [268] M.V. Yezhelyev, A. Al-Hajj, C. Morris, A.I. Marcus, T. Liu, M. Lewis, C. Cohen, P.
[221] S. Hinds, S. Myrskog, L. Levina, G. Koleilat, J. Yang, S.O. Kelley, E.H. Sargent, Zrazhevskiy, J.W. Simons, A. Rogatko, S. Nie, X. Gao, R.M. O’Regan, Advanced
Journal of the American Chemical Society 129 (2007) 7218–7219. Materials 19 (2007) 3146–3151.
[222] P.M. Allen, M.G. Bawendi, Journal of the American Chemical Society 130 (2008) [269] J. Liu, S.K. Lau, V.A. Varma, B.A. Kairdolf, S. Nie, Analytical Chemistry 82 (2010)
9240–9241. 6237–6243.
[223] H. Wang, S. Lou, Z. Tang, W. Xu, H. Shang, H. Shen, L.S. Li, Dalton Transactions 41 [270] J. Xu, S. Muller, S. Nannapaneni, L. Pan, Y. Wang, X. Peng, D. Wang, M. Tighiouart,
(2012) 12726–12732. Z. Chen, N.F. Saba, J.J. Beitler, D.M. Shin, Z.G. Chen, European Journal of Cancer 48
[224] M. Wang, C.C. Mi, Y.X. Zhang, J.L. Liu, F. Li, C.B. Mao, S.K. Xu, The Journal of (2012) 1682–1691.
Physical Chemistry. C 113 (2009) 19021–19027. [271] J.C. Boyer, F.C. van Veggel, Nanoscale 2 (2010) 1417–1419.
[225] R. Xie, X. Peng, Journal of the American Chemical Society 131 (2009) 10645– [272] F. Wang, X. Liu, Chemical Society Reviews 38 (2009) 976–989.
10651. [273] J. Zhou, Z. Liu, F. Li, Chemical Society Reviews 41 (2012) 1323–1349.
[226] X.H. Zhong, Z.H. Zhang, S.H. Liu, M.Y. Han, W. Knoll, The Journal of Physical [274] M. Wang, G. Abbineni, A. Clevenger, C. Mao, S. Xu, Nanomedicine 7 (2011) 710–
Chemistry. B 108 (2004) 15552–15559. 729.
[227] M. Protiere, P. Reiss, Small 3 (2007) 399–403. [275] L.Y. Ang, M.E. Lim, L.C. Ong, Y. Zhang, Nanomedicine 6 (2011) 1273–1288.
[228] X.H. Zhong, Y.Y. Feng, W. Knoll, M.Y. Han, Journal of the American Chemical [276] O. Ehlert, R. Thomann, M. Darbandi, T. Nann, ACS Nano 2 (2008) 120–124.
Society 125 (2003) 13559–13563. [277] X.C. Ye, J.E. Collins, Y.J. Kang, J. Chen, D.T.N. Chen, A.G. Yodh, C.B. Murray,
[229] A.M. Smith, A.M. Mohs, S. Nie, Nature Nanotechnology 4 (2009) 56–63. Proceedings of the National Academy of Sciences of the United States of America
[230] W.C. Chan, S. Nie, Science 281 (1998) 2016–2018. 107 (2010) 22430–22435.
[231] X. Yu, L. Chen, Y. Deng, K. Li, Q. Wang, Y. Li, S. Xiao, L. Zhou, X. Luo, J. Liu, D. Pang, [278] F. Wang, X. Liu, Journal of the American Chemical Society 130 (2008) 5642–
Journal of Fluorescence 17 (2007) 243–247. 5643.
[232] W.C. Law, K.T. Yong, I. Roy, H. Ding, R. Hu, W. Zhao, P.N. Prasad, Small 5 (2009) [279] F. Wang, J.A. Wang, X.G. Liu, Angewandte Chemie (International ed. in English)
1302–1310. 49 (2010) 7456–7460.
[233] J. Aldana, Y.A. Wang, X. Peng, Journal of the American Chemical Society 123 [280] X. Bai, H.W. Song, G.H. Pan, Y.Q. Lei, T. Wang, X.G. Ren, S.Z. Lu, B. Dong, Q.L. Dai, L.
(2001) 8844–8850. Fan, The Journal of Physical Chemistry. C 111 (2007) 13611–13617.
[234] W.R. Algar, U.J. Krull, Chemphyschem 8 (2007) 561–568. [281] S.G. Xiao, X.L. Yang, Modern Physics Letters B 25 (2011) 265–272.
[235] W. Liu, M. Howarth, A.B. Greytak, Y. Zheng, D.G. Nocera, A.Y. Ting, M.G. Bawendi, [282] G.K. Liu, H.Z. Zhuang, X.Y. Chen, Nano Letters 2 (2002) 535–539.
Journal of the American Chemical Society 130 (2008) 1274–1284. [283] S. Schietinger, S. Menezes Lde, B. Lauritzen, O. Benson, Nano Letters 9 (2009)
[236] A. Sukhanova, J. Devy, L. Venteo, H. Kaplan, M. Artemyev, V. Oleinikov, D. Klinov, 2477–2481.
M. Pluot, J.H. Cohen, I. Nabiev, Analytical Biochemistry 324 (2004) 60–67. [284] M. Wang, G. Abbineni, A. Clevenger, C.B. Mao, S.K. Xu, Nanomedicine: Nano-
[237] W. Jiang, S. Mardyani, H. Fischer, W.C.W. Chan, Chemistry of Materials 18 (2006) technology, Biology, and Medicine 7 (2011) 710–729.
872–878. [285] G.S. Yi, H.C. Lu, S.Y. Zhao, G. Yue, W.J. Yang, D.P. Chen, L.H. Guo, Nano Letters 4
[238] A.M. Smith, S. Nie, Journal of the American Chemical Society 130 (2008) 11278– (2004) 2191–2196.
11279. [286] X. Wang, J. Zhuang, Q. Peng, Y. Li, Nature 437 (2005) 121–124.
[239] M. Bruchez Jr., M. Moronne, P. Gin, S. Weiss, A.P. Alivisatos, Science 281 (1998) [287] X. Wang, J. Zhuang, Q. Peng, Y. Li, Inorganic Chemistry 45 (2006) 6661–6665.
2013–2016. [288] L. Wang, Y. Li, Nano Letters 6 (2006) 1645–1649.
[240] D. Gerion, F. Pinaud, S.C. Williams, W.J. Parak, D. Zanchet, S. Weiss, A.P. Alivi- [289] L.Y. Wang, Y.D. Li, Chemistry of Materials 19 (2007) 727–734.
satos, The Journal of Physical Chemistry. B 105 (2001) 8861–8871. [290] F. Zhang, Y. Wan, T. Yu, F. Zhang, Y. Shi, S. Xie, Y. Li, L. Xu, B. Tu, D. Zhao,
[241] S.T. Selvan, T.T. Tan, J.Y. Ying, Advanced Materials 17 (2005) 1620–1625. Angewandte Chemie (International ed. in English) 46 (2007) 7976–7979.
[242] X. Gao, Y. Cui, R.M. Levenson, L.W. Chung, S. Nie, Nature Biotechnology 22 (2004) [291] F. Zhang, J. Li, J. Shan, L. Xu, D. Zhao, Chemistry 15 (2009) 11010–11019.
969–976. [292] F. Zhang, D. Zhao, ACS Nano 3 (2009) 159–164.
[243] R. Hardman, Environmental Health Perspectives 114 (2006) 165–172. [293] Z.H. Xu, C.X. Li, P.P. Yang, C.M. Zhang, S.S. Huang, J. Lin, Crystal Growth & Design 9
[244] A.M. Derfus, W.C.W. Chan, S.N. Bhatia, Nano Letters 4 (2004) 11–18. (2009) 4752–4758.
[245] J. Lovric, H.S. Bazzi, Y. Cuie, G.R. Fortin, F.M. Winnik, D. Maysinger, Journal of [294] L.W. Yang, H.L. Han, Y.Y. Zhang, J.X. Zhong, The Journal of Physical Chemistry. C
Molecular Medicine 83 (2005) 377–385. 113 (2009) 18995–18999.
[247] A. Hoshino, K. Hanaki, K. Suzuki, K. Yamamoto, Biochemical and Biophysical [295] J.J. Li, L.W. Yang, Y.Y. Zhang, J.X. Zhong, C.Q. Sun, P.K. Chu, Optical Materials 33
Research Communications 314 (2004) 46–53. (2011) 882–887.
[248] E.B. Voura, J.K. Jaiswal, H. Mattoussi, S.M. Simon, Nature Medicine 10 (2004) [296] D. Chen, P. Huang, Y. Yu, F. Huang, A. Yang, Y. Wang, Chemical Communications
993–998. 47 (2011) 5801–5803.
[249] J.K. Jaiswal, H. Mattoussi, J.M. Mauro, S.M. Simon, Nature Biotechnology 21 [297] Y.H. Wang, R.X. Cai, Z.H. Liu, Crystengcomm 13 (2011) 1772–1774.
(2003) 47–51. [298] L.Y. Wang, P. Li, Y.D. Li, Advanced Materials 19 (2007) 3304–3307.
[250] J.P. Zimmer, S.W. Kim, S. Ohnishi, E. Tanaka, J.V. Frangioni, M.G. Bawendi, Journal [299] H. Hu, Z. Chen, T. Cao, Q. Zhang, M. Yu, F. Li, T. Yi, C. Huang, Nanotechnology 19
of the American Chemical Society 128 (2006) 2526–2527. (2008) 375702.
[251] R. Xie, K. Chen, X. Chen, X. Peng, Nano Research 1 (2008) 457–464. [300] C.M. Zhang, P.A. Ma, C.X. Li, G.G. Li, S.S. Huang, D.M. Yang, M.M. Shang, X.J. Kang,
[252] L. Li, T.J. Daou, I. Texier, T.K.C. Tran, Q.L. Nguyen, P. Reiss, Chemical Materials 21 J. Lin, Journal of Material Chemistry 21 (2011) 717–723.
(2009) 2422–2429. [301] J.Y. Sun, J.B. Xian, H.Y. Du, The Journal of Physical Chemistry of Solids 72 (2011)
[253] R.G. Aswathy, Y. Yoshida, T. Maekawa, D.S. Kumar, Analytical and Bioanalytical 207–213.
Chemistry 397 (2010) 1417–1435. [302] J.Y. Sun, J.B. Xian, Z.G. Xia, H.Y. Du, Journal of Rare Earths 28 (2010) 219–
[254] J. Gao, K. Chen, R. Xie, J. Xie, S. Lee, Z. Cheng, X. Peng, X. Chen, Small 6 (2010) 256– 221.
261. [303] G.F. Wang, Q. Peng, Y.D. Li, Chemical Communications 46 (2010) 7528–7529.
[255] D.R. Larson, W.R. Zipfel, R.M. Williams, S.W. Clark, M.P. Bruchez, F.W. Wise, W.W. [304] X.H. Chuai, D.S. Zhang, D. Zhao, K.Z. Zheng, C.F. He, F. Shi, L.L. Wang, H. Chen, W.P.
Webb, Science 300 (2003) 1434–1436. Qin, Materials Letters 65 (2011) 2368–2370.
[256] B. Ballou, B.C. Lagerholm, L.A. Ernst, M.P. Bruchez, A.S. Waggoner, Bioconjugate [305] D. Li, C.R. Ding, G. Song, S.Z. Lu, Z. Zhang, Y.U. Shi, H. Shen, Y.L. Zhang, H.Q.
Chemistry 15 (2004) 79–86. Ouyang, H. Wang, The Journal of Physical Chemistry. C 114 (2010) 21378–
[257] L. Ye, K.T. Yong, L. Liu, I. Roy, R. Hu, J. Zhu, H. Cai, W.C. Law, J. Liu, K. Wang, Y. Liu, 21384.
Y. Hu, X. Zhang, M.T. Swihart, P.N. Prasad, Nature Nanotechnology 7 (2012) 453– [306] J.H. Zeng, J. Su, Z.H. Li, R.X. Yan, Y.D. Li, Advanced Materials 17 (2005) 2119–
458. 2123.
[258] K. Chen, J. Xie, H. Xu, D. Behera, M.H. Michalski, S. Biswal, A. Wang, X. Chen, [307] X.P. Fan, D.B. Pi, F. Wang, J.R. Qiu, M.Q. Wang, IEEE Transactions on Nanotech-
Biomaterials 30 (2009) 6912–6919. nology 5 (2006) 123–128.
[259] G. Prencipe, S.M. Tabakman, K. Welsher, Z. Liu, A.P. Goodwin, L. Zhang, J. Henry, [308] G.F. Wang, Q. Peng, Y.D. Li, Journal of the American Chemical Society 131 (2009)
H. Dai, Journal of the American Chemical Society 131 (2009) 4783–4787. 14200–14201.
68 H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69

[309] M. Pedroni, F. Piccinelli, T. Passuello, M. Giarola, G. Mariotto, S. Polizzi, M. [360] D.K. Chatteriee, A.J. Rufalhah, Y. Zhang, Biomaterials 29 (2008) 937–943.
Bettinelli, A. Speghini, Nanoscale 3 (2011) 1456–1460. [361] L.A. Cheng, K. Yang, S.A. Zhang, M.W. Shao, S.T. Lee, Z.A. Liu, Nano Research 3
[310] J.Y. Sun, J.B. Xian, H.Y. Du, Applied Surface Science 257 (2011) 3592–3595. (2010) 722–732.
[311] J.Y. Sun, J.B. Xian, X.Y. Zhang, H.Y. Du, Journal of Rare Earths 29 (2011) 32–38. [362] L.Q. Xiong, Z.G. Chen, M.X. Yu, F.Y. Li, C. Liu, C.H. Huang, Biomaterials 30 (2009)
[312] T. Pang, W.H. Cao, M.M. Xing, X.X. Luo, X.F. Yang, Optical Materials 33 (2011) 5592–5600.
485–489. [363] L. Xiong, Z. Chen, Q. Tian, T. Cao, C. Xu, F. Li, Analytical Chemistry 81 (2009)
[313] J.H. Zeng, T. Xie, Z.H. Li, Y.D. Li, Crystal Growth & Design 7 (2007) 2774–2777. 8687–8694.
[314] F. Wang, Y. Han, C.S. Lim, Y. Lu, J. Wang, J. Xu, H. Chen, C. Zhang, M. Hong, X. Liu, [364] X.F. Yu, Z. Sun, M. Li, Y. Xiang, Q.Q. Wang, F. Tang, Y. Wu, Z. Cao, W. Li,
Nature 463 (2010) 1061–1065. Biomaterials 31 (2010) 8724–8731.
[315] Z. Li, Y. Zhang, Nanotechnology 19 (2008) 345606. [365] A. Xia, Y. Gao, J. Zhou, C. Li, T. Yang, D. Wu, L. Wu, F. Li, Biomaterials 32 (2011)
[316] H. Schafer, P. Ptacek, H. Eickmeier, M. Haase, Advanced Functional Materials 19 7200–7208.
(2009) 3091–3097. [366] Q. Xiao, W. Bu, Q. Ren, S. Zhang, H. Xing, F. Chen, M. Li, X. Zheng, Y. Hua, L. Zhou,
[317] Y. Wei, F.Q. Lu, X.R. Zhang, D.P. Chen, Chemistry of Materials 18 (2006) 5733– W. Peng, H. Qu, Z. Wang, K. Zhao, J. Shi, Biomaterials 33 (2012) 7530–7539.
5737. [367] S. Link, M.A. El-Sayed, The Journal of Physical Chemistry B 103 (1999) 8410–
[318] M. Wang, J.L. Liu, Y.X. Zhang, W. Hou, X.L. Wu, S.K. Xu, Materials Letters 63 8426.
(2009) 325–327. [368] M.C. Daniel, D. Astruc, Chemical Reviews 104 (2004) 293–346.
[319] C.H. Liu, J. Sun, H. Wang, D. Chen, Scripta Materialia 58 (2008) 89–92. [369] J. Yguerabide, E.E. Yguerabide, Analytical Biochemistry 262 (1998) 157–176.
[320] F. Shi, J.S. Wang, X.S. Zhai, D. Zhao, W.P. Qin, Crystengcomm 13 (2011) 3782– [370] H. Horvath, Journal of Quantitative Spectroscopy & Radiative Transfer 110
3787. (2009) 787–799.
[321] D.Q. Chen, Y.L. Yu, F. Huang, A.P. Yang, Y.S. Wang, Journal of Material Chemistry [371] E. Boisselier, D. Astruc, Chemical Society Reviews 38 (2009) 1759–1782.
21 (2011) 6186–6192. [372] H.Y. Wu, H.C. Chu, T.J. Kuo, C.L. Kuo, M.H. Huang, Chemistry of Materials 17
[322] Y.W. Zhang, X. Sun, R. Si, L.P. You, C.H. Yan, Journal of the American Chemical (2005) 6447–6451.
Society 127 (2005) 3260–3261. [373] S. Lal, S.E. Clare, N.J. Halas, Accounts of Chemical Research 41 (2008) 1842–1851.
[323] J.C. Boyer, F. Vetrone, L.A. Cuccia, J.A. Capobianco, Journal of the American [374] Y.N. Xia, W.Y. Li, C.M. Cobley, J.Y. Chen, X.H. Xia, Q. Zhang, M.X. Yang, E.C. Cho,
Chemical Society 128 (2006) 7444–7445. P.K. Brown, Accounts of Chemical Research 44 (2011) 914–924.
[324] J.C. Boyer, L.A. Cuccia, J.A. Capobianco, Nano Letters 7 (2007) 847–852. [375] S.J. Oldenburg, R.D. Averitt, S.L. Westcott, N.J. Halas, Chemical Physics Letters
[325] H.X. Mai, Y.W. Zhang, L.D. Sun, C.R. Yan, The Journal of Physical Chemistry. C 111 288 (1998) 243–247.
(2007) 13730–13739. [376] E.S. Glazer, C. Zhu, K.L. Massey, C.S. Thompson, W.D. Kaluarachchi, A.N. Hamir,
[326] Y.P. Du, Y.W. Zhang, Z.G. Yan, L.D. Sun, S. Gao, C.H. Yan, Chemistry, An Asian S.A. Curley, Clinical Cancer Research 16 (2010) 5712–5721.
Journal 2 (2007) 965–974. [377] M. Raoof, S.J. Corr, W.D. Kaluarachchi, K.L. Massey, K. Briggs, C. Zhu, M.A. Cheney,
[327] J. Shan, X. Qin, N. Yao, Y. Ju, Nanotechnology 18 (2007) 445607. L.J. Wilson, S.A. Curley, Nanomedicine 8 (2012) 1096.
[328] J.N. Shan, Y.G. Ju, Nanotechnology 20 (2009) 155101. [378] J. Turkevich, P.C. Stevenson, J. Hillier, Discussions of the Faraday Society 11
[329] S.J. Budijono, J.N. Shan, N. Yao, Y. Miura, T. Hoye, R.H. Austin, Y.G. Ju, R.K. (1951) 55–75.
Prud’homme, Chemistry of Materials 22 (2010) 311–318. [379] G. Frens, Nature Physical Science 241 (1973) 20–22.
[330] L.L. Li, R.B. Zhang, L.L. Yin, K.Z. Zheng, W.P. Qin, P.R. Selvin, Y. Lu, Angewandte [380] J. Kimling, M. Maier, B. Okenve, V. Kotaidis, H. Ballot, A. Plech, The Journal of
Chemie (International ed. in English) 51 (2012) 6121–6125. Physical Chemistry B 110 (2006) 15700–15707.
[331] G.S. Yi, G.M. Chow, Advanced Functional Materials 16 (2006) 2324–2329. [381] C.J. Murphy, T.K. San, A.M. Gole, C.J. Orendorff, J.X. Gao, L. Gou, S.E. Hunyadi, T. Li,
[332] G.S. Yi, Y.F. Peng, Z.Q. Gao, Chemistry of Materials 23 (2011) 2729–2734. The Journal of Physical Chemistry. B 109 (2005) 13857–13870.
[333] D. Li, B.A. Dong, X. Bai, Y. Wang, H.W. Song, The Journal of Physical Chemistry. C [382] S.T. Hussain, M. Iqbal, M. Mazhar, Journal of Nanoparticle Research 11 (2009)
114 (2010) 8219–8226. 1383–1391.
[334] B.A. Dong, S. Xu, J.A. Sun, S. Bi, D. Li, X. Bai, Y. Wang, L.P. Wang, H.W. Song, Journal [383] K. Esumi, A. Kameo, A. Suzuki, K. Torigoe, Colloids Surfaces A: Physicochemical
of Material Chemistry 21 (2011) 6193–6200. Engineering Aspects 189 (2001) 155–161.
[335] H. Schafer, P. Ptacek, K. Kompe, M. Haase, Chemistry of Materials 19 (2007) [384] M. Brust, M. Walker, D. Bethell, D.J. Schiffrin, R. Whyman, Journal of the Chemical
1396–1400. Society, Chemical Communications (7) (1994) 801–802.
[336] T.Y. Cao, T.S. Yang, Y. Gao, Y. Yang, H. Hu, F.Y. Li, Inorganic Chemistry [385] X.J. Yin, S.G. Chen, A.G. Wu, Micro & Nano Letters 5 (2010) 270–273.
Communications 13 (2010) 392–394. [386] S.P. Dubey, M. Lahtinen, M. Sillanpaa, Colloids and Surfaces A: Physicochemical
[337] J. Zhou, M. Yu, Y. Sun, X. Zhang, X. Zhu, Z. Wu, D. Wu, F. Li, Biomaterials 32 (2011) and Engineering Aspects 364 (2010) 34–41.
1148–1156. [387] K. Roy, S. Lahiri, Green Chemistry 8 (2006) 1063–1066.
[338] G. Chen, T.Y. Ohulchanskyy, W.C. Law, H. Agren, P.N. Prasad, Nanoscale 3 (2011) [388] C. Xu, J. Xie, D. Ho, C. Wang, N. Kohler, E.G. Walsh, J.R. Morgan, Y.E. Chin, S. Sun,
2003–2008. Angewandte Chemie (International ed. in English) 47 (2008) 173–176.
[339] C. Liu, Z. Wang, H. Jia, Z. Li, Chemical Communications 47 (2011) 4661–4663. [389] H. Yu, M. Chen, P.M. Rice, S.X. Wang, R.L. White, S. Sun, Nano Letters 5 (2005)
[340] C.L. Zhang, Y.X. Yuan, S.M. Zhang, Y.H. Wang, Z.H. Liu, Angewandte Chemie 379–382.
(International ed. in English) 50 (2011) 6851–6854. [390] Y.Y. Yu, S.S. Chang, C.L. Lee, C.R.C. Wang, The Journal of Physical Chemistry B 101
[341] R. Naccache, F. Vetrone, V. Mahalingam, L.A. Cuccia, J.A. Capobianco, Chemistry (1997) 6661–6664.
of Materials 21 (2009) 717–723. [391] N.R. Jana, L. Gearheart, C.J. Murphy, The Journal of Physical Chemistry B 105
[342] L. Xiong, T. Yang, Y. Yang, C. Xu, F. Li, Biomaterials 31 (2010) 7078–7085. (2001) 4065–4067.
[343] Q.T. Chen, X. Wang, F.H. Chen, Q.B. Zhang, B. Dong, H. Yang, G.X. Liu, Y.M. Zhu, [392] A. Gole, C.J. Murphy, Chemistry of Materials 16 (2004) 3633–3640.
Journal of Material Chemistry 21 (2011) 7661–7667. [393] B. Nikoobakht, M.A. El-Sayed, Chemistry of Materials 15 (2003) 1957–1962.
[344] R. Kumar, M. Nyk, T.Y. Ohulchanskyy, C.A. Flask, P.N. Prasad, Advanced Func- [394] N.R. Jana, L. Gearheart, C.J. Murphy, Advanced Materials 13 (2001) 1389–
tional Materials 19 (2009) 853–859. 1393.
[345] M. Nyk, R. Kumar, T.Y. Ohulchanskyy, E.J. Bergey, P.N. Prasad, Nano Letters 8 [395] Y.N. Xia, Y.J. Xiong, B. Lim, S.E. Skrabalak, Angewandte Chemie (International ed.
(2008) 3834–3838. in English) 48 (2009) 60–103.
[346] J.C. Boyer, M.P. Manseau, J.I. Murray, F.C. van Veggel, Langmuir 26 (2010) 1157– [396] L.R. Hirsch, A.M. Gobin, A.R. Lowery, F. Tam, R.A. Drezek, N.J. Halas, J.L. West,
1164. Annals of Biomedical Engineering 34 (2006) 15–22.
[347] Q.B. Zhang, K. Song, J.W. Zhao, X.G. Kong, Y.J. Sun, X.M. Liu, Y.L. Zhang, Q.H. Zeng, [397] W. Stober, A. Fink, E. Bohn, Journal of Colloid and Interface Science 26 (1968) 62–
H. Zhang, Journal of Colloid and Interface Science 336 (2009) 171–175. 69.
[348] J. Shen, L.D. Sun, Y.W. Zhang, C.H. Yan, Chemical Communications 46 (2010) [398] N.J. Halas, MRS Bulletin 30 (2005) 362–367.
5731–5733. [399] C. Radloff, N.J. Halas, Nano Letters 4 (2004) 1323–1327.
[349] A. Dong, X. Ye, J. Chen, Y. Kang, T. Gordon, J.M. Kikkawa, C.B. Murray, Journal of [400] R. Bardhan, S. Mukherjee, N.A. Mirin, S.D. Levit, P. Nordlander, N.J. Halas, The
the American Chemical Society 133 (2011) 998–1006. Journal of Physical Chemistry. C 114 (2010) 7378–7383.
[350] R. Abdul Jalil, Y. Zhang, Biomaterials 29 (2008) 4122–4128. [401] J.Z. Zhang, The Journal of Physical Chemistry Letters 1 (2010) 686–695.
[351] J.N. Shan, Y.G. Ju, Applied Physics Letters 91 (2007) 123103. [402] Y.D. Jin, X.H. Gao, Nature Nanotechnology 4 (2009) 571–576.
[352] S. Jiang, Y. Zhang, K.M. Lim, E.K. Sim, L. Ye, Nanotechnology 20 (2009) 155101. [403] Y.D. Jin, X.H. Gao, Journal of the American Chemical Society 131 (2009) 17774–
[353] Z. Chen, H. Chen, H. Hu, M. Yu, F. Li, Q. Zhang, Z. Zhou, T. Yi, C. Huang, Journal of 17776.
the American Chemical Society 130 (2008) 3023–3029. [404] Y.G. Sun, B.T. Mayers, Y.N. Xia, Nano Letters 2 (2002) 481–485.
[354] J. Zhou, Y. Sun, X.X. Du, L.Q. Xiong, H. Hu, F.Y. Li, Biomaterials 31 (2010) 3287– [405] Y.G. Sun, Y.N. Xia, Science 298 (2002) 2176–2179.
3295. [406] Y.G. Sun, Y.N. Xia, Journal of the American Chemical Society 126 (2004) 3892–
[355] H. Hu, M.X. Yu, F.Y. Li, Z.G. Chen, X. Gao, L.Q. Xiong, C.H. Huang, Chemistry of 3901.
Materials 20 (2008) 7003–7009. [407] S.E. Skrabalak, J.Y. Chen, Y.G. Sun, X.M. Lu, L. Au, C.M. Cobley, Y.N. Xia, Accounts
[356] S. Jiang, Y. Zhang, K.M. Lim, E.K.W. Sim, L. Ye, Nanotechnology 20 (2009) of Chemical Research 41 (2008) 1587–1595.
155101–155109. [408] S.E. Skrabalak, L. Au, X.D. Li, Y.N. Xia, Nature Protocols 2 (2007) 2182–2190.
[357] R.A. Jalil, Y. Zhang, Biomaterials 29 (2008) 4122–4128. [409] J.Y. Chen, M.X. Yang, Q.A. Zhang, E.C. Cho, C.M. Cobley, C. Kim, C. Glaus, L.H.V.
[358] S.F. Lim, R. Riehn, C.K. Tung, W.S. Ryu, R. Zhuo, J. Dalland, R.H. Austin, Nano- Wang, M.J. Welch, Y.N. Xia, Advanced Functional Materials 20 (2010) 3684–
technology 20 (2009) 405701. 3694.
[359] S.A. Hilderbrand, F. Shao, C. Salthouse, U. Mahmood, R. Weissleder, Chemical [410] C.M. Cobley, L. Au, J. Chen, Y. Xia, Expert Opinion on Drug Delivery 7 (2010) 577–
Communications (28) (2009) 4188–4190. 587.
H. Chen et al. / Materials Science and Engineering R 74 (2013) 35–69 69

[411] M.S. Yavuz, Y. Cheng, J. Chen, C.M. Cobley, Q. Zhang, M. Rycenga, J. Xie, C. Kim, [447] M. Eghtedari, A. Oraevsky, J.A. Copland, N.A. Kotov, A. Conjusteau, M. Motamedi,
K.H. Song, A.G. Schwartz, L.V. Wang, Y. Xia, Nature Materials 8 (2009) 935–939. Nano Letters 7 (2007) 1914–1918.
[412] W. Li, X. Cai, C. Kim, G. Sun, Y. Zhang, R. Deng, M. Yang, J. Chen, S. Achilefu, L.V. [448] C.W. Wei, S.W. Huang, C.R. Wang, P.C. Li, IEEE Transactions on Ultrasonics,
Wang, Y. Xia, Nanoscale 3 (2011) 1724–1730. Ferroelectrics, and Frequency Control 54 (2007) 1131–1141.
[413] G.D. Moon, S.W. Choi, X. Cai, W. Li, E.C. Cho, U. Jeong, L.V. Wang, Y. Xia, Journal of [449] X.M. Yang, S.E. Skrabalak, Z.Y. Li, Y.N. Xia, L.H.V. Wang, Nano Letters 7 (2007)
the American Chemical Society 133 (2011) 4762–4765. 3798–3802.
[414] J.C. Love, L.A. Estroff, J.K. Kriebel, R.G. Nuzzo, G.M. Whitesides, Chemical Reviews [450] K.H. Song, C.H. Kim, C.M. Cobley, Y.N. Xia, L.V. Wang, Nano Letters 9 (2009) 183–
105 (2005) 1103–1169. 188.
[415] W. Lu, C. Xiong, G. Zhang, Q. Huang, R. Zhang, J.Z. Zhang, C. Li, Clinical Cancer [451] K. Wilson, K. Homan, S. Emelianov, Nature Communications 3 (2012) 618.
Research 15 (2009) 876–886. [452] M. Fleischm, P.J. Hendra, A.J. Mcquilla, Chemical Physics Letters 26 (1974) 163–166.
[416] M.P. Melancon, W. Lu, Z. Yang, R. Zhang, Z. Cheng, A.M. Elliot, J. Stafford, T. Olson, [453] D.L. Jeanmaire, R.P. Vanduyne, Journal of Electroanalytical Chemistry 84 (1977)
J.Z. Zhang, C. Li, Molecular Cancer Therapeutics 7 (2008) 1730–1739. 1–20.
[417] W. Lu, G. Zhang, R. Zhang, L.G. Flores 2nd, Q. Huang, J.G. Gelovani, C. Li, Cancer [454] M.G. Albrecht, J.A. Creighton, Journal of the American Chemical Society 99
Research 70 (2010) 3177–3188. (1977) 5215–5217.
[418] G. von Maltzahn, J.H. Park, A. Agrawal, N.K. Bandaru, S.K. Das, M.J. Sailor, S.N. [455] Y. Lu, G.L. Liu, J. Kim, Y.X. Mejia, L.P. Lee, Nano Letters 5 (2005) 119–124.
Bhatia, Cancer Research 69 (2009) 3892–3900. [456] G.L. Liu, Y. Lu, J. Kim, J.C. Doll, L.P. Lee, Advanced Materials 17 (2005) 2683–2688.
[419] G. Zhang, Z. Yang, W. Lu, R. Zhang, Q. Huang, M. Tian, L. Li, D. Liang, C. Li, [457] S. Keren, C. Zavaleta, Z. Cheng, A. de la Zerda, O. Gheysens, S.S. Gambhir,
Biomaterials 30 (2009) 1928–1936. Proceedings of the National Academy of Sciences of the United States of America
[420] J. Cao, E.K. Galbraith, T. Sun, K.T.V. Grattan, Sensors and Actuators B: Chemical 105 (2008) 5844–5849.
169 (2012) 360–367. [458] S. Lee, S. Kim, J. Choo, S.Y. Shin, Y.H. Lee, H.Y. Choi, S. Ha, K. Kang, C.H. Oh,
[421] D. Sahu, H.C. Chu, P.J. Yang, H.C. Lin, Macromolecular Chemistry and Physics 213 Analytical Chemistry 79 (2007) 916–922.
(2012) 1550–1558. [459] S. Zong, Z. Wang, J. Yang, C. Wang, S. Xu, Y. Cui, Talanta 97 (2012) 368–375.
[422] Y. Jung, R. Reif, Y. Zeng, R.K. Wang, Nano Letters 11 (2011) 2938–2943. [460] J.V. Jokerst, Z. Miao, C. Zavaleta, Z. Cheng, S.S. Gambhir, Small 7 (2011) 625–633.
[423] Q. Dai, J. Coutts, J.H. Zou, Q. Huo, Chemical Communications (25) (2008) 2858– [461] M.F. Kircher, A. de la Zerda, J.V. Jokerst, C.L. Zavaleta, P.J. Kempen, E. Mittra, K.
2860. Pitter, R. Huang, C. Campos, F. Habte, R. Sinclair, C.W. Brennan, I.K. Mellinghoff,
[424] X.G. Hu, X.H. Gao, Physical Chemistry Chemical Physics 13 (2011) 10028–10035. E.C. Holland, S.S. Gambhir, Nature Medicine 18 (2012) 829–834.
[425] Y. Pan, S. Neuss, A. Leifert, M. Fischler, F. Wen, U. Simon, G. Schmid, W. Brandau, [462] M.L. James, S.S. Gambhir, Physiological Reviews 92 (2012) 897–965.
W. Jahnen-Dechent, Small 3 (2007) 1941–1949. [463] Q.Y. Cai, S.H. Kim, K.S. Choi, S.Y. Kim, S.J. Byun, K.W. Kim, S.H. Park, S.K. Juhng,
[426] H. Ghandehari, A. Malugin, H. Herd, A. Gormley, T. Yu, Abstracts of Papers of the K.H. Yoon, Investigative Radiology 42 (2007) 797–806.
American Chemical Society 241 (2011) 21. [464] D. Kim, S. Park, J.H. Lee, Y.Y. Jeong, S. Jon, Nanomedicine: Nanotechnology,
[427] A.M. Alkilany, P.K. Nagaria, C.R. Hexel, T.J. Shaw, C.J. Murphy, M.D. Wyatt, Small 5 Biology, and Medicine 3 (2007) 352.
(2009) 701–708. [465] R. Popovtzer, A. Agrawal, N.A. Kotov, A. Popovtzer, J. Balter, T.E. Carey, R.
[428] A.M. Alkilany, C.J. Murphy, Journal of Nanoparticle Research 12 (2010) 2313– Kopelman, Nano Letters 8 (2008) 4593–4596.
2333. [466] D. Kim, S. Park, J.H. Lee, Y.Y. Jeong, S. Jon, Journal of the American Chemical
[429] T.S. Hauck, A.A. Ghazani, W.C. Chan, Small 4 (2008) 153–159. Society 129 (2007) 7661–7665.
[430] A.P. Leonov, J. Zheng, J.D. Clogston, S.T. Stern, A.K. Patri, A. Wei, ACS Nano 2 [467] T. Reuveni, M. Motiei, Z. Romman, A. Popovtzer, R. Popovtzer, International
(2008) 2481–2488. Journal of Nanomedicine 6 (2011) 2859–2864.
[431] A.M. Alkilany, P.K. Nagaria, M.D. Wyatt, C.J. Murphy, Langmuir 26 (2010) 9328– [468] C.A. Mirkin, R.L. Letsinger, R.C. Mucic, J.J. Storhoff, Nature 382 (1996) 607–609.
9333. [469] M. Filippova, L.S. Ross, S.S. Gill, Insect Molecular Biology 7 (1998) 223–232.
[432] A.M. Alkilany, C.J. Murphy, Langmuir 25 (2009) 13874–13879. [470] R. Elghanian, J.J. Storhoff, R.C. Mucic, R.L. Letsinger, C.A. Mirkin, Science 277
[433] H. Takahashi, Y. Niidome, T. Niidome, K. Kaneko, H. Kawasaki, S. Yamada, (1997) 1078–1081.
Langmuir 22 (2006) 2–5. [471] R.C. Jin, G.S. Wu, Z. Li, C.A. Mirkin, G.C. Schatz, Journal of the American Chemical
[434] X. Qian, X.H. Peng, D.O. Ansari, Q. Yin-Goen, G.Z. Chen, D.M. Shin, L. Yang, A.N. Society 125 (2003) 1643–1654.
Young, M.D. Wang, S. Nie, Nature Biotechnology 26 (2008) 83–90. [472] Y.W.C. Cao, R.C. Jin, C.A. Mirkin, Science 297 (2002) 1536–1540.
[435] S.K. Libutti, G.F. Paciotti, A.A. Byrnes, H.R. Alexander Jr., W.E. Gannon, M. Walker, [473] C. Mirkin, Abstracts of Papers of the American Chemical Society 225 (2003)
G.D. Seidel, N. Yuldasheva, L. Tamarkin, Clinical Cancer Research 16 (2010) U963–U964.
6139–6149. [474] S.I. Stoeva, J.S. Lee, C.S. Thaxton, C.A. Mirkin, Angewandte Chemie (International
[436] J.G. Fujimoto, C. Pitris, S.A. Boppart, M.E. Brezinski, Neoplasia 2 (2000) 9–25. ed. in English) 45 (2006) 3303–3306.
[437] G.J. Tearney, M.E. Brezinski, B.E. Bouma, S.A. Boppart, C. Pitris, J.F. Southern, J.G. [475] L.R. Hirsch, J.B. Jackson, A. Lee, N.J. Halas, J. West, Analytical Chemistry 75 (2003)
Fujimoto, Science 276 (1997) 2037–2039. 2377–2381.
[438] L.R. Hirsch, R.J. Stafford, J.A. Bankson, S.R. Sershen, B. Rivera, R.E. Price, J.D. Hazle, [476] W.T. Lu, R. Arumugam, D. Senapati, A.K. Singh, T. Arbneshi, S.A. Khan, H.T. Yu, P.C.
N.J. Halas, J.L. West, Proceedings of the National Academy of Sciences of the Ray, ACS Nano 4 (2010) 1739–1749.
United States of America 100 (2003) 13549–13554. [477] X. Qian, X. Zhou, S. Nie, Journal of the American Chemical Society 130 (2008)
[439] J. Chen, F. Saeki, B.J. Wiley, H. Cang, M.J. Cobb, Z.Y. Li, L. Au, H. Zhang, M.B. 14934–14935.
Kimmey, X.D. Li, Y.N. Xia, Nano Letters 5 (2005) 473–477. [478] M.Y. Sha, H. Xu, M.J. Natan, R. Cromer, Journal of the American Chemical Society
[440] A.L. Oldenburg, M.N. Hansen, T.S. Ralston, A. Wei, S.A. Boppart, Journal of 130 (2008) 17214–17215.
Materials Chemistry 19 (2009) 6407. [479] X. Wang, X.M. Qian, J.J. Beitler, Z.G. Chen, F.R. Khuri, M.M. Lewis, H.J.C. Shin, S.M.
[441] A.M. Gobin, M.H. Lee, N.J. Halas, W.D. James, R.A. Drezek, J.L. West, Nano Letters 7 Nie, D.M. Shin, Cancer Research 71 (2011) 1526–1532.
(2007) 1929–1934. [480] A.Y. Louie, Chemical Reviews 110 (2010) 3146–3195.
[442] C. Loo, A. Lin, L. Hirsch, M.H. Lee, J. Barton, N.J. Halas, J. West, R. Drezek, [481] R.H.I. Veld, G. Storm, W.E. Hennink, F. Kiessling, T. Lammers, Nanoscale 3 (2011)
Technology in Cancer Research & Treatment 3 (2004) 33–40. 4022–4034.
[443] L.H.V. Wang, S. Hu, Science 335 (2012) 1458–1462. [482] T. Lammers, F. Kiessling, W.E. Hennink, G. Storm, Molecular Pharmaceutics 7
[444] L.V. Wang, Medical Physics 35 (2008) 5758–5767. (2010) 1899–1912.
[445] G. Ku, L.H.V. Wang, Optical Letters 30 (2005) 507–509. [483] F.M. Kievit, M.Q. Zhang, Advanced Materials 23 (2011) 217–247.
[446] X.D. Wang, Y.J. Pang, G. Ku, X.Y. Xie, G. Stoica, L.H.V. Wang, Nature Biotechnology [484] L.S. Wang, M.C. Chuang, J.A. Ho, International Journal of Nanomedicine 7 (2012)
21 (2003) 803–806. 4679–4695.

Das könnte Ihnen auch gefallen