Theranostic Applications of Carbon Nanomaterials in Cancer: Focus on Imaging and Cargo Delivery Daiqin Chen, Casey A. Dougherty, Kaicheng Zhu, Hao Hong PII: DOI: Reference:

S0168-3659(15)00251-5 doi: 10.1016/j.jconrel.2015.04.021 COREL 7642

To appear in:

Journal of Controlled Release

Received date: Revised date: Accepted date:

7 March 2015 17 April 2015 18 April 2015

Please cite this article as: Daiqin Chen, Casey A. Dougherty, Kaicheng Zhu, Hao Hong, Theranostic Applications of Carbon Nanomaterials in Cancer: Focus on Imaging and Cargo Delivery, Journal of Controlled Release (2015), doi: 10.1016/j.jconrel.2015.04.021

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT

Theranostic Applications of Carbon Nanomaterials in Cancer: Focus on Imaging and Cargo Delivery

RI

PT

Daiqin Chen1,2, Casey A. Dougherty1,2, Kaicheng Zhu1,2, Hao Hong1,2,3*

1

SC

Center for Molecular Imaging, University of Michigan Health Systems, Ann Arbor, Michigan 48109, United States 2

NU

Department of Radiology, University of Michigan Health Systems, Ann Arbor, Michigan 48109, United States 3

MA

University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan 48109, United States

AC CE P

TE

D

Requests for reprints: Hao Hong, PhD, Departments of Radiology, University of Michigan Health Systems, Room A520, 109 Zina Pitcher Place, Ann Arbor, MI 48109-2200, USA. Fax: 1-734-763-5447 Tel: 1-734-615-4634 E-mail: [email protected]

1

ACCEPTED MANUSCRIPT Contents 1. Introduction ............................................................................................................................ 4 2. Functionalization of carbon nanomaterials ............................................................................ 6

PT

3. Theranostic applications of fullerenes (nontubular type) ...................................................... 7 3.1 Cancer imaging with fullerenes ........................................................................................ 8

RI

3.2 Cargo delivery to cancer with fullerenes .......................................................................... 9 4. Theranostic applications of carbon nanotubes ....................................................................... 9

SC

4.1 Cancer imaging with fullerenes ...................................................................................... 10 4.2 Cargo delivery to cancer with fullerenes ........................................................................ 12

NU

5. Theranostic applications of single-walled carbon nanohorns .............................................. 12 5.1 Cancer imaging with SWNHs ........................................................................................ 13

MA

5.2 Cargo delivery to cancer with SWNHs .......................................................................... 14 6. Theranostic applications of nanodiamonds .......................................................................... 14 6.1 Cancer imaging with nanodiamonds .............................................................................. 14

D

6.2 Cargo delivery to cancer with nanodiamonds ................................................................ 16

TE

7. Theranostic applications of carbon nanodots ....................................................................... 16 7.1 Cancer imaging with carbon nanodots ........................................................................... 16 7.2 Cargo delivery to cancer with carbon nanodots ............................................................. 17

AC CE P

8. Theranostic applications of nanographenes ......................................................................... 18 8.1 Cancer imaging with nanographenes ............................................................................. 18 8.2 Cargo delivery to cancer with nanographenes................................................................ 20 9. Conclusions and future perspectives .................................................................................... 22 Acknowledgments.................................................................................................................... 23 References ................................................................................................................................ 24

2

ACCEPTED MANUSCRIPT ABSTRACT: Carbon based nanomaterials have attracted significant attention over the past decades due

PT

to their unique physical properties, versatile functionalization chemistry, and biological compatibility. In this review, we will summarize the current state-of-the-art applications of

RI

carbon nanomaterials in cancer imaging and drug delivery/therapy. The carbon nanomaterials

SC

will be categorized into fullerenes, nanotubes, nanohorns, nanodiamonds, nanodots and graphene

NU

derivatives based on their morphologies. The chemical conjugation/functionalization strategies of each category will be introduced before focusing on their applications in cancer imaging

MA

(fluorescence/bioluminescence, magnetic resonance (MR), positron emission tomography (PET), single-photon emission computed tomography (SPECT), photoacoustic, Raman imaging, etc.)

D

and cargo (chemo/gene/therapy) delivery. The advantages and limitations of each category and

TE

the potential clinical utilization of these carbon nanomaterials will be discussed. Multifunctional

AC CE P

carbon nanoplatforms have the potential to serve as optimal candidates for image-guided delivery vectors for cancer.

KEYWORDS: carbon nanomaterials, theranostics, cancer, molecular imaging, drug delivery

3

ACCEPTED MANUSCRIPT 1. INTRODUCTION With an estimation of 1,658,370 new cases and 589,430 deaths in the United States in

PT

2015, cancer is considered as the second leading health threat [1]. Despite great progress made in the past few decades, early diagnosis and efficient treatment of cancer are still challenges to

RI

overcome [2, 3]. The current standard management of cancer patients involves stage

SC

determination (by imaging or biopsies), chemo/radiation therapy, and/or surgical resection.

NU

Molecular imaging, a useful tool to monitor in vivo biochemical events, can provide invaluable insights into both cancer diagnosis and therapeutic response monitoring [4]. Different molecular

MA

imaging techniques, including optical imaging (fluorescence/bioluminescence/Raman), magnetic resonance imaging (MRI), computed tomography (CT), positron emission tomography (PET),

D

single photon emission computed tomography (SPECT), and ultrasound (US), have furthered our

TE

understanding of cancer initiation, progression, and metastasis.

AC CE P

Post cancer diagnosis/detection, chemotherapy is the most frequently used method for cancer treatment. In order to obtain the best curative effect while avoiding negative side effects, it is important to develop drug delivery systems with strong cancer targeting abilities, as well as controllable/on-demand release properties. Nanomaterials have been stated to be one of the most promising platforms for cancer imaging and drug delivery since they have the ability to synergistically combine diagnosis and therapy into one material [5, 6]. A material that combines diagnosis and therapy has been termed a “theranostic” [7, 8].

4

ACCEPTED MANUSCRIPT Nanomaterials can be generally classified into organic nanomaterials, inorganic nanomaterials and hybrid nanomaterials [9-11]. The theranostic applications of organic

PT

nanomaterials (primarily including liposomes, micelles, dendrimers, and protein-based

RI

nanomaterials) in cancer have been previously summarized elsewhere [12, 13]. Inorganic nanomaterials (e.g. silica nanoparticles, gold nanoparticles, iron oxide nanoparticles and

SC

carbonaceous nanomaterials etc.) are more actively being used for cancer theranostic

NU

applications due to their unique physical properties (which usually improve the therapeutic outputs) and comparatively lower production cost [14-16]. Among these inorganic nanomaterials,

MA

carbonaceous nanomaterials are rising stars due to their excellent mechanical, thermal and optical properties (Figure 1) [17, 18]. For instance, most of these carbonaceous nanomaterials

TE

D

possess strong absorption in the infrared (IR) or near infrared (NIR) regions, which is useful for photothermal therapy (PTT) of cancer. Some inorganic nanomaterials (e.g. carbon nanotubes or

AC CE P

nanodots) can also produce fluorescence in the visible and infrared regions for fluorescence imaging [19, 20]. In addition, carbonaceous nanomaterials are able to transform the energy from a laser into acoustic signals, which makes them promising agents for photoacoustic imaging (PAI) [21, 22]. Finally, intrinsic Raman vibration signals from carbonaceous materials can also provide a reliable method to monitor their distribution and metabolism in vivo [23, 24].

5

ACCEPTED MANUSCRIPT Figure 1 Radioisotopes

PT

Chemo drugs

MA

Carbon nanomaterials

NU

SC

RI

Genes

AC CE P

Polymer functionalization

TE

D

PDT & photothermal

Image-guided cancer therapy Targeting ligands

Imaging labels

Legend to Figure 1. The schematic illustration of theranostic applications of carbon nanomaterials in cancer.

6

ACCEPTED MANUSCRIPT Due to the unique sp2 carbon structure and inherent hydrophobic nature of carbonaceous nanomaterials, the nanomaterials have the potential to be useful drug delivery vectors. Multiple

PT

copies of drugs or DNA/RNA molecules can easily be absorbed onto their surface through

RI

hydrophobic interactions or π-π stacking [25-27]. After loading the drugs/genes, the carbonaceous nanomaterials (or their assemblies) can accumulate into tumors in vivo through

SC

either “active” targeting (by introduction of a targeting ligand for overexpressed receptors in

NU

tumor/vasculatures) or by the enhanced permeability and retention (EPR) effect. Drug/gene cargos on carbonaceous nanomaterials possess distinct pharmacokinetic behavior compared to

MA

free molecules, which may not only enhance the tumor killing efficiency but also decrease toxicity in surrounding healthy tissue. In some cases, carbonaceous nanomaterials (e.g. fullerene)

TE

D

can also be used as anticancer agents themselves and provide synergetic antitumor effects with a given drug [28]. Moreover, the addition of stimuli (e.g. light or heat) responsive polymers to

AC CE P

carbonaceous nanomaterials can also be extremely useful for selective and controllable drug release [29, 30].

Much research effort has been devoted to carbonaceous nanomaterials as platforms for cancer diagnosis and drug delivery due to their many aforementioned advantages, leading to the goal of personalized cancer therapy [7, 31]. By combining imaging labels with therapeutics in the same platform, the location of the tumor can be precisely delineated, and the optimal drug doses as well as therapeutic time frame can be determined by acquiring the real-time drug distribution information in vivo. Image-guided carbonaceous nanomaterials can evaluate the therapeutic efficacy of a given treatment by monitoring the abundance fluctuation of target proteins/receptors as well. Theranostic applications of carbonaceous nanomaterials will be discussed in the current review article. They will be categorized into fullerenes, nanotubes, 7

ACCEPTED MANUSCRIPT nanohorns, nanodiamond, nanodots and graphene derivatives according to their morphologies. The functionalization (i.e. chemical modification) of these materials before their biomedical uses

PT

will be briefly introduced. The molecular imaging and cargo delivery applications of these

RI

materials will be the major focus. Lastly, the safety/toxicity concerns of each carbonaceous

SC

nanomaterial will be covered.

2. FUNCTIONALIZATION OF CARBON NANOMATERIALS

NU

Since most of pristine carbonaceous nanomaterials are highly hydrophobic (due to the sp2 carbon nanostructure), proper functionalization is necessary before their biomedical applications.

MA

Generally speaking, there are two primary strategies of functionalization: covalent or noncovalent (Table 1). Covalent modifications usually involve introduction of hydrophilic

TE

D

functional groups (e.g. hydroxyl groups, carboxyl groups, or amino groups) for the further conjugation of protecting polymers (such as poly(ethylene glycol) [PEG]), targeting ligands, or

AC CE P

drug/gene cargos [32-35]. For example, carboxyl groups (-COOH) can be formed by oxidation by agents like nitric acid. Besides oxidation, the cycloaddition reaction is another covalent approach to modify carbonaceous nanomaterials, in which the aromatic rings inside the material structures are used as the reaction target [27].

8

ACCEPTED MANUSCRIPT Table 1 Functionalization strategies for carbonaceous nanomaterials Functionalization molecule(s)

Major Purpose(s)

Potential toxicity concerns

Representative References

PEG derivatives Chitosan

Stabilization Stabilization and gene delivery Stabilization and gene delivery Imaging label

Biocompatible Biocompatible

[63, 95, 174] [96, 210, 231]

BSA M13 phage

DNA DNA/siRNA Nanostructures (IONP, QDs, gold NPs etc.) Gadolinium (Gd) Chemo drugs Photosensitizers 9

RI

Imaging label

Mostly biocompatible

[73, 161]

Biocompatible Potential immune response

[95, 178, 242] [100, 164]

Biocompatible Biocompatible

[243] [119]

Biocompatible Potential immune response

[123] [70, 71]

TE

Pluronic F127 Chitosan

[85, 161] [132]

Stabilization Stabilization and gene delivery Stabilization Stabilization and gene loading Stabilization Stabilization and tumor targeting Stabilization

AC CE P

PEG derivatives Polyethylenimine (PEI)

[104, 146, 209]

Toxicity from premature release Tumor therapy Toxicity from premature release Tumor Potential immune targeting response

D

Antibodies, proteins, or peptides Fluorophores Noncovalent

Radiation exposure

MA

Chemo drugs

Imaging label

SC

Radioisotopes and chelators Gadolinium (Gd)

Potential immune response

NU

Polyethylenimine (PEI)

PT

Covalent

Potential immune response Gene therapy Gene inhibition in normal tissues Imaging label, MPS capture and PDT, and PTT potential metal contamination Imaging label Toxicity from premature release Tumor therapy Toxicity from premature release PDT Damage to normal tissues by ROS

[57, 97] [47, 97, 104, 174, 221]

[207] [122, 164, 212] [63, 72, 147, 203]

[116, 204] [56, 57, 119] [55, 102, 103, 121, 168, 223]

ACCEPTED MANUSCRIPT The covalently functionalized carbonaceous nanomaterials are usually stable. However, the limitation lies in that this type of functionalization method inevitably destroys partial material

PT

structure causing the loss of certain intrinsic properties (e.g. photothermal capacities). Compared

RI

with covalent functionalization, the reaction condition of noncovalent functionalization is comparatively mild, which involves coating the carbonaceous nanomaterials with amphiphilic

SC

molecules. The hydrophobic motifs of the amphiphilic molecules could be anchored onto the

NU

material’s surface with the hydrophilic ends extending to the aqueous solution and maintaining the stability of the whole material. Noncovalent interactions include electrostatic forces, π-π

MA

interactions, hydrogen bonding, or van der Waals forces [36, 37]. As we can expect, lower stability of noncovalent conjugates is the major concern for this type of functionalization. For

TE

D

example, Chung et al. attempted to conjugate lysozyme onto the surface of nanodiamonds via electrostatic interactions [38]. Although direct absorption based on electrostatic interactions was

AC CE P

observed to be fast and easy, the stability of formed conjugates was far from satisfactory. A lot of thought about stability and design of the nanomaterial must be taken into account before choosing an appropriate functionalization method for any carbonaceous nanomaterial. A balance between stability and structural integrity must be maintained before any further biomedical applications. The functionalization strategy is one of the most significant factors that can control the subsequent loading efficiency of drug/gene/imaging labels. For those materials which are not sensitive to structural alteration (e.g. carbon nanotube, nanodiamonds, or nanographene oxide), a covalent functionalization can be preferred, but other factors (e.g. effective cargo release, circulation half-life, or reagent availability) must be considered. Carbonaceous nanomaterials are only effective for cancer theranostic applications once the correct functionalization strategy is chosen and applied. 10

ACCEPTED MANUSCRIPT 3. THERANOSTIC APPLICATIONS OF FULLERENES (NONTUBULAR TYPE) Fullerene is the first category of carbonaceous nanomaterial frequently used for cancer

PT

theranostic applications. Since the initial discovery of the first fullerene, C60, in 1985, the fullerene family has been expanding rapidly with more members being identified [39]. Among

RI

these fullerene derivatives, C60 (OH)x and C82 (OH)22 are two of the most investigated fullerene

SC

species [40]. Their unique structures provide fullerenes with some very attractive and unique

NU

properties [27]. For example, gadolinium ions (Gd, T1-weighted) can be incorporated into the carbon cage of the C82 molecules, making Gd@C82 derivatives promising MRI contrast agents

MA

[41]. Photodynamic therapy (PDT) relies on reactive oxygen species (ROS, e.g. singlet oxygen) to be emitted from a photosensitizer (PS) under laser irradiation to destroy cancer cells [42].

D

Many fullerene derivatives can serve as effective photosensitizers for PDT under laser irradiation

TE

and work as a “radical sponge”, demonstrating anticancer properties by themselves [43].

AC CE P

Fullerenes are also reported to be excellent cargo delivering vectors for anticancer drugs and genes (DNA/RNA) – both enhanced cytotoxicity and gene transfection efficiency are observed with the use of fullerenes as a non-viral vector [44]. 3.1. Cancer imaging with fullerenes The most dominant utilization of fullerene in cancer imaging is for MRI. To be more specific, metallofullerene complexes (e.g. gadolinium-containing or iron oxide nanoparticle [IONP, T2-weighted]-containing) are generally considered to be good MRI contrast agents [45]. In

one

study,

a

water-soluble

gadolinium

endohedral

fulleride,

Gd@C82O6(OH)16-

(NHCH2CH2COOH)8 (AAD-EMFs) was synthesized with good stability and water proton relaxivity [46], which is ideal for MRI applications. In a follow-up report, these AAD-EMFs were further conjugated with an antibody against green fluorescence protein (GFP) [47]. With the assistance of GFP, the cellular distribution of AAD-EMFs can be more accurately detected. 11

ACCEPTED MANUSCRIPT Other groups also explored the morphological impact of the gadolinium-fullerene complex to MR relaxation behavior [48, 49]. Details of other gadolinium-fullerene complexes and their MR

PT

relaxation behavior can be found in references [50, 51]. IONP-decorated fullerene represents

RI

another category of T2-weighted MRI agents. A recent study used IONP-C60 composite for MRI, PDT, and PTT triple applications [52]. Adopting folic acid (FA) as the tumor targeting molecule,

SC

this hybrid IONP-C60 could not only visualize tumor in vivo (Figure 2A), but also provided

NU

synergistic PDT/PTT for effective and selective ablation of tumor.

MA

Figure 2

Folic acid (FA)

(A)

IONP

(B)

AC CE P

TE

D

C60 fullerene PEG Control

C60-IONP-PEG C60-IONP-PEG C60-IONP-PEG -FA + magnet -FA

PEI

DOX

DOX

Free DOX

C60-PEI-DOX

ROS

Control Control

C60-PEI-DOX + laser

C60-PEI-DOX + laser

Legend to Figure 2. (A) Structure of C60-IONP-PEG-FA and its application as an MRI-guidable PDT/PTT agent for tumors. Significant “darkening” of tumor area based on T2-weighted MR images indicated selective accumulation of C60-IONP-PEG-FA from folate receptor targeting and magnetic attraction. Adapted with permission from reference [52]. (B) Structure and application of C60-PEI-DOX for synergistic cancer therapy by DOX and PDT. C60-PEI-DOX demonstrated good uptake in melanoma cells and significant production of cellular ROS after laser irradiation. Histological examination also confirmed that treatment with C60-PEI-DOX plus laser irradiation produced maximal cancer cell killing efficiency. Adapted with permission from reference [57].

12

ACCEPTED MANUSCRIPT Fullerene derivatives are also useful for fluorescence imaging of cancer. One example is a hyaluronated (for CD44-targeting) C60 fullerene with a strong intrinsic NIR fluorescence

PT

emission. The hyaluranoted C60 fullerene was used for both in vivo fluorescence imaging of

RI

HCT-116 tumors (CD44+) and photodynamic killing of tumors [53]. Since the intrinsic fluorescence signal from fullerene is not very strong, external fluorophores could also be

SC

attached to facilitate the visualization of fullerene complexes in vivo. For example, a hybrid

NU

nanoconjugate was formed from the condensation between hyaluronated C60 fullerene and upconversion nanocrystals [54]. Although only cell imaging and ROS validation were carried out

MA

in this study, this fullerene complex could be potentially used for in vivo fluorescence imaging and PDT.

TE

D

Despite their usefulness as imaging contrast agents, fullerene derivatives are rarely adopted solely for cancer imaging/detection purposes. With synergistic integration into other

AC CE P

applications (e.g. drug delivery), imaging can also provide guidance to assess delivery efficiency or therapeutic responses.

3.2. Cargo delivery to cancer with fullerenes The unique structure of fullerene derivatives render them broadly useful for delivery of different cargos. Recently, an Asn-Gly-Arg (NGR, against CD13 on the tumor cells) functionalized

C60 fullerene was used as a targeted delivery system for a PDT agent (2-

methoxyestradiol [2-ME]) [55]. Joint photosensitization effects were witnessed for C60 fullerene to boost the cell apoptosis efficacy of 2-ME on MCF-7 breast cancer cells. Other chemotherapy drugs, such as doxorubicin (DOX) and pacilitaxel, were also loaded into C60 [56, 57], which was confirmed to possess enhanced tumor killing capacity over free individual drug. In one of these studies, cancer cell death was the consequence from synergistic pH-sensitive DOX release and PDT-mediated ROS (Figure 2B) [57]. 13

ACCEPTED MANUSCRIPT With the introduction of proper cationic groups, fullerenes can also be attractive carriers for gene delivery due to the carbon cages inside fullerene [58, 59], which can stabilize DNA-

PT

fullerene complexes. A cationic tetra (piperazino) fullerene epoxide (TPFE) was synthesized as

RI

an in vivo gene delivery carrier [59]. The initial validation was carried out after loading the fullerene with DNA encoding enhanced green fluorescent protein gene (EGFP). The gene

SC

delivery by TPFE was efficient and organ selective, causing no noticeable toxicity. Although

NU

cancer-selective gene delivery (e.g. therapeutic siRNA) with nontubular fullerenes has not been carried out to date, these studies validated the value of using fullerenes as a gene delivery vector

MA

for cancer treatment.

Various fullerene-based multifunctional vectors (e.g. combination of drug delivery with

TE

D

imaging or PDT/PTT) have been developed recently [52, 60-62]. One recent example was the utilization of a PEGylated fullerene decorated with IONPs for targeted delivery of PDT-sensitive

AC CE P

hematoporphyrin monomethyl ether (HMME), readily monitored by MRI [63]. The PDT efficacy of HMME delivered by this composite was dramatically improved both in vitro and in vivo when compared with that of free HMME, monitored by MRI and other evidences. 4. THERANOSTIC APPLICATIONS OF CARBON NANOTUBES Although strictly speaking, carbon nanotubes (CNTs) belong to a sub-category of fullerene derivatives, their wide-spread biomedical applications necessitate a separate section to illustrate their leading role for cancer theranostics inside the whole carbon nanomaterial family. CNTs are carbon allotropes which possess a cylindrical nanostructure with a very high length-to-diameter ratio. According to the numbers of sheets of sp2 carbon atoms, CNTs can be generally categorized into single-walled nanotubes (SWNTs) and multi-walled nanotubes (MWNTs). CNTs retain several unique properties, including strong resonant Raman scattering and photoluminescence in the NIR region, and good laser-to-acoustic transferring capacity [64]. The 14

ACCEPTED MANUSCRIPT strong NIR absorption of CNTs enables them to be used as a potential candidate for cancer PTT. The large area inside and outside CNTs provide tremendous accommodation to various

PT

therapeutic molecules [25].

RI

4.1. Cancer imaging with CNTs

SC

We will focus on the updates within the passing five years since the progress in molecular imaging with SWNTs has been summarized previously [65]. Among all the imaging

NU

techniques, fluorescence imaging is the most frequently adopted for CNTs in cancer imaging/detection. The intrinsic emission in the second NIR region (1100-1400 nm, NIRII) from

MA

CNTs could be readily used for in vivo tumor detection [66, 67]. Since the initial success of CNT imaging producing high-resolution tumor vessel visualization [19], different studies have

TE

D

confirmed that CNTs do in fact produce high signal-to-noise ratios. Most organisms investigated have extremely low absorption in the second NIR wavelength region [68, 69]. In an interesting

AC CE P

study, M13 phage-conjugated SWNTs with stable display of tumor-targeting sequence were used for NIR fluorescence imaging of prostate tumors in mice [70]. The same group improved their results by adopting a new tumor-targeting sequence on M13-SWNTs, which could facilitate the detection and excision of tumors with submillimeter diameters (Figure 3A) [71]. At the same time, CNTs could also be conjugated with multiple fluorophores with different emission wavelengths (e.g. quantum dots [QD] and polydiacetylene) [72, 73] for fluorescence imaging of cancer cells based on fluorescence resonance energy transfer (FRET). Besides fluorescence imaging, bioluminescence imaging was also used recently to monitor the distribution of CNTs in vivo after conjugation of a thermostable luciferase onto CNTs [74].

15

ACCEPTED MANUSCRIPT Figure 3 Unguided M13-SWNT guided tumor excision

M13-SWNT guided

Post-surgery (unguided)

(B)

MA

NU

1h

4h

Post-surgery (M13-SWNT guided)

SC

Pre-surgery

RI

PT

(A)

D

SWNT-PEI /siRNA

SWNT-PEI /siRNA/NGR

Upper: SWNT-PEI/siRNA/NGR Down: SWNT-PEI/siRNA/NGR + laser

AC CE P

TE

Legend to Figure 3. (A) The use of M13 functionalized SWNTs for NIR fluorescence-guided tumor excision. Peptide sequence targeting secreted protein, acidic and rich in cysteines (SPARC) was displayed on M13. The fluorescence from M13-SWNTs was successfully used for detection and excision of submillimeter tumors which were not removable without M13-SWNTs guidance. SBP stands for SPARC binding peptide. Adapted with permission from reference [71]. (B) The structure of SWNT-PEI and its application for siRNA/PDT synergistic therapy of prostate tumors. SWNT-PEI loaded with siRNA (with or without NGR targeting peptide) could enter nucleus of PC-3 cells for effective siRNA delivery at 4 h post-treatment. In vivo siRNA/PTT demonstrated significantly beneficial therapeutic response when compared with only RNA interference induced by siRNA loaded, NGR conjugated SWNT-PEI (SWNTPEI/siRNA/NGR). Adapted with permission from reference [104]. Another primary application for CNTs in cancer imaging is to serve as an ultrasound contrast agent [75, 76] or a PAI agent [77, 78]. PAI detects the ultrasound signals excited by optical lasers, and it promises to have much higher spatial resolution over fluorescence imaging since ultrasound is less scatterable than photons [79]. The initial PAI study of CNTs was carried out in 2008, in which cyclic Arg-Gly-Asp (RGD) functionalized SWNTs showed strong accumulation into the tumor [21]. Afterwards, different strategies were used to improve PAI contrast for CNTs. For example, SWNTs were recently conjugated with indocyanine green (ICG) and used to identify sentinel lymph nodes (SLNs), with approximately four folds of signal enhancement being observed after ICG conjugation [80]. Another research group doped silicacoated gold nanorods (GNRs) onto the surface of MWNTs to boost PAI signals for gastric cancer delineation [81]. 16

ACCEPTED MANUSCRIPT The Raman scattering is useful for long term monitoring of CNTs since it is durable and resistant to photo-bleaching. Multicolor Raman imaging of live cells has been reported using C) and a single laser excitation [23]. By

12

C/13C isotope compositions, five-color Raman imaging with SWNTs was done in a

RI

changing

13

PT

SWNTs containing different carbon isotopes (12C or

follow-up study [82]. The same research group also reported the first ex vivo and in vivo Raman

SC

imaging of CNTs [21, 24]. With the support of Raman imaging, the intracellular distribution of

NU

CNTs could be determined post treatment in cancer cells [83]. With the coating of noble metals

imaging and photothermal therapy [84].

MA

(e.g. gold or silver), CNTs can also be used for both surface enhanced Raman scattering (SERS)

Due to the chemical versatility and structural uniqueness, CNTs can be also used in other

TE

D

imaging techniques, including PET [85], MRI [86, 87], or multimodality imaging [88-91].

AC CE P

4.2 Cargo delivery to cancer with CNTs CNTs have been intensively investigated as drug carriers, with DOX being the most common model drug [92]. DOX-loaded CNTs have a potent accumulation in different tumor types and the on-demand release of DOX is witnessed upon the delivery of CNTs to the tumor [93]. Stimuli-responsive cargo delivery (e.g. promotion of drug uptake/release in tumor by a laser) into tumors is also achievable by utilization of CNTs’ unique optical properties [94]. The aforementioned imaging capacity of CNTs allows them to be used as imaging guided therapeutic platforms for cancer as well. When accompanied with a targeting ligand (i.e. a molecule with strong affinity against a given receptor overexpressed in a tumor), CNTs could be adopted as tumor-selective delivery vectors. Functionalized MWNTs were used for the delivery of DOX to glioma by utilization of angiopep-2 (ANG) as the targeting ligand (for low-density lipoprotein receptor-related protein [LRP] overexpressed in glioma) [95]. Improved therapeutic efficacy was observed for DOX17

ACCEPTED MANUSCRIPT MWNTs-PEG-ANG when compared with free DOX. Another example of targeted delivery of DOX is the use of chitosan modified SWNTs conjugated with folic acid (FA) in order to target

PT

folate receptor (FR) positive cancer cells [96]. The SWNTs effectively inhibited the growth of

RI

hepatocellular carcinoma with less observable side effects over free DOX. In most cases, anticancer drugs were loaded onto CNTs through noncovalent interactions, although covalent

SC

binding can also be employed for hydrophilic drugs. For example, cisplatin and epidermal

NU

growth factor (EGF) were covalently attached to SWNTs and had enhanced killing efficacy of cancer cells that were positive for EGF receptors [97].

MA

With proper functionalization, CNTs can also be a good gene delivery vector of plasmid DNA, small interfering RNA (siRNA), and micro RNA [98, 99]. For example, SWNTs were

TE

D

functionalized non-covalently with succinated polyethyleimine (PEI-SA) in one study and loaded with siRNA for transdermal siRNA delivery in a murine melanoma model with an attenuated

AC CE P

tumor growth being observed [100]. CNTs have also been used for thermal therapy [101] and PDT agent delivery [102, 103]. A recent trend has been to combine different cargo loadings into CNTs to maximize the therapeutic response. SWNTs were functionalized in one recent report with PEI as well as tumor-targeting NGR peptide for both siRNA delivery and PTT [104]. In tumor-bearing mice the delivery system exhibited potent tumor accumulation without obvious toxicity in main organs, and the combination of RNAi and PTT significantly enhanced the therapeutic output (Figure 3B).

18

ACCEPTED MANUSCRIPT 5. THERANOSTIC APPLICATIONS OF SINGLE WALLED CARBON NANOHORNS As a close relative (with conical structure) to CNTs, carbon nanohorns (CNHs) have certain

PT

unique and attractive properties, although their use in biomedical applications is still at a preliminary stage. Single walled carbon nanohorns (SWNHs) are the chief member of the CNH

RI

family – defined as an assembly of tubular units that are 2-5 nm in diameter and 40-50 nm in

SC

length [105, 106]. According to the different morphologies of these aggregates, SWNHs can be

NU

classified into either dahlia-like type, bud-like type or seed-like type [32]. The dahlia-like SWNHs were most frequently used for cancer theranostic applications, which are composed of

MA

nearly 2,000 tubular unites and have a spherical structure with a diameter of 80-100 nm [107, 108]. SWNHs have many properties in common with SWNTs due to structural similarity.

D

However, SWNHs have several advantages over SWNTs including more uniform and

TE

controllable morphology [109], easier large-scale production and free of metal contamination

AC CE P

(considered to be the main attribution to CNTs’ toxicity) [110], and improved biocompatibility [111]. Hence, SWNHs are considered to have a higher potential for clinical use [112]. 5.1. Cancer imaging with SWNHs As a comparatively new material for biomedical applications, cancer imaging studies with SWNHs are quite limited. The primary imaging techniques adopted is MRI since SWNHs can be easily complexed with gadolinium [113, 114] or IONPs [115] (this property is shared by other carbon nanomaterials with similar structure such as fullerene or CNTs). For example, Gd3N@C80@SWNHs was coupled with QDs in one study to visualize U87 tumors in the mouse brain with MRI [116]. IONPs have also been stably attached onto SWNHs with holes opened (SWNHox) via a thermal transformation [117] – the as-prepared IONP-functionalized SWNHox was very stable and had clear detection in the T2-weighted MRI images.

19

ACCEPTED MANUSCRIPT Recently, SWNHs were used as a PAI guided PTT platform [118]. A PEG derivative, poly(maleic anhydride-alt-1-octadecene-PEG) (C18PMH-PEG), was utilized to functionalize

PT

SWNHs (Figure 4A). The obtained SWNHs/C18PMH-PEG was highly dispersible and stable in

RI

aqueous solutions. With strong absorbance in the NIR region, it could be used for synergistic PAI/PTT. After the accumulation of SWNHs/C18PMH-PEG in the peripheric vessels of the

SC

tumor, the boundary of tumor was clearly delineated in PAI to achieve more accurate tumor

NU

ablation.

AC CE P

TE

D

MA

Figure 4

Legend to Figure 4. (A) The structure and application of SWNHs/C18 PMH-PEG for PAI guided PTT. The PA images of tumor sites at different time were shown for mouse injected with SWNHs/C18 PMH-PEG. Whole animal images before and after laser irradiation were also shown for mouse injected with SWNHs/C18 PMH-PEG or saline. Adapted with permission from reference [118]. (B) The schematic illustration of SWNHs used as a tool for photothermally enhanced DOX release in tumor. Thermal images and histological examination confirmed the temperature rise in tumor area, which demonstrated the boosted cancer killing efficacy from both DOX and PTT. Adapted with permission from reference [119].

20

ACCEPTED MANUSCRIPT 5.2. Cargo delivery to cancer with SWNHs The large areas both outside and inside the horn structure of SWNHs allow for

PT

accommodation of different cargo molecules [107, 108]. Recently, DOX-loaded SWNHs (DOXSWNHs) were used for photothermal enhanced chemotherapy [119]. After stabilization with

RI

chitosan, the obtained complex demonstrated good biocompatibility and photothermal properties.

SC

A mild heating from the laser irradiation was found to effectively promote the release of DOX from the complex, therefore effectively enhancing the toxicity of DOX to cancer cells (Figure

NU

4B). Another example of SWNH drug loading is cisplatin loaded in SWNHox via a solvent

MA

evaporation procedure [120]. The incorporated cisplatin was completely released from SWNHox resulting in increased anticancer efficacy. The same group has used SWNHox for loading zinc

D

phthalocyanine (ZnPc) for synergistic PDT/PTT [121]. After subsequent functionalization with

TE

BSA, a single 670 nm laser was used to trigger PDT/PTT at the same time. Almost full tumor

phototherapy.

AC CE P

disappearance post treatment was observed, confirming the potential of SWNHs in cancer

SWNHs have shown promise as effective gene carriers as well [108]. Polyamidoamine (PAMAM) modified SWNHs were used to deliver siRNA into prostate cancer cells (PC-3), and the results showed initial evidence of gene transfection by SWNHs with decreased toxicity to normal cells [122]. This hybrid platform possesses several advantages: the PAMAM functionalization provides strong electrostatic interactions with DNA/RNA, the PAMAM functionality also provides good solubility while decreasing toxicity, and the PAMAM allows for further biological modifications. Another application of SWNHs in gene therapy is to utilize the photothermal effect induced by SWNHs for regulation of gene expression [123]. For example, BSA modified SWNHs were combined with NIR laser irradiation to trigger heat shock

21

ACCEPTED MANUSCRIPT promoter-mediated gene expression. The positive results of this study provide an important insight in the development of light-triggered gene manipulation.

PT

6. THERANOSTIC APPLICATIONS OF CARBON NANODIAMONDS As another important member of carbonaceous nanomaterial family, nanodiamond

RI

(ND) has an octahedral architecture ranging in size from 5-50nm. NDs are becoming

SC

increasingly useful in therapeutic and diagnostic applications due to their biocompatibility,

NU

scalability, and easy surface modification [124]. NDs also possesses an inherent stable fluorescence with the introduction of nitrogen into its internal structure.

MA

6.1. Cancer imaging with nanodiamonds

NDs are effective imaging agents due to their bright intrinsic fluorescence after the

15

N or

13

C, for fluorescence imaging or MRI [124]. The amount of nitrogen

TE

isotopes, such as

D

addition of nitrogen and easy modification with an organic fluorophore or MRI detectable

AC CE P

implemented into the ND allows for control over the fluorescence intensity and emission [125], which could be used for single molecule tracking inside a cell. The inherent fluorescence intensity from an ND is significantly higher compared to an AlexFluor dye at the same wavelength, so the ND does not need to be further functionalized in order to have strong imaging capabilities [126]. This strong fluorescence from NDs is particularly useful for low-background imaging in vivo [127]. In order to obtain even higher brightness from the fluorescence of the ND, helium has been used to bombard the ND powder [128]. With a larger degree of vacancy caused by helium ions inside the ND, more nitrogen was introduced, increasing the fluorescence intensity observed from the ND. Despite the active research into ND fluorescence, it was not until recently that fluorescent NDs were used for in vivo tracking of cancer cells in a murine model (Figure 5A) [129].

22

ACCEPTED MANUSCRIPT Figure 5

NU

DOX

MA

(B)

SC

RI

PT

(A)

D

ND-DOX (NDX)

4 h efflux

TE

1 h incubation

AC CE P

Legend to Figure 5. (A) Fluorescent NDs were used for in vivo lung cancer cell tracking in a murine model Adapted with permission from reference [129]. (B) DOX-loaded ND could prevent DOX from efflux from cancer cells and boost the therapeutic response in a murine liver cancer model. Adapted with permission from reference [137].

23

ACCEPTED MANUSCRIPT There has been no report to date adopting magnetic NDs for in vivo detection of cancer, although some researchers claimed that Gd-NDs conjugates could be considered “game

PT

changing” and provide dramatically enhanced MR signal intensity [130-132]. To prepare NDs

RI

for cancer detection/imaging applications, various imaging labels were simultaneously incorporated into NDs. For example, one early study explored the in vivo distribution pattern of

SC

amino-functionalized NDs by using the positron-emitting isotope

18

F (t1/2 = 110 min) as the

NU

imaging label [133]. Iron nanoparticle (ferrocene) bound NDs were used in another study for fluorescence and MRI based cancer cell tracking [134]. Also, a new imaging concept, based on

MA

the optical detection of NDs in a magnetic field gradient, was proposed recently to achieve higher sensitivity and ND detection with better spatial resolution [135]. This imaging method has

TE

D

been named “nanodiamond imaging”, and it could be extremely useful for applications such as circulating tumor cell (CTC) tracking or stem cell tracking in vivo.

AC CE P

6.2. Cargo delivery to cancer with nanodiamonds NDs can be useful vectors to provide cancer-targeted drug delivery with adjustable blood circulation and potentially decrease drug resistance of cancer cells. It was shown that NDs conjugated with DOX embedded in a microfilm produced a long term slow release of DOX for over a month [136].

ND-DOX conjugates were recently used in murine models of mammary and liver cancer to determine their distribution compared to free DOX [137]. The ND-DOX conjugates showed increased tumor growth inhibition and apoptosis compared to typical DOX treatment (Figure 5B) with decreased chemotherapeutic toxicity. Consistent therapeutic effect with better kidney and liver tolerance was confirmed in another report [138], and similar results were also obtained in a lung cancer model [139]. Other ND-drug conjugates also have shown improved tumor therapy efficacy, including 10-hydroxycampotothecin (HCPT) [140] and daunorubicin [141]. 24

ACCEPTED MANUSCRIPT Besides the loading of chemotherapeutic drugs, NDs can also be used for carrying proteins [142] or genes [143-146]. After conjugation with cationic compounds such as PEI [146],

PT

protamine sulfate [145], or cationic polymers [143, 144], NDs demonstrated potent siRNA

RI

delivery efficiency into various types of tumors. Unlike previously mentioned carbon nanomaterials, NDs are not intrinsically usable for PDT or PTT of cancer, but NDs can still be

SC

used for delivering PTT/PDT agents to cancer cells after proper hybridization. For example, NDs

NU

were complexed with gold/silver nanoparticles and used for efficient photothermal killing of

MA

cancer cells [147].

7. THERANOSTIC APPLICATIONS OF CARBON NANODOTS

D

Carbon nanodots (C-dots) are quantum sized carbon analogues with bright and tunable

TE

photoluminescence [148]. Interestingly, the C-dots are photoluminescent only after surface passivation. Massive research efforts have been devoted to utilize the bright and colorful

AC CE P

luminescent agents for imaging applications [149-152]. Unlike other carbonaceous nanomaterials, the C-dots have inherent aqueous solubility: no complicated functionalization is necessary for CDots to be stabilized in buffers. Large scale production of C-dots could be economically practical since many cheap and widely available materials can be used for C-dot synthesis [153-155]. Cdots are considered much more biocompatible compared to other materials, such as quantum dots, since heavy metal catalysts is not required in their synthesis.

25

ACCEPTED MANUSCRIPT 7.1. Cancer imaging with carbon nanodots Like NDs, the intrinsic photoluminescence in C-dots allows the material to be used as an

PT

imaging agent. Produced initially by laser ablation and nitric acid treatment [148], C-dots possess bright and colorful luminescence with a strong dependence (tunable) on the excitation

RI

wavelength. The first in vivo imaging of these materials used C-dots doped with ZnS in order to

SC

increase their fluorescence [156]. It was found that C-dots retained their strong

NU

photoluminescence in vivo with no observable acute toxicity to the tested mice. In another study, identical C-dots with diameters of 3–4 nm were obtained from carbon nanotubes and graphite

MA

[157]. Fluorescence imaging was also carried out to study the distribution of these C-dots. C-dots have also been applied in cancer cell identification [158] and fluorescence imaging-guided drug

D

delivery [159].

TE

Aside from fluorescence imaging, C-dots can be readily used for multimodal imaging.

AC CE P

For example, after doping with gadolinium, C-Dots were used for both fluorescence imaging and MRI [160]. In another report, the distribution, clearance and tumor uptake of C-dots were systematically investigated by fluorescence and PET (Figure 6A) after conjugation with positron-emitting

64

Cu (t1/2 = 12.7 h) and a NIR fluorophore [161]. The combination of NIR

imaging and PET imaging could provide synergistic information to assess the in vivo fate of Cdots.

26

ACCEPTED MANUSCRIPT Figure 6

(B)

RI

PT

(A)

NU

SC

0.05 (scaled counts/ g)

0 s.c

i.m.

MA

i.v.

TE

D

Legend to Figure 6. (A) The schematic structure of C-dots used for in vivo imaging and the fluorescence images of tumor-bearing mice with different administrative ways of C-dots: subcutaneous injection (s.c.), intramuscular (i.m.) injection and intraveneous injection (i.v.). White arrow indicates tumor; red arrow indicates kidney. Adapted with permission from reference [161].

AC CE P

(B) Scheme for synthesis and theranostic application of CD-Oxa. In vivo fluorescence images and the tumor size of tumor-bearing mouse at different time intervals after injection of CD-Oxa. Adapted with permission from reference [162].

27

ACCEPTED MANUSCRIPT 7.2. Cargo delivery to cancer with carbon nanodots C-dots can also serve as a carrier for delivery of cancer therapeutics. One example uses

PT

C-dots covalently attached to a chemotherapeutic [162]. C-dot oxaliplatin conjugates significantly improved the anticancer efficacy of oxaliplatin. Moreover, the distribution and the

RI

pharmacokinetics of this C-dot conjugate could be monitored in a real-time scale due to the

SC

inherent imaging capabilities of C-dots (Figure 6B), which facilitated the optimization of the administration of drugs and the therapeutic evaluation after treatment. Different complexes from

NU

C-dots (e.g. C-dots@GNRs or C-dots@mesoporous silica nanoparticles) have also been formed

MA

for the controlled release of DOX [159, 163]. The drug loading efficiency of the C-dot DOX hybrid platforms was very high and the release of DOX was triggered under laser irradiation by

D

the photothermal effect [163] or pH-mediated dissociation [159]. The strong inherent

TE

fluorescence of the C-dots was used for determining the distribution of DOX.

AC CE P

C-dots can also serve as vectors for transferring DNA/RNA, proteins, or PDT agents. For example, PEI adsorbed C-dots were used in one study to deliver siRNA against survivin into human gastric cancer cells [164]. The C-dot complex induced more than 90% of the target protein knockdown, resulting in significant cell apoptosis. Ribonuclease A was loaded onto Cdots in another study for simultaneous therapy and fluorescence imaging in gastric tumor-bearing mice [165]. After conjugation with a photosensitizer, C-dots were used for simultaneous fluorescence imaging and PDT [166-169]. The C-dots/PS conjugation strategy has several advantages including therapeutic response monitoring [166, 169], extended tissue penetration of conventional PSs [167], and laser power decrease for effective PDT [168].

28

ACCEPTED MANUSCRIPT 8. THERANOSTIC APPLICATIONS OF NANOGRAPHENES Another category of carbon nanomaterial is nanosized graphene which includes its

PT

derivatives graphene oxide (GO), reduced graphene oxide (RGO), and hybrid graphene nanostructures [170-172]. Graphene is a two-dimensional (2d) carbon sheet assembled in a

RI

honeycomb structure, and it can be used as the basic composing piece for other carbon materials

SC

of different dimensions [173]. The unique physical and chemical characteristics of graphene (and

NU

its derivatives), such as ultra-high surface area, robust mechanical strength, rapid electron transfer ability, and versatile surface modification capacity, attracts enormous research interests.

MA

8.1 Cancer imaging with nanographenes Cancer imaging with nanographenes is as effective as using CNTs. An initial PET

D

imaging of cancer study with GO nanosheets used an antibody against CD105 as the targeting

TE

ligand with 64Cu as a PET label. The targeted GO nanosheet resulted in enhanced tumor uptake.

radiogalium

66

AC CE P

To broaden the clinical applicability of the GO conjugates, other PET labels including Ga (t1/2 = 9.3 h) [175] and

61

Cu (t1/2 = 3.3 h) [176] were used. In another study,

vascular endothelial growth factor 121 (VEGF121) was selected as a targeting ligand to further improve tumor targeting efficiency (Figure 7A) [177]. The antibody and

64

Cu were attached

onto RGO conjugates with a slightly modified conjugation strategy (i.e. noncovalent stacking of C18PMH-PEG-NH2 was used to functionalize RGO surface instead of covalent binding) [178]. Conjugation strategies of graphene derivatives for biomedical applications have been reviewed in detail [179].

29

ACCEPTED MANUSCRIPT Figure 7 NOTA

(A)

(B)

Fluorescence

PT

64Cu

PAI 64Cu-NOTA-GO-PEG-VEGF

121

RI

VEGF121

10 %ID/g

Treatment

-VEGF121

0 %ID/g

NU

64Cu-NOTA-GO-PEG 64 Cu-NOTA-GO-PEG

SC

100 nm

PDT + PTT

PDT

PTT

Saline

MA

Legend to Figure 7. (A) The structure and applications of graphene derivatives as a PET imaging contrast agent for tumor detection. Adapted with permission from reference [177].

AC CE P

TE

D

(B) An imaging-guided hybrid PDT/PTT therapy nanovector based on graphene oxide. Adapted with permission from reference [228].

30

ACCEPTED MANUSCRIPT Researchers from Oxford University conducted the initial research report of using GO as a scaffold to create SPECT imaging agents [180].

111

In (t1/2 = 2.8 d) was adopted as the

PT

radiolabel and its chelator, p–NH2–Bn-DTPA, was introduced onto the surface of GO via π-π

RI

stacking. An anti-HER2 (human epidermal growth factor receptor 2) antibody, trastuzumab, was also conjugated onto the GO surface to increase uptake in HER2-positive tumors.

111

In-GO-

SC

trastuzumab could not only detect primary tumor site, but also locate metastases in lymph nodes.

199

198,199

Au (t1/2:

198

Au: 2.7 d;

NU

In a recent report, GO sheets were also shown to be labeled with an

Au: 3.1 d) nanoparticle (named 198,199Au@AF-GO) [181]. In rats bearing fibrosarcoma tumors, Au@AF-GO exhibited fast penetration and strong accumulation (a surprisingly high tumor-

MA

198,199

D

to-muscle ratio of 167 could be obtained at 4 h post-injection).

TE

Three major types of MR label can be used to produce graphene-based MRI contrast agents – IONPs, manganese (Mn2+), and Gd. Initial production of the different MR labelled

AC CE P

nanosheets was carried out by decomposition of an iron precursor onto RGO via high temperature [182]. MnFe2O4 nanoparticles were also used to decorate GO to serve as a potential T2 contrast agent for MRI [183]. Mn2+–intercalated graphene nanoplatelets functionalized with dextran has been another potential choice as an MRI agent [184]. Fluorinated GO (FGO) is another interesting category of MRI detectable graphene derivatives [30]. FGO is used as a magnetically responsive drug carrier which can potentially be used for both MRI and photoacoustic imaging [185].

31

ACCEPTED MANUSCRIPT Similar to CNTs, nanosized GO produce fluorescence in the visible and IR regions [186]. The IR-A fluorescence emissions (~1100 to 2200 nm) from GO can be detected by an InGaAs

PT

detector. Despite successful cell imaging with low background, the inherently low quantum yield

RI

of GO fluorescence hinders its in vivo applications. Adopting direct emission from GO as an imaging label is not optimal due to the limited resources of an InGaAs detector. To address

SC

direct emission limitations, different fluorophores were conjugated onto graphene derivatives.

NU

One example is a Cy7 labeled, PEG-functionalized GO nanosheets used for fluorescence tumor detection [187]. From this study, increased uptake was observed in tumors, with relatively low

MA

accumulation in the mononuclear phagocyte system (MPS). QD-tagged RGOs with bright

TE

tracking of drug behavior [190].

D

fluorescence were also used for live cell imaging [188], tumor imaging and PTT [189], and

Another emerging fluorescent graphene is a graphene quantum dot (GQD), which can be

AC CE P

produced from a variety of synthetic methods [191, 192]. Compared to “conventional” heavy metal based QDs, these GQDs are chemically inert, and usually possess good solubility, photostability, low toxicity [193], and high quantum-yield fluorescence emissions. Tumortargeting ligands such as hyaluronic acid (for CD44) [194] or folic acid [195] could be easily coupled onto GQDs for tumor targeting. In addition to tumor cell detection and recognition, GQDs can also be useful to monitor biochemical pathways, such as apoptosis [196]. Different GQD based hybrid nanostructures, primarily silica nanomaterials-GQDs [197-199], have also been synthesized for fluorescence imaging and drug delivery applications.

32

ACCEPTED MANUSCRIPT GOs were also considered highly promising PA contrast agents [200]. In a more recent study, GO nanosheets were produced with a modified Hummers method by microwave-assisted

PT

pure nitronium ion oxidation instead of KMnO4. The resulting GO nanosheets exhibited strong

RI

absorption in the visible and NIR regions and strong PA signals from NIR excitation [201]. Strong NIR absorption and excitation from these materials is useful for long-term tracking of

SC

cancer cells. BSA was used in another study as both a reductant and stabilizer to a nanosized

NU

RGO for both PAI and PTT [202].

MA

8.2 Cargo delivery to cancer with nanographenes The delocalized π electrons and highly available surface area on graphene derivatives provide good loading capacity for various molecules. Despite the fact that they have been

D

frequently adopted as drug/gene delivery vectors, in vivo drug delivery with graphene derivatives

TE

are still in its infancy. The overall benefits of nanographene as delivery vectors of anti-cancer

AC CE P

drugs and genes have been reviewed elsewhere [26, 171]. DOX was loaded onto GO-IONP-PEG to be used for MRI detectable and magnetically targeted drug delivery [203]. T2 MRI demonstrated accumulation of GO-IONP-PEG in the tumor site: this study for the first time established the role of graphene derivatives as an in vivo drug delivery vector. Following a similar strategy, a GO-based T1 MRI detectable nanoplatform was developed via a GO-gadolinium complex [204], which showed strong uptake and subsequent cytotoxicity in HepG2 cancer cells after DOX loading. By chemical deposition of silver nanoparticles onto GO, GO@Ag was used for DOX loading and selective tumor targeting after NGR peptide (against CD13) conjugation [205]. With better DOX delivery to the tumor and an improved release profile over free DOX, the nanomaterial could serve as an X-ray contrast agent and a platform to combine local chemotherapy with PTT. GO nanosheets with PAI capacity was 33

ACCEPTED MANUSCRIPT also developed with DOX loading, which monitored the therapy response over time and revealed subtle microvascular changes during chemotherapy [206].

PT

Graphene derivatives are active candidates for directing a gene to the cancerous sites.

RI

They have been used for delivering DNA [207] or RNA [208] to cancer cells, with PEI [209] or

SC

chitosan [210] being two primary mediators to improve the DNA/RNA binding and condensation. For example, GO-PEG-PEI was used in a study and showed increased gene

NU

transfection efficiency as well as reduced cytotoxicity compared to free PEI [211]. Interestingly,

MA

the authors utilized the NIR absorbance of GO to enhance the cellular uptake of GO-PEG-PEI by using a low power NIR laser irradiation to increase membrane permeability. This GO-PEG-PEI

D

was able to deliver siRNA into cells under NIR irradiation with obvious down-regulation of the

TE

target gene. The use of the NIR laser irradiation is known as “light-controllable gene delivery”. Another study used similarly structured GO conjugates for delivering Stat3-specific siRNA into

[212].

AC CE P

melanomas to produce significant regression in tumor growth due to increased gene transfection

Nanographenes were demonstrated to have higher heat production capacity when compared with CNTs under the same conditions [213]. An initial PTT study used Cy5 labeled GO nanosheets to determine long circulation time and high tumor accumulation [187]. A laser energy density of ~ 2 W cm-2 was used for tumor irradiation with efficient tumor ablation. GOIONP-Au-PEG was also synthesized with improved photothermal tumor ablation [214]. Another IONP-GO complex was also formed by one-step adherence of IONP to carboxylic GO via hydrophobic interactions [215], which displayed a good photothermal efficiency. Recently, BaGdF5 nanoparticles were attached on the surface of GO nanosheets to form the GO/BaGdF5/PEG [216]. Efficient tumor penetration of GO/BaGdF5/PEG led to efficient 34

ACCEPTED MANUSCRIPT photothermal ablation of tumor in vivo, monitored by MRI and CT. GO nanoconjugates can also be used for diagnosis and ablation of tumor-drained regional lymph nodes [217].

PT

The reduction of GO can lead to enhanced absorbance in the NIR region [171], so RGOs

RI

can be considered more ideal for PTT applications. Single-layered RGO nanosheets (~ 20 nm) exhibited 6-fold higher NIR absorption that other non-reduced GO materials [218]. RGO-PEG

SC

conjugates were used to completely eliminate tumors by low-power laser irradiation in tumor

NU

area (0.15 W cm-2) post intratumor injection of RGO-PEG [219]. A RGO-IONP-PEG complex was also developed with good tumor targeting and improved PTT tumor ablation over GO [220].

MA

RGO nanomesh (RGONM) was another recently developed structures of graphene with over 20fold higher NIR absorption [221]. As-synthesized RGONMs were functionalized by PEG, RGD,

TE

laser power (0.1 W cm-2).

D

and Cy7 and completely eliminated U87MG tumors followed by irradiation with an ultralow

AC CE P

The first study of PDT with graphene derivatives used ZnPc as a PS [222]. The GO-ZnPc complex was internalized in MCF-7 cells, exhibiting a pronounced cytotoxicity under Xe light irradiation. Adopting a similar PS-attaching strategy, a folic acid-conjugated GO was used to load chlorin e6 (Ce6) for selective PDT in tumor cells [223]. Clinical translation of current PDT agents are often limited by their low singlet oxygen (1O2) quantum yields. One GQD-based PDT agent could produce 1O2 via a multistate sensitization process, resulting in a quantum yield of ~1.3, the highest recorded for PDT agents [224]. The fluorescence of PSs are often drastically quenched via an energy/charge transfer process after their interactions with graphene derivatives, which sometimes limits their detectability. To solve this problem, a PTT agent was prepared from sinoporphyrin sodium (DVDMS) loaded, PEGylated GO (named GO-PEG-DVDMS) which possessed more stable fluorescence for imaging guided PDT [225]. The uniqueness of 35

ACCEPTED MANUSCRIPT DVDMS is that its fluorescence could be enhanced via intramolecular charge transfer post loading onto GO-PEG. Another PS molecule, 2-(1-hexyloxyethyl)-2-devinyl pyropheophorbide-

PT

alpha (HPPH), was loaded onto PEG-GO. In vivo distribution and delivery of the obtained 64

Cu

RI

complex could be tracked by both fluorescence imaging and PET after radiolabeling with [226]. Improved cancer cell killing efficacy was witnessed in all of these reports.

SC

Graphene-based nanoplatforms are useful for multiple hybrid therapies, such as

NU

PDT/PTT and PTT/chemotherapy. Carboxylated graphene nanodots (cGdots) has been used for imaging-guided PDT/PTT without requirement of an extra PS [227]. cGdots have shown to

MA

effectively shrink MDA-MB-231 tumors compared with a saline-treated control group upon NIR laser irradiation. The luminescence emission from cGdots enabled visualization of tumor tissue

TE

D

to better interpret the treatment efficacy in mice. DVDMS-loaded GO-PEG (GO-PEG-DVDMS) was also used in another study for combined PDT and PTT [228]. The synergistic PDT/PTT

AC CE P

efficacy was monitored by fluorescence/PA dual-modal imaging (Figure 7B). One graphene based nanohybrid was engineered via a facile one-pot process consisting of Fe3O4, Pluronic F127 (PF127), and graphene nanosheets [229]. DOX-loaded nanohybrids showed a significant cytotoxicity to HeLa cells, which could be intensified post laser irradiation. Another peptidemodified magnetic graphene-based mesoporous silica was synthesized with good NIR absorbance, facile magnetic separation, large T2 relaxation rates, and a high DOX loading capacity [230]. With DOX loading, it could integrate MRI, magnetic targeting and receptormediated active targeting, and chemo-photothermal therapy for a synergistic therapy of glioma.

36

ACCEPTED MANUSCRIPT Graphene derivatives are also good choices for hybrid cargo loading. For example, a chitosan functionalized magnetic graphene nanoparticle (CMG) was used for simultaneous gene

PT

and drug delivery [231]. After intravenous administration of GFP-plasmid encapsulated within

RI

DOX-CMGs into lung tumor-bearing mice, both GFP expression and DOX accumulation at the tumor site were shown. In another study, PAMAM-grafted Gd-GO nanoparticles were evaluated

SC

as carriers to deliver both chemotherapeutic drugs and miRNAs to cancer cells [232].

NU

9. CONCLUSIONS AND FUTURE PERSEPCTIVES As we can draw a preliminary conclusion based on aforementioned summarized research

MA

progress, carbonaceous nanomaterials are extremely useful tools for theranostic applications in cancer. The unique physical properties (e.g. intrinsic fluorescence or NIR absorbance), chemical

D

versatility, and superior cargo loading capacity are the prerequisite for them to be used as potent

TE

cancer therapeutics. The cancer imaging and cargo delivery applications of six leading players

AC CE P

(with CNTs and nanographenes as two leading horses) from carbonaceous nanomaterial family were discussed in this review article. We are fully aware that there are still other existing carbon nanostructures (e.g. carbon nano-onions [233]), which may be useful for theranostic applications in cancer. However, their limited case numbers restrained the devotion of a separate section for illustration – interested readers can refer to literature [234] for details. The structural integrity is critical for certain carbonaceous nanomaterials (e.g. fullerenes or CNHs), thus suitable medication/functionalization methods should be chosen carefully to improve their biocompatibility, solubility, and cargo loading efficiency (Table 1). Continued research into optimized surface modifications will allow for more effective loading and release of relevant moieties. The future research should be directed on development of functionalization strategies which can result in accurate release of the cargo from the vector. Although concepts such as “stimuli-responsive” drug delivery have been proposed utilizing the heat-driving cargo 37

ACCEPTED MANUSCRIPT release from carbonaceous nanomaterials [29, 94], the ability to control the system is still comparatively low (e.g. with limited tissue penetration), with inevitable heat damage to

PT

surrounding fragile tissues. To partially address this dilemma, drug loading with an environment

RI

sensitive chemical bond (e.g. pH-sensitive) can be helpful [235].

SC

With the increasing complexity of carbonaceous nanomaterials, knowledge of their in vivo kinetics is crucial to boost their performance. Imaging techniques are powerful tools to

NU

determine in vivo kinetics since they can not only provide the precise distribution profile of

MA

carbonaceous nanomaterials, but also give evaluation on therapeutic response [6]. However, each individual imaging technique has its pros and cons. For example, optical imaging

D

(fluorescence/bioluminescence/Raman) is highly sensitive and cost-effective with good security,

TE

but their tissue penetration is rather limited. MRI possesses high spatial resolution for tissues with good penetration depth, while its detection sensitivity for a given molecule is not optimal.

AC CE P

PET or SPECT imaging can provide highly sensitive and quantitative detection of trace materials, but the radiation exposure and relatively limited spatial resolution are major disadvantages. The integration of multi-modalities into a single platform may provide an answer to partially solve this conundrum – with different imaging modalities synergistically combined (e.g. MRI/PET to provide both high-resolution images and accurate molecular profile) in different studies for carbonaceous nanomaterials, more precise information can be collected to prepare them as the “next-generation” delivering vectors for cancer therapy.

38

ACCEPTED MANUSCRIPT To minimize the impact of carbonaceous nanomaterials to normal tissues/organs, their toxicity should be properly evaluated [236, 237]. As most reports claimed that carbonaceous

PT

nanomaterials are quite biocompatible after proper functionalization, skepticism can be raised

RI

due to their accumulation in cells from MPS (e.g. macrophages), incomplete excretion from the test subjects, and limited biodegradability. For example, induction of ROS from carbonaceous

SC

nanomaterials (especially with a tubular morphology) could be one leading factor to cause cell

NU

death [238]. At the same time, as metal catalysts are needed in the production of some carbonaceous nanomaterials (e.g. CNTs), heavy metal ions release from the material even after

MA

functionalization could be another source of toxicity. Combined with toxicological assessment methods, imaging techniques could be used to provide a better insight into their distribution,

TE

D

pharmacokinetic behavior, and eventually long-term toxicity. Some carbonaceous nanomaterials with small sizes, like fullerene, C-dots, were able to be cleared completely within weeks and do

AC CE P

not have long term toxicity concerns [27, 239]. There has also been some recent progress on the development of a “biodegradable” carbonaceous nanomaterial [240, 241], and will hopefully design a “deliver and degrade” vector with carbonaceous nanomaterials for ideal pharmacokinetic behavior and low long term toxicity. There is potential for future clinical usage of carbonaceous nanomaterials. With the multi-functionality of these materials, future developments can involve the production of carbonaceous nanomaterials with the capacity of detecting and responding to the dynamic changes of proteins or DNA/RNA in living systems (i.e. a “smart” surgeon-like material). Joint efforts from scientists and continued research can further develop advanced carbonaceous nanomaterials for cancer theranostic applications.

39

ACCEPTED MANUSCRIPT ACKNOWLEDGEMENTS The authors acknowledge financial support from the Department of Radiology at

AC CE P

TE

D

MA

NU

SC

RI

PT

University of Michigan Health Systems.

40

ACCEPTED MANUSCRIPT REFERENCES [1] R.L. Siegel, K.D. Miller, A. Jemal, Cancer statistics, 2015, CA Cancer J. Clin., 65 (2015) 5-29. [2] A. Carbone, E. Vaccher, A. Gloghini, L. Pantanowitz, A. Abayomi, P. de Paoli, S. Franceschi,

PT

Diagnosis and management of lymphomas and other cancers in HIV-infected patients, Nat. Rev. Clin. Oncol., 11 (2014) 223-238.

RI

[3] N.F. Walker, C. Gan, J. Olsburgh, M.S. Khan, Diagnosis and management of intradiverticular bladder tumours, Nat. Rev. Urol., 11 (2014) 383-390.

SC

[4] M.L. James, S.S. Gambhir, A molecular imaging primer: modalities, imaging agents, and applications, Physiol. Rev., 92 (2012) 897-965.

NU

[5] A. Schroeder, D.A. Heller, M.M. Winslow, J.E. Dahlman, G.W. Pratt, R. Langer, T. Jacks, D.G. Anderson, Treating metastatic cancer with nanotechnology, Nat. Rev. Cancer, 12 (2012) 39-50.

MA

[6] A.S. Thakor, S.S. Gambhir, Nanooncology: the future of cancer diagnosis and therapy, CA Cancer J. Clin., 63 (2013) 395-418.

[7] S.M. Janib, A.S. Moses, J.A. MacKay, Imaging and drug delivery using theranostic nanoparticles,

D

Adv. Drug Deliv. Rev., 62 (2010) 1052-1063.

TE

[8] H. Koo, M.S. Huh, I.-C. Sun, S.H. Yuk, K. Choi, K. Kim, I.C. Kwon, In vivo targeted delivery of nanoparticles for theranosis, Acc. Chem. Res., 44 (2011) 1018-1028.

AC CE P

[9] C. Sanchez, P. Belleville, M. Popall, L. Nicole, Applications of advanced hybrid organic–inorganic nanomaterials: from laboratory to market, Chem. Soc. Rev., 40 (2011) 696-753. [10] A.C. Grimsdale, K. Müllen, The chemistry of organic nanomaterials, Angew. Chem. Int. Ed., 44 (2005) 5592-5629.

[11] G. Garnweitner, M. Niederberger, Organic chemistry in inorganic nanomaterials synthesis, J. Mater. Chem., 18 (2008) 1171-1182.

[12] V. Lopez-Davila, A.M. Seifalian, M. Loizidou, Organic nanocarriers for cancer drug delivery, Curr. Opin. Pharmacol., 12 (2012) 414-419. [13] W. Lohcharoenkal, L. Wang, Y.C. Chen, Y. Rojanasakul, Protein nanoparticles as drug delivery carriers for cancer therapy, Biomed. Res. Int., 2014 (2014) 180549. [14] S. Shi, F. Chen, W. Cai, Biomedical applications of functionalized hollow mesoporous silica nanoparticles: focusing on molecular imaging, Nanomedicine (Lond), 8 (2013) 2027-2039. [15] R. Cao-Milan, L.M. Liz-Marzan, Gold nanoparticle conjugates: recent advances toward clinical applications, Expert Opin. Drug Deliv., 11 (2014) 741-752. [16] L. Tong, M. Zhao, S. Zhu, J. Chen, Synthesis and application of superparamagnetic iron oxide nanoparticles in targeted therapy and imaging of cancer, Front Med., 5 (2011) 379-387. 41

ACCEPTED MANUSCRIPT [17] H. Gong, R. Peng, Z. Liu, Carbon nanotubes for biomedical imaging: the recent advances, Adv. Drug Deliv. Rev., 65 (2013) 1951-1963. [18] D.J. Lim, M. Sim, L. Oh, K. Lim, H. Park, Carbon-based drug delivery carriers for cancer therapy,

PT

Arch. Pharm. Res., 37 (2014) 43-52. [19] K. Welsher, Z. Liu, S.P. Sherlock, J.T. Robinson, Z. Chen, D. Daranciang, H. Dai, A route to

RI

brightly fluorescent carbon nanotubes for near-infrared imaging in mice, Nat. Nanotechnol., 4 (2009) 773-780.

SC

[20] H. Tao, K. Yang, Z. Ma, J. Wan, Y. Zhang, Z. Kang, Z. Liu, In vivo NIR fluorescence imaging, biodistribution, and toxicology of photoluminescent carbon dots produced from carbon nanotubes and

NU

graphite, Small, 8 (2012) 281-290.

[21] A. De La Zerda, C. Zavaleta, S. Keren, S. Vaithilingam, S. Bodapati, Z. Liu, J. Levi, B.R. Smith, T.-

MA

J. Ma, O. Oralkan, Z. Cheng, X. Chen, H. Dai, B.T. Khuri-Yakub, S.S. Gambhir, Carbon nanotubes as photoacoustic molecular imaging agents in living mice, Nat. Nanotechnol., 3 (2008) 557-562. [22] J.-W. Kim, E.I. Galanzha, E.V. Shashkov, H.-M. Moon, V.P. Zharov, Golden carbon nanotubes as

D

multimodal photoacoustic and photothermal high-contrast molecular agents, Nat. Nanotechnol., 4 (2009)

TE

688-694.

[23] Z. Liu, X. Li, S.M. Tabakman, K. Jiang, S. Fan, H. Dai, Multiplexed multicolor Raman imaging of

13540-13541.

AC CE P

live cells with isotopically modified single walled carbon nanotubes, J. Am. Chem. Soc., 130 (2008)

[24] Z. Liu, C. Davis, W. Cai, L. He, X. Chen, H. Dai, Circulation and long-term fate of functionalized, biocompatible single-walled carbon nanotubes in mice probed by Raman spectroscopy, Proc. Natl. Acad. Sci. USA, 105 (2008) 1410-1415.

[25] B.S. Wong, S.L. Yoong, A. Jagusiak, T. Panczyk, H.K. Ho, W.H. Ang, G. Pastorin, Carbon nanotubes for delivery of small molecule drugs, Adv. Drug Deliv. Rev., 65 (2013) 1964-2015. [26] K. Yang, L. Feng, Z. Liu, The advancing uses of nano-graphene in drug delivery, Expert Opin. Drug Deliv., (2014) 1-12. [27] X. Yang, A. Ebrahimi, J. Li, Q. Cui, Fullerene-biomolecule conjugates and their biomedicinal applications, Int. J. Nanomedicine, 9 (2014) 77-92. [28] Y. Liu, F. Jiao, Y. Qiu, W. Li, F. Lao, G. Zhou, B. Sun, G. Xing, J. Dong, Y. Zhao, Z. Chai, C. Chen, The effect of Gd@C-82(OH)(22) nanoparticles on the release of Th1/Th2 cytokines and induction of TNF-alpha mediated cellular immunity, Biomaterials, 30 (2009) 3934-3945. [29] X. Zhu, Y. Xie, Y. Zhang, H. Huang, S. Huang, L. Hou, H. Zhang, Z. Li, J. Shi, Z. Zhang, Thermosensitive liposomes loaded with doxorubicin and lysine modified single-walled carbon nanotubes as tumor-targeting drug delivery system, J. Biomater. Appl., 29 (2014) 769-779. 42

ACCEPTED MANUSCRIPT [30] Z. Zhang, J. Wang, X. Nie, T. Wen, Y. Ji, X. Wu, Y. Zhao, C. Chen, Near infrared laser induced targeted cancer therapy using thermo-responsive polymer encapsulated gold nanorods, J. Am. Chem. Soc., 136 (2014) 7317-7326.

PT

[31] Z. Liu, X.-J. Liang, Nano-carbons as theranostics, Theranostics, 2 (2012) 235. [32] S. Zhu, G. Xu, Single-walled carbon nanohorns and their applications, Nanoscale, 2 (2010) 2538-

RI

2549.

[33] G. Pagona, G. Mountrichas, G. Rotas, N. Karousis, S. Pispas, N. Tagmatarchis, Properties,

SC

applications and functionalisation of carbon nanohorns, Int. J. Nanotechnol., 6 (2009) 176-195. [34] M. Zhang, M. Yudasaka, K. Ajima, J. Miyawaki, S. Iijima, Light-assisted oxidation of single-wall

NU

carbon nanohorns for abundant creation of oxygenated groups that enable chemical modifications with proteins to enhance biocompatibility, ACS Nano, 1 (2007) 265-272.

Nanosci. Nanotechnol., 15 (2015) 989-999.

MA

[35] L. Lai, A.S. Barnard, Functionalized nanodiamonds for biological and medical applications, J.

[36] Y. Xing, L.M. Dai, Nanodiamonds for nanomedicine, Nanomedicine, 4 (2009) 207-218.

TE

Chem. Eur. J., 14 (2008) 1382-1390.

D

[37] A. Krueger, New carbon materials: Biological applications of functionalized nanodiamond materials,

[38] P.H. Chung, E. Perevedentseva, J.S. Tu, C.C. Chang, C.L. Cheng, Spectroscopic study of biofunctionalized nanodiamonds, Diam. Relat. Mater., 15 (2006) 622-625.

AC CE P

[39] P. Anilkumar, F. Lu, L. Cao, P.G. Luo, J.H. Liu, S. Sahu, K.N. Tackett, II, Y. Wang, Y.P. Sun, Fullerenes for applications in biology and medicine, Curr. Med. Chem., 18 (2011) 2045-2059. [40] Z. Chen, L. Ma, Y. Liu, C. Chen, Applications of functionalized fullerenes in tumor theranostics, Theranostics, 2 (2012) 238-250.

[41] J.-P. Zheng, M.-M. Zhen, C.-R. Wang, C.-Y. Shu, Recent progress of molecular imaging probes based on gadofullerenes, Chinese J. Anal. Chem., 40 (2012) 1607-1614. [42] D.E. Dolmans, D. Fukumura, R.K. Jain, Photodynamic therapy for cancer, Nat. Rev. Cancer, 3 (2003) 380-387. [43] Y.-Y. Huang, S.K. Sharma, R. Yin, T. Agrawal, L.Y. Chiang, M.R. Hamblin, Functionalized fullerenes in photodynamic therapy, J. Biomed. Nanotechnol., 10 (2014) 1918-1936. [44] A. Dellinger, Z. Zhou, J. Connor, A.B. Madhankumar, S. Pamujula, C.M. Sayes, C.L. Kepley, Application of fullerenes in nanomedicine: an update, Nanomedicine, 8 (2013) 1191-1208. [45] S.A. Anderson, K.K. Lee, J.A. Frank, Gadolinium-fullerenol as a paramagnetic contrast agent for cellular imaging, Invest. Radiol., 41 (2006) 332-338.

43

ACCEPTED MANUSCRIPT [46] C.-Y. Shu, E.-Y. Zhang, J.-F. Xiang, C.-F. Zhu, C.-R. Wang, X.-L. Pei, H.-B. Han, Aggregation studies

of

the

water-soluble

gadofullerene

magnetic

resonance

imaging

contrast

agent:

Gd@C82O6(OH)16(NHCH2CH2COOH)8x, J. Phys. Chem. B, 110 (2006) 15597-15601.

PT

[47] C.-Y. Shu, X.-Y. Ma, J.-F. Zhang, F.D. Corwin, J.H. Sim, E.-Y. Zhang, H.C. Dorn, H.W. Gibson, P.P. Fatouros, C.-R. Wang, X.-H. Fang, Conjugation of a water-soluble gadolinium endohedral fulleride

RI

with an antibody as a magnetic resonance imaging contrast agent, Bioconjug. Chem., 19 (2008) 651-655. [48] P. Adiseshaiah, A. Dellinger, D. MacFarland, S. Stern, M. Dobrovolskaia, L. Ileva, A.K. Patri, M.

SC

Bernardo, D.B. Brooks, Z. Zhou, S. McNeil, C. Kepley, A novel gadolinium-based trimetasphere metallofullerene for application as a magnetic resonance imaging contrast agent, Invest. Radiol., 48

NU

(2013) 745-754.

[49] J. Zhang, Y. Ye, Y. Chen, C. Pregot, T. Li, S. Balasubramaniam, D.B. Hobart, Y. Zhang, S. Wi,

MA

R.M. Davis, L.A. Madsen, J.R. Morris, S.M. LaConte, G.T. Yee, H.C. Dorn, Gd 3N@C84(OH)x: a new egg-shaped metallofullerene magnetic resonance imaging contrast agent, J. Am. Chem. Soc., 136 (2014) 2630-2636.

D

[50] P.P. Fatouros, M.D. Shultz, Metallofullerenes: a new class of MRI agents and more?, Nanomedicine

TE

(Lond), 8 (2013) 1853-1864.

[51] K.B. Ghiassi, M.M. Olmstead, A.L. Balch, Gadolinium-containing endohedral fullerenes: structures and function as magnetic resonance imaging (MRI) agents, Dalton Trans., 43 (2014) 7346-7358.

AC CE P

[52] J. Shi, L. Wang, J. Gao, Y. Liu, J. Zhang, R. Ma, R. Liu, Z. Zhang, A fullerene-based multifunctional nanoplatform for cancer theranostic applications, Biomaterials, 35 (2014) 5771-5784. [53] D.S. Kwag, K. Park, K.T. Oh, E.S. Lee, Hyaluronated fullerenes with photoluminescent and antitumoral activity, Chem. Commun. (Camb), 49 (2013) 282-284. [54] X. Wang, C.X. Yang, J.T. Chen, X.P. Yan, A dual-targeting upconversion nanoplatform for twocolor fluorescence imaging-guided photodynamic therapy, Anal. Chem., 86 (2014) 3263-3267. [55] J. Shi, Z. Wang, L. Wang, H. Wang, L. Li, X. Yu, J. Zhang, R. Ma, Z. Zhang, Photodynamic therapy of a 2-methoxyestradiol tumor-targeting drug delivery system mediated by Asn-Gly-Arg in breast cancer, Int. J. Nanomedicine, 8 (2013) 1551-1562. [56] T.Y. Zakharian, A. Seryshev, B. Sitharaman, B.E. Gilbert, V. Knight, L.J. Wilson, A fullerenepaclitaxel chemotherapeutic: Synthesis, characterization, and study of biological activity in tissue culture, J. Am. Chem. Soc., 127 (2005) 12508-12509. [57] J. Shi, Y. Liu, L. Wang, J. Gao, J. Zhang, X. Yu, R. Ma, R. Liu, Z. Zhang, A tumoral acidic pHresponsive drug delivery system based on a novel photosensitizer (fullerene) for in vitro and in vivo chemo-photodynamic therapy, Acta Biomater., 10 (2014) 1280-1291.

44

ACCEPTED MANUSCRIPT [58] H. Isobe, W. Nakanishi, N. Tomita, S. Jinno, H. Okayama, E. Nakamura, Gene delivery by aminofullerenes: Structural requirements for efficient transfection, Chem. Asian J., 1 (2006) 167-175. [59] R. Maeda-Mamiya, E. Noiri, H. Isobe, W. Nakanishi, K. Okamoto, K. Doi, T. Sugaya, T. Izumi, T.

PT

Homma, E. Nakamura, In vivo gene delivery by cationic tetraamino fullerene, Proc. Natl. Acad. Sci. USA, 107 (2010) 5339-5344.

RI

[60] I. Blazkova, N. Hoai Viet, M. Kominkova, R. Konecna, D. Chudobova, L. Krejcova, P. Kopel, D. Hynek, O. Zitka, M. Beklova, V. Adam, R. Kizek, Fullerene as a transporter for doxorubicin investigated

SC

by analytical methods and in vivo imaging, Electrophoresis, 35 (2014) 1040-1049. [61] J. Fan, G. Fang, F. Zeng, X. Wang, S. Wu, Water-dispersible fullerene aggregates as a targeted

NU

anticancer prodrug with both chemo- and photodynamic therapeutic actions, Small, 9 (2013) 613-621. [62] X. Guo, R. Ding, Y. Zhang, L. Ye, X. Liu, C. Chen, Z. Zhang, Y. Zhang, Dual role of

MA

photosensitizer and carrier material of fullerene in micelles for chemo-photodynamic therapy of cancer, J. Pharm. Sci., 103 (2014) 3225-3234.

[63] J. Shi, X. Yu, L. Wang, Y. Liu, J. Gao, J. Zhang, R. Ma, R. Liu, Z. Zhang, PEGylated fullerene/iron

D

oxide nanocomposites for photodynamic therapy, targeted drug delivery and MR imaging, Biomaterials,

TE

34 (2013) 9666-9677.

[64] Z. Liu, K. Yang, S.-T. Lee, Single-walled carbon nanotubes in biomedical imaging, J. Mater. Chem., 21 (2011) 586-598.

AC CE P

[65] H. Hong, T. Gao, W. Cai, Molecular imaging with single-walled carbon nanotubes, Nano Today, 4 (2009) 252-261.

[66] J.T. Robinson, K. Welsher, S.M. Tabakman, S.P. Sherlock, H. Wang, R. Luong, H. Dai, High performance in vivo near-IR (>1 mum) imaging and photothermal cancer therapy with carbon nanotubes, Nano Res., 3 (2010) 779-793.

[67] J.T. Robinson, G. Hong, Y. Liang, B. Zhang, O.K. Yaghi, H. Dai, In vivo fluorescence imaging in the second near-infrared window with long circulating carbon nanotubes capable of ultrahigh tumor uptake, J. Am. Chem. Soc., 134 (2012) 10664-10669. [68] K. Welsher, S.P. Sherlock, H. Dai, Deep-tissue anatomical imaging of mice using carbon nanotube fluorophores in the second near-infrared window, Proc. Natl. Acad. Sci. USA, 108 (2011) 8943-8948. [69] J. Lefebvre, D.G. Austing, J. Bond, P. Finnie, Photoluminescence imaging of suspended singlewalled carbon nanotubes, Nano Lett., 6 (2006) 1603-1608. [70] H. Yi, D. Ghosh, M.-H. Ham, J. Qi, P.W. Barone, M.S. Strano, A.M. Belcher, M13 Phagefunctionalized single-walled carbon nanotubes as nanoprobes for second near-infrared window fluorescence imaging of targeted tumors, Nano Lett., 12 (2012) 1176-1183.

45

ACCEPTED MANUSCRIPT [71] D. Ghosh, A.F. Bagley, Y.J. Na, M.J. Birrer, S.N. Bhatia, A.M. Belcher, Deep, noninvasive imaging and surgical guidance of submillimeter tumors using targeted M13-stabilized single-walled carbon nanotubes, Proc. Natl. Acad. Sci. USA, 111 (2014) 13948-13953.

PT

[72] M.L. Chen, Y.J. He, X.W. Chen, J.H. Wang, Quantum dots conjugated with Fe 3O4-filled carbon nanotubes for cancer-targeted imaging and magnetically guided drug delivery, Langmuir, 28 (2012)

RI

16469-16476.

[73] K.S. Yang, J.S. Yun, J.C. Kim, J. Min, T.J. Park, J.K. Ahn, H. Kim do, Polydiacetylene single-

SC

walled carbon nanotubes nano-hybrid for cellular imaging applications, J. Nanosci. Nanotechnol., 12 (2012) 377-385.

NU

[74] R. El-Sayed, M. Eita, A. Barrefelt, F. Ye, H. Jain, M. Fares, A. Lundin, M. Crona, K. Abu-Salah, M. Muhammed, M. Hassan, Thermostable luciferase from Luciola cruciate for imaging of carbon nanotubes

MA

and carbon nanotubes carrying doxorubicin using in vivo imaging system, Nano Lett., 13 (2013) 13931398.

[75] L.G. Delogu, G. Vidili, E. Venturelli, C. Menard-Moyon, M.A. Zoroddu, G. Pilo, P. Nicolussi, C.

D

Ligios, D. Bedognetti, F. Sgarrella, R. Manetti, A. Bianco, Functionalized multiwalled carbon nanotubes

TE

as ultrasound contrast agents, Proc. Natl. Acad. Sci. USA, 109 (2012) 16612-16617. [76] H. Wu, H. Shi, H. Zhang, X. Wang, Y. Yang, C. Yu, C. Hao, J. Du, H. Hu, S. Yang, Prostate stem cell antigen antibody-conjugated multiwalled carbon nanotubes for targeted ultrasound imaging and drug

AC CE P

delivery, Biomaterials, 35 (2014) 5369-5380. [77] L. Xiang, Y. Yuan, D. Xing, Z. Ou, S. Yang, F. Zhou, Photoacoustic molecular imaging with antibody-functionalized single-walled carbon nanotubes for early diagnosis of tumor, J. Biomed. Opt., 14 (2009) 021008.

[78] A. de la Zerda, Z. Liu, S. Bodapati, R. Teed, S. Vaithilingam, B.T. Khuri-Yakub, X. Chen, H. Dai, S.S. Gambhir, Ultrahigh sensitivity carbon nanotube agents for photoacoustic molecular imaging in living mice, Nano Lett., 10 (2010) 2168-2172. [79] Y. Zhang, H. Hong, W. Cai, Photoacoustic imaging, Cold Spring Harb. Protoc., 9 (2011) 1015-1025. [80] J. Koo, M. Jeon, Y. Oh, H.W. Kang, J. Kim, C. Kim, J. Oh, In vivo non-ionizing photoacoustic mapping of sentinel lymph nodes and bladders with ICG-enhanced carbon nanotubes, Phys. Med. Biol., 57 (2012) 7853-7862. [81] C. Wang, C. Bao, S. Liang, H. Fu, K. Wang, M. Deng, Q. Liao, D. Cui, RGD-conjugated silicacoated gold nanorods on the surface of carbon nanotubes for targeted photoacoustic imaging of gastric cancer, Nanoscale Res. Lett., 9 (2014) 264.

46

ACCEPTED MANUSCRIPT [82] Z. Liu, S. Tabakman, S. Sherlock, X. Li, Z. Chen, K. Jiang, S. Fan, H. Dai, Multiplexed Five-Color Molecular imaging of cancer cells and tumor tissues with carbon nanotube Raman tags in the nearinfrared, Nano Res., 3 (2010) 222-233.

PT

[83] C. Lamprecht, N. Gierlinger, E. Heister, B. Unterauer, B. Plochberger, M. Brameshuber, P. Hinterdorfer, S. Hild, A. Ebner, Mapping the intracellular distribution of carbon nanotubes after targeted

RI

delivery to carcinoma cells using confocal Raman imaging as a label-free technique, J. Phys. Condens. Matter., 24 (2012) 164206.

SC

[84] X. Wang, C. Wang, L. Cheng, S.T. Lee, Z. Liu, Noble metal coated single-walled carbon nanotubes for applications in surface enhanced Raman scattering imaging and photothermal therapy, J. Am. Chem.

NU

Soc., 134 (2012) 7414-7422.

[85] A. Ruggiero, C.H. Villa, J.P. Holland, S.R. Sprinkle, C. May, J.S. Lewis, D.A. Scheinberg, M.R.

MA

McDevitt, Imaging and treating tumor vasculature with targeted radiolabeled carbon nanotubes, Int. J. Nanomedicine, 5 (2010) 783-802.

[86] Y. Liu, T.C. Hughes, B.W. Muir, L.J. Waddington, T.R. Gengenbach, C.D. Easton, T.M. Hinton,

D

B.A. Moffat, X. Hao, J. Qiu, Water-dispersible magnetic carbon nanotubes as T2-weighted MRI contrast

TE

agents, Biomaterials, 35 (2014) 378-386.

[87] A. Al Faraj, A.P. Shaik, A.S. Shaik, Magnetic single-walled carbon nanotubes as efficient drug delivery nanocarriers in breast cancer murine model: noninvasive monitoring using diffusion-weighted

AC CE P

magnetic resonance imaging as sensitive imaging biomarker, Int. J. Nanomedicine, 10 (2015) 157-168. [88] S. Shen, F. Kong, X. Guo, L. Wu, H. Shen, M. Xie, X. Wang, Y. Jin, Y. Ge, CMCTS stabilized Fe3O4 particles with extremely low toxicity as highly efficient near-infrared photothermal agents for in vivo tumor ablation, Nanoscale, 5 (2013) 8056-8066. [89] M. Das, S.R. Datir, R.P. Singh, S. Jain, Augmented anticancer activity of a targeted, intracellularly activatable, theranostic nanomedicine based on fluorescent and radiolabeled, methotrexate-folic Acidmultiwalled carbon nanotube conjugate, Mol. Pharm., 10 (2013) 2543-2557. [90] C. Liang, S. Diao, C. Wang, H. Gong, T. Liu, G. Hong, X. Shi, H. Dai, Z. Liu, Tumor metastasis inhibition by imaging-guided photothermal therapy with single-walled carbon nanotubes, Adv. Mater., 26 (2014) 5646-5652. [91] B.T. Cisneros, J.J. Law, M.L. Matson, A. Azhdarinia, E.M. Sevick-Muraca, L.J. Wilson, Stable confinement of positron emission tomography and magnetic resonance agents within carbon nanotubes for bimodal imaging, Nanomedicine (Lond), 9 (2014) 2499-2509. [92] S.K. Vashist, D. Zheng, G. Pastorin, K. Al-Rubeaan, J.H.T. Luong, F.-S. Sheu, Delivery of drugs and biomolecules using carbon nanotubes, Carbon, 49 (2011) 4077-4097.

47

ACCEPTED MANUSCRIPT [93] L. Meng, X. Zhang, Q. Lu, Z. Fei, P.J. Dyson, Single walled carbon nanotubes as drug delivery vehicles: Targeting doxorubicin to tumors, Biomaterials, 33 (2012) 1689-1698. [94] J.J. Mulvey, C.H. Villa, M.R. McDevitt, F.E. Escorcia, E. Casey, D.A. Scheinberg, Self-assembly of

PT

carbon nanotubes and antibodies on tumours for targeted amplified delivery, Nat. Nanotechnol., 8 (2013) 763-771.

RI

[95] J. Ren, S. Shen, D. Wang, Z. Xi, L. Guo, Z. Pang, Y. Qian, X. Sun, X. Jiang, The targeted delivery of anticancer drugs to brain glioma by PEGylated oxidized multi-walled carbon nanotubes modified with

SC

angiopep-2, Biomaterials, 33 (2012) 3324-3333.

[96] Z. Ji, G. Lin, Q. Lu, L. Meng, X. Shen, L. Dong, C. Fu, X. Zhang, Targeted therapy of SMMC-7721

NU

liver cancer in vitro and in vivo with carbon nanotubes based drug delivery system, J. Colloid Interface Sci., 365 (2012) 143-149.

MA

[97] A.A. Bhirde, V. Patel, J. Gavard, G. Zhang, A.A. Sousa, A. Masedunskas, R.D. Leapman, R. Weigert, J.S. Gutkind, J.F. Rusling, Targeted killing of cancer cells in vivo and in vitro with EGF-directed carbon nanotube-based drug delivery, ACS Nano, 3 (2009) 307-316.

D

[98] W. Cheung, F. Pontoriero, O. Taratula, A.M. Chen, H. He, DNA and carbon nanotubes as medicine,

TE

Adv. Drug Deliv. Rev., 62 (2010) 633-649.

[99] M. Karimi, N. Solati, A. Ghasemi, M.A. Estiar, M. Hashemkhani, P. Kiani, E. Mohamed, A. Saeidi, M. Taheri, P. Avci, A.R. Aref, M. Amiri, F. Baniasadi, M.R. Hamblin, Carbon nanotubes part II: a

AC CE P

remarkable carrier for drug and gene delivery, Expert. Opin. Drug Deliv., 2015 (Epub). [100] K.S. Siu, D. Chen, X. Zheng, X. Zhang, N. Johnston, Y. Liu, K. Yuan, J. Koropatnick, E.R. Gillies, W.P. Min, Non-covalently functionalized single-walled carbon nanotube for topical siRNA delivery into melanoma, Biomaterials, 35 (2014) 3435-3442. [101] R. Singh, S.V. Torti, Carbon nanotubes in hyperthermia therapy, Adv, Drug Deliv, Rev., 65 (2013) 2045-2060.

[102] J. Shi, R. Ma, L. Wang, J. Zhang, R. Liu, L. Li, Y. Liu, L. Hou, X. Yu, J. Gao, Z. Zhang, The application of hyaluronic acid-derivatized carbon nanotubes in hematoporphyrin monomethyl ether-based photodynamic therapy for in vivo and in vitro cancer treatment, Int. J. Nanomedicine, 8 (2013) 23612373. [103] L. Wang, J. Shi, R. Liu, Y. Liu, J. Zhang, X. Yu, J. Gao, C. Zhang, Z. Zhang, Photodynamic effect of functionalized single-walled carbon nanotubes: a potential sensitizer for photodynamic therapy, Nanoscale, 6 (2014) 4642-4651. [104] L. Wang, J. Shi, H. Zhang, H. Li, Y. Gao, Z. Wang, H. Wang, L. Li, C. Zhang, C. Chen, Z. Zhang, Y. Zhang, Synergistic anticancer effect of RNAi and photothermal therapy mediated by functionalized single-walled carbon nanotubes, Biomaterials, 34 (2013) 262-274. 48

ACCEPTED MANUSCRIPT [105] H. Kataura, Y. Kumazawa, Y. Maniwa, I. Umezu, S. Suzuki, Y. Ohtsuka, Y. Achiba, Optical properties of single-wall carbon nanotubes, Synth. Metals, 103 (1999) 2555-2558. [106] K. Murata, K. Kaneko, F. Kokai, K. Takahashi, M. Yudasaka, S. Iijima, Pore structure of single-

PT

wall carbon nanohorn aggregates, Chem. Phys. Lett., 331 (2000) 14-20. [107] K. Ajima, M. Yudasaka, T. Murakami, A. Maigne, K. Shiba, S. Iijima, Carbon nanohorns as

RI

anticancer drug carriers, Mol. Pharm., 2 (2005) 475-480.

Advances, 4 (2014) 27315-27321.

SC

[108] J. Guerra, M.A. Herrero, E. Vazquez, Carbon nanohorns as alternative gene delivery vectors, RSC

[109] M. Zhang, T. Yamaguchi, S. Iijima, M. Yudasaka, Size-dependent biodistribution of carbon

NU

nanohorns in vivo, Nanomedicine, 9 (2013) 657-664.

[110] T. Azami, D. Kasuya, R. Yuge, M. Yudasaka, S. Iijima, T. Yoshitake, Y. Kubo, Large-scale

MA

production of single-wall carbon nanohorns with high purity, J. Phys. Chem. C, 112 (2008) 1330-1334. [111] J. Miyawaki, M. Yudasaka, T. Azami, Y. Kubo, S. Iijima, Toxicity of single-walled carbon nanohorns, ACS Nano, 2 (2008) 213-226.

D

[112] F. Zhou, C. Li, W. Zhong, Y. Hu, W. Shen, Applications of single-walled carbon nanohorns in

TE

biomedicine, Chemistry, 75 (2012) 1095-1099.

[113] A. Hashimoto, H. Yorimitsu, K. Ajima, K. Suenaga, H. Isobe, A. Miyawaki, M. Yudasaka, S. Iijima, E. Nakamura, Selective deposition of a gadolinium(III) cluster in a hole opening of single-wall

AC CE P

carbon nanohorn, Proc. Natl. Acad. Sci. USA, 101 (2004) 8527-8530. [114] J. Miyawaki, S. Matsumura, R. Yuge, T. Murakami, S. Sato, A. Tomida, T. Tsuruo, T. Ichihashi, T. Fujinami, H. Irie, K. Tsuchida, S. Iijima, K. Shiba, M. Yudasaka, Biodistribution and ultrastructural localization of single-walled carbon nanohorns determined in vivo with embedded Gd2O3 labels, ACS Nano, 3 (2009) 1399-1406.

[115] J. Miyawaki, M. Yudasaka, H. Imai, H. Yorimitsu, H. Isobe, E. Nakamura, S. Iijima, Synthesis of ultrafine Gd2O3 nanoparticles inside single-wall carbon nanohorns, J. Phys. Chem. B, 110 (2006) 51795181. [116] J. Zhang, J. Ge, M.D. Shultz, E. Chung, G. Singh, C. Shu, P.P. Fatouros, S.C. Henderson, F.D. Corwin, D.B. Geohegan, In vitro and in vivo studies of single-walled carbon nanohorns with encapsulated metallofullerenes and exohedrally functionalized quantum dots, Nano Lett., 10 (2010) 2843-2848. [117] J. Miyawaki, M. Yudasaka, H. Imai, H. Yorimitsu, H. Isobe, E. Nakamura, S. Iijima, In vivo magnetic resonance imaging of single-walled carbon nanohorns by labeling with magnetite nanoparticles, Adv. Mater., 18 (2006) 1010-1014.

49

ACCEPTED MANUSCRIPT [118] D. Chen, C. Wang, X. Nie, S. Li, R. Li, M. Guan, Z. Liu, C. Chen, C. Wang, C. Shu, L. Wan, Photoacoustic imaging guided near-infrared photothermal therapy using highly water-dispersible singlewalled carbon nanohorns as theranostic agents, Adv. Funct. Mater., 24 (2014) 6621-6628.

PT

[119] D. Chen, C. Wang, F. Jiang, Z. Liu, C. Shu, L.-J. Wan, In vitro and in vivo photothermally enhanced chemotherapy by single-walled carbon nanohorns as a drug delivery system, J. Mater. Chem. B,

RI

2 (2014) 4726-4732.

[120] K. Ajima, T. Murakami, Y. Mizoguchi, K. Tsuchida, T. Ichihashi, S. Iijima, M. Yudasaka,

SC

Enhancement of in vivo anticancer effects of cisplatin by incorporation inside single-wall carbon nanohorns, ACS Nano, 2 (2008) 2057-2064.

NU

[121] M. Zhang, T. Murakami, K. Ajima, K. Tsuchida, A.S. Sandanayaka, O. Ito, S. Iijima, M. Yudasaka, Fabrication of ZnPc/protein nanohorns for double photodynamic and hyperthermic cancer phototherapy,

MA

Proc. Natl. Acad. Sci. USA, 105 (2008) 14773-14778.

[122] J. Guerra, M. Antonia Herrero, B. Carrion, F.C. Perez-Martinez, M. Lucio, N. Rubio, M. Meneghetti, M. Prato, V. Cena, E. Vazquez, Carbon nanohorns functionalized with polyamidoamine

D

dendrimers as efficient biocarrier materials for gene therapy, Carbon, 50 (2012) 2832-2844.

TE

[123] E. Miyako, T. Deguchi, Y. Nakajima, M. Yudasaka, Y. Hagihara, M. Horie, M. Shichiri, Y. Higuchi, F. Yamashita, M. Hashida, Y. Shigeri, Y. Yoshida, S. Iijima, Photothermic regulation of gene

7528.

AC CE P

expression triggered by laser-induced carbon nanohorns, Proc. Natl. Acad. Sci. USA, 109 (2012) 7523-

[124] R. Kaur, I. Badea, Nanodiamonds as novel nanomaterials for biomedical applications: drug delivery and imaging systems, Int. J. Nanomedicine, 8 (2013) 203-220. [125] C. Bradac, T. Gaebel, N. Naidoo, M.J. Sellars, J. Twamley, L.J. Brown, A.S. Barnard, T. Plakhotnik, A.V. Zvyagin, J.R. Rabeau, Observation and control of blinking nitrogen-vacancy centres in discrete nanodiamonds, Nat. Nanotechnol., 5 (2010) 345-349. [126] C.C. Fu, H.Y. Lee, K. Chen, T.S. Lim, H.Y. Wu, P.K. Lin, P.K. Wei, P.H. Tsao, H.C. Chang, W. Fann, Characterization and application of single fluorescent nanodiamonds as cellular biomarkers, Proc. Natl. Acad. Sci. USA, 104 (2007) 727-732. [127] R. Igarashi, Y. Yoshinari, H. Yokota, T. Sugi, F. Sugihara, K. Ikeda, H. Sumiya, S. Tsuji, I. Mori, H. Tochio, Y. Harada, M. Shirakawa, Real-time background-free selective imaging of fluorescent nanodiamonds in vivo, Nano Lett., 12 (2012) 5726-5732. [128] Y.R. Chang, H.Y. Lee, K. Chen, C.C. Chang, D.S. Tsai, C.C. Fu, T.S. Lim, Y.K. Tzeng, C.Y. Fang, C.C. Han, H.C. Chang, W. Fann, Mass production and dynamic imaging of fluorescent nanodiamonds, Nat. Nanotechnol., 3 (2008) 284-288.

50

ACCEPTED MANUSCRIPT [129] Y.Y. Hui, L.J. Su, O.Y. Chen, Y.T. Chen, T.M. Liu, H.C. Chang, Wide-field imaging and flow cytometric analysis of cancer cells in blood by fluorescent nanodiamond labeling and time gating, Sci. Rep., 4 (2014) 5574.

PT

[130] F.Q. Hu, H.M. Joshi, V.P. Dravid, T.J. Meade, High-performance nanostructured MR contrast probes, Nanoscale, 2 (2010) 1884-1891.

RI

[131] T. Nakamura, T. Ohana, H. Yabuno, R. Kasai, T. Suzuki, T. Hasebe, Simple fabrication of Gd(III)-

contrast agent, Appl. Phys. Express, 6 (2013) 015001.

SC

DTPA-nanodiamond particles by chemical modification for use as magnetic resonance imaging (MRI)

[132] L.M. Manus, D.J. Mastarone, E.A. Waters, X.Q. Zhang, E.A. Schultz-Sikma, K.W. MacRenaris, D.

NU

Ho, T.J. Meade, Gd(III)-nanodiamond conjugates for MRI contrast enhancement, Nano Lett., 10 (2010) 484-489.

MA

[133] S. Rojas, J.D. Gispert, R. Martin, S. Abad, C. Menchon, D. Pareto, V.M. Victor, M. Alvaro, H. Garcia, J.R. Herance, Biodistribution of amino-functionalized diamond nanoparticles. In vivo studies based on 18F radionuclide emission, ACS Nano, 5 (2011) 5552-5559.

D

[134] Z.Y. Lien, T.C. Hsu, K.K. Liu, W.S. Liao, K.C. Hwang, J.I. Chao, Cancer cell labeling and tracking

TE

using fluorescent and magnetic nanodiamond, Biomaterials, 33 (2012) 6172-6185. [135] A.N. Hegyi, E. Yablonovitch, Nanodiamond imaging: a new molecular imaging approach, Conf. Proc. IEEE Eng. Med. Biol. Soc., 2012 (2012) 2639-2642.

AC CE P

[136] R. Lam, M. Chen, E. Pierstorff, H. Huang, E.J. Osawa, D. Ho, Nanodiamond-embedded microfilm devices for localized chemotherapeutic elution, ACS Nano, 2 (2008) 2095-2102. [137] E.K. Chow, X.Q. Zhang, M. Chen, R. Lam, E. Robinson, H.J. Huang, D. Schaffer, E. Osawa, A. Goga, D. Ho, Nanodiamond therapeutic delivery agents mediate enhanced chemoresistant tumor treatment, Sci. Transl. Med., 3 (2011) 73ra21. [138] Y.Q. Li, Y.L. Tong, R.X. Cao, Z.M. Tian, B.S. Yang, P. Yang, In vivo enhancement of anticancer therapy using bare or chemotherapeutic drug-bearing nanodiamond particles, Int. J. Nanomedicine, 9 (2014) 1065-1082. [139] J.S. Xiao, X.P. Duan, Q. Yin, Z.W. Zhang, H.J. Yu, Y.P. Li, Nanodiamonds-mediated doxorubicin nuclear delivery to inhibit lung metastasis of breast cancer, Biomaterials, 34 (2013) 9648-9656. [140] J. Li, Y. Zhu, W.X. Li, X.Y. Zhang, Y. Peng, Q. Huang, Nanodiamonds as intracellular transporters of chemotherapeutic drug, Biomaterials, 31 (2010) 8410-8418. [141] H.B. Man, H. Kim, H.J. Kim, E. Robinson, W.K. Liu, E.K. Chow, D. Ho, Synthesis of nanodiamond-daunorubicin

conjugates

Nanomedicine, 10 (2014) 359-369.

51

to

overcome

multidrug

chemoresistance

in

leukemia,

ACCEPTED MANUSCRIPT [142] R.A. Shimkunas, E. Robinson, R. Lam, S. Lu, X. Xu, X.Q. Zhang, H. Huang, E. Osawa, D. Ho, Nanodiamond-insulin complexes as pH-dependent protein delivery vehicles, Biomaterials, 30 (2009) 5720-5728.

PT

[143] A. Alhaddad, M.P. Adam, J. Botsoa, G. Dantelle, S. Perruchas, T. Gacoin, C. Mansuy, S. Lavielle, C. Malvy, F. Treussart, J.R. Bertrand, Nanodiamond as a vector for siRNA delivery to Ewing sarcoma

RI

cells, Small, 7 (2011) 3087-3095.

[144] A. Alhaddad, C. Durieu, G. Dantelle, E. Le Cam, C. Malvy, F. Treussart, J.R. Bertrand, Influence

SC

of the internalization pathway on the efficacy of siRNA delivery by cationic fluorescent nanodiamonds in the Ewing sarcoma cell model, PLoS One, 7 (2012) e52207.

NU

[145] M. Cao, X. Deng, S. Su, F. Zhang, X. Xiao, Q. Hu, Y. Fu, B.B. Yang, Y. Wu, W. Sheng, Y. Zeng, Protamine sulfate-nanodiamond hybrid nanoparticles as a vector for MiR-203 restoration in esophageal

MA

carcinoma cells, Nanoscale, 5 (2013) 12120-12125.

[146] H. Kim, H.B. Man, B. Saha, A.M. Kopacz, O.S. Lee, G.C. Schatz, D. Ho, W.K. Liu, Multiscale Simulation as a Framework for the Enhanced Design of Nanodiamond-Polyethylenimine-based Gene

D

Delivery, J. Phys. Chem. Lett., 3 (2012) 3791-3797.

TE

[147] L.C. Cheng, H.M. Chen, T.C. Lai, Y.C. Chan, R.S. Liu, J.C. Sung, M. Hsiao, C.H. Chen, L.J. Her, D.P. Tsai, Targeting polymeric fluorescent nanodiamond-gold/silver multi-functional nanoparticles as a light-transforming hyperthermia reagent for cancer cells, Nanoscale, 5 (2013) 3931-3940.

AC CE P

[148] Y.P. Sun, B. Zhou, Y. Lin, W. Wang, K.A.S. Fernando, P. Pathak, M.J. Meziani, B.A. Harruff, X. Wang, H.F. Wang, P.J.G. Luo, H. Yang, M.E. Kose, B.L. Chen, L.M. Veca, S.Y. Xie, Quantum-sized carbon dots for bright and colorful photoluminescence, J. Am. Chem. Soc., 128 (2006) 7756-7757. [149] L. Cao, X. Wang, M.J. Meziani, F. Lu, H. Wang, P.G. Luo, Y. Lin, B.A. Harruff, L.M. Veca, D. Murray, S.-Y. Xie, Y.-P. Sun, Carbon dots for multiphoton bioimaging, J. Am. Chem. Soc., 129 (2007) 11318-11319.

[150] Z.-A. Qiao, Y. Wang, Y. Gao, H. Li, T. Dai, Y. Liu, Q. Huo, Commercially activated carbon as the source for producing multicolor photoluminescent carbon dots by chemical oxidation, Chem. Commum., 46 (2010) 8812-8814. [151] L. Cao, S.-T. Yang, X. Wang, P.G. Luo, J.-H. Liu, S. Sahu, Y. Liu, Y.-P. Sun, Competitive Performance of Carbon "Quantum" Dots in Optical Bioimaging, Theranostics, 2 (2012) 295-301. [152] E.J. Goh, K.S. Kim, Y.R. Kim, H.S. Jung, S. Beack, W.H. Kong, G. Scarcelli, S.H. Yun, S.K. Hahn, Bioimaging of hyaluronic acid derivatives using nanosized carbon dots, Biomacromolecules, 13 (2012) 2554-2561.

52

ACCEPTED MANUSCRIPT [153] S. Sahu, B. Behera, T.K. Maiti, S. Mohapatra, Simple one-step synthesis of highly luminescent carbon dots from orange juice: application as excellent bio-imaging agents, Chem. Commum., 48 (2012) 8835-8837.

watermelon peel as a carbon source, Mater. Lett., 66 (2012) 222-224.

PT

[154] J. Zhou, Z. Sheng, H. Han, M. Zou, C. Li, Facile synthesis of fluorescent carbon dots using

RI

[155] L. Wang, H.S. Zhou, Green Synthesis of luminescent nitrogen-doped carbon dots from milk and its imaging application, Anal. Chem., 86 (2014) 8902-8905.

SC

[156] S.-T. Yang, L. Cao, P.G. Luo, F. Lu, X. Wang, H. Wang, M.J. Meziani, Y. Liu, G. Qi, Y.-P. Sun, Carbon dots for optical imaging in vivo, J. Am. Chem. Soc., 131 (2009) 11308-11309.

NU

[157] H. Tao, K. Yang, Z. Ma, J. Wan, Y. Zhang, Z. Kang, Z. Liu, In vivo NIR fluorescence imaging, biodistribution, and toxicology of photoluminescent carbon dots produced from carbon nanotubes and

MA

graphite, Small, 8 (2012) 281-290.

[158] Q. Liu, S. Xu, C. Niu, M. Li, D. He, Z. Lu, L. Ma, N. Na, F. Huang, H. Jiang, J. Ouyang, Distinguish cancer cells based on targeting turn-on fluorescence imaging by folate functionalized green

D

emitting carbon dots, Biosens. Bioelectron., 64 (2015) 119-125.

TE

[159] L. Zhou, Z. Li, Z. Liu, J. Ren, X. Qu, Luminescent carbon dot-gated nanovehicles for pH-triggered intracellular controlled release and imaging, Langmuir, 29 (2013) 6396-6403. [160] Y. Xu, X.H. Jia, X.B. Yin, X.W. He, Y.K. Zhang, Carbon quantum dot stabilized gadolinium

AC CE P

nanoprobe prepared via a one-pot hydrothermal approach for magnetic resonance and fluorescence dualmodality bioimaging, Anal. Chem., 86 (2014) 12122-12129. [161] X. Huang, F. Zhang, L. Zhu, K.Y. Choi, N. Guo, J. Guo, K. Tackett, P. Anilkumar, G. Liu, Q. Quan, H.S. Choi, G. Niu, Y.-P. Sun, S. Lee, X. Chen, Effect of injection routes on the biodistribution, clearance, and tumor uptake of carbon dots, ACS Nano, 7 (2013) 5684-5693. [162] M. Zheng, S. Liu, J. Li, D. Qu, H. Zhao, X. Guan, X. Hu, Z. Xie, X. Jing, Z. Sun, Integrating oxaliplatin with highly luminescent carbon dots: an unprecedented theranostic agent for personalized medicine, Adv. Mater., 26 (2014) 3554-3560. [163] S. Pandey, M. Thakur, A. Mewada, D. Anjarlekar, N. Mishra, M. Sharon, Carbon dots functionalized gold nanorod mediated delivery of doxorubicin: tri-functional nano-worms for drug delivery, photothermal therapy and bioimaging, J. Mater. Chem. B, 1 (2013) 4972-4982. [164] Q. Wang, C. Zhang, G. Shen, H. Liu, H. Fu, D. Cui, Fluorescent carbon dots as an efficient siRNA nanocarrier for its interference therapy in gastric cancer cells, J. Nanobiotechnology, 12 (2014) 288. [165] H. Liu, Q. Wang, G. Shen, C. Zhang, C. Li, W. Ji, C. Wang, D. Cui, A multifunctional ribonuclease A-conjugated carbon dot cluster nanosystem for synchronous cancer imaging and therapy, Nanoscale Res. Lett., 9 (2014) 397. 53

ACCEPTED MANUSCRIPT [166] P. Huang, J. Lin, X. Wang, Z. Wang, C. Zhang, M. He, K. Wang, F. Chen, Z. Li, G. Shen, D. Cui, X. Chen, Light-triggered theranostics based on photosensitizer-conjugated carbon dots for simultaneous enhanced-fluorescence imaging and photodynamic therapy, Adv. Mater., 24 (2012) 5104-5110.

PT

[167] C. Fowley, N. Nomikou, A.P. McHale, B. McCaughan, J.F. Callan, Extending the tissue penetration capability of conventional photosensitisers: a carbon quantum dot-protoporphyrin IX

RI

conjugate for use in two-photon excited photodynamic therapy, Chem. Commun. (Camb), 49 (2013) 8934-8936.

SC

[168] J. Wang, Z. Zhang, S. Zha, Y. Zhu, P. Wu, B. Ehrenberg, J.Y. Chen, Carbon nanodots featuring efficient FRET for two-photon photodynamic cancer therapy with a low fs laser power density,

NU

Biomaterials, 35 (2014) 9372-9381.

[169] Y. Choi, S. Kim, M.-H. Choi, S.-R. Ryoo, J. Park, D.-H. Min, B.-S. Kim, Highly biocompatible

Adv. Funct. Mater., 24 (2014) 5781-5789.

MA

carbon nanodots for simultaneous bioimaging and targeted photodynamic therapy in vitro and in vivo,

[170] L. Feng, Z. Liu, Graphene in biomedicine: opportunities and challenges, Nanomedicine (Lond), 6

D

(2011) 317-324.

Soc. Rev., 42 (2013) 530-547.

TE

[171] K. Yang, L. Feng, X. Shi, Z. Liu, Nano-graphene in biomedicine: theranostic applications, Chem.

[172] L. Feng, L. Wu, X. Qu, New horizons for diagnostics and therapeutic applications of graphene and

AC CE P

graphene oxide, Adv. Mater., 25 (2013) 168-186. [173] A.K. Geim, K.S. Novoselov, The rise of graphene, Nat. Mater., 6 (2007) 183-191. [174] H. Hong, K. Yang, Y. Zhang, J.W. Engle, L. Feng, Y. Yang, T.R. Nayak, S. Goel, J. Bean, C.P. Theuer, T.E. Barnhart, Z. Liu, W. Cai, In vivo targeting and imaging of tumor vasculature with radiolabeled, antibody-conjugated nanographene, ACS Nano, 6 (2012) 2361-2370. [175] H. Hong, Y. Zhang, J.W. Engle, T.R. Nayak, C.P. Theuer, R.J. Nickles, T.E. Barnhart, W. Cai, In vivo targeting and positron emission tomography imaging of tumor vasculature with 66Ga-labeled nanographene, Biomaterials, 33 (2012) 4147-4156. [176] Y. Zhang, T.R. Nayak, H. Hong, W. Cai, Graphene: a versatile nanoplatform for biomedical applications, Nanoscale, 4 (2012) 3833-3842. [177] S. Shi, K. Yang, H. Hong, F. Chen, H.F. Valdovinos, S. Goel, T.E. Barnhart, Z. Liu, W. Cai, VEGFR targeting leads to significantly enhanced tumor uptake of nanographene oxide in vivo, Biomaterials, 39 (2015) 39-46. [178] S. Shi, K. Yang, H. Hong, H.F. Valdovinos, T.R. Nayak, Y. Zhang, C.P. Theuer, T.E. Barnhart, Z. Liu, W. Cai, Tumor vasculature targeting and imaging in living mice with reduced graphene oxide, Biomaterials, 34 (2013) 3002-3009. 54

ACCEPTED MANUSCRIPT [179] S. Shi, F. Chen, E.B. Ehlerding, W. Cai, Surface engineering of graphene-based nanomaterials for biomedical applications, Bioconjug. Chem., 25 (2014) 1609-1619. [180] B. Cornelissen, S. Able, V. Kersemans, P.A. Waghorn, S. Myhra, K. Jurkshat, A. Crossley, K.A.

PT

Vallis, Nanographene oxide-based radioimmunoconstructs for in vivo targeting and SPECT imaging of HER2-positive tumors, Biomaterials, 34 (2013) 1146-1154.

imaging of tumors by

198,199

RI

[181] Y. Fazaeli, O. Akhavan, R. Rahighi, M.R. Aboudzadeh, E. Karimi, H. Afarideh, In vivo SPECT Au-labeled graphene oxide nanostructures, Mater. Sci. Eng. C Mater. Biol.

SC

Appl., 45 (2014) 196-204.

[182] H.P. Cong, J.J. He, Y. Lu, S.H. Yu, Water-soluble magnetic-functionalized reduced graphene oxide

NU

sheets: in situ synthesis and magnetic resonance imaging applications, Small, 6 (2010) 169-173. [183] E. Peng, E.S. Choo, P. Chandrasekharan, C.T. Yang, J. Ding, K.H. Chuang, J.M. Xue, Synthesis of

MA

manganese ferrite/graphene oxide nanocomposites for biomedical applications, Small, 8 (2012) 36203630.

[184] S. Kanakia, J.D. Toussaint, S.M. Chowdhury, G. Lalwani, T. Tembulkar, T. Button, K.R. Shroyer,

D

W. Moore, B. Sitharaman, Physicochemical characterization of a novel graphene-based magnetic

TE

resonance imaging contrast agent, Int. J. Nanomedicine, 8 (2013) 2821-2833. [185] R. Romero-Aburto, T.N. Narayanan, Y. Nagaoka, T. Hasumura, T.M. Mitcham, T. Fukuda, P.J. Cox, R.R. Bouchard, T. Maekawa, D.S. Kumar, S.V. Torti, S.A. Mani, P.M. Ajayan, Fluorinated

5637.

AC CE P

graphene oxide; a new multimodal material for biological applications, Adv. Mater., 25 (2013) 5632-

[186] X. Sun, Z. Liu, K. Welsher, J.T. Robinson, A. Goodwin, S. Zaric, H. Dai, Nano-graphene oxide for cellular imaging and drug delivery, Nano Res., 1 (2008) 203-212. [187] K. Yang, S. Zhang, G. Zhang, X. Sun, S.T. Lee, Z. Liu, Graphene in mice: ultrahigh in vivo tumor uptake and efficient photothermal therapy, Nano Lett., 10 (2010) 3318-3323. [188] M.L. Chen, J.W. Liu, B. Hu, M.L. Chen, J.H. Wang, Conjugation of quantum dots with graphene for fluorescence imaging of live cells, Analyst, 136 (2011) 4277-4283. [189] S.H. Hu, Y.W. Chen, W.T. Hung, I.W. Chen, S.Y. Chen, Quantum-dot-tagged reduced graphene oxide nanocomposites for bright fluorescence bioimaging and photothermal therapy monitored in situ, Adv. Mater., 24 (2012) 1748-1754. [190] M.L. Chen, Y.J. He, X.W. Chen, J.H. Wang, Quantum-dot-conjugated graphene as a probe for simultaneous cancer-targeted fluorescent imaging, tracking, and monitoring drug delivery, Bioconjug. Chem., 24 (2013) 387-397. [191] J. Lu, P.S. Yeo, C.K. Gan, P. Wu, K.P. Loh, Transforming C60 molecules into graphene quantum dots, Nat. Nanotechnol., 6 (2011) 247-252. 55

ACCEPTED MANUSCRIPT [192] L. Zhang, Y. Xing, N. He, Y. Zhang, Z. Lu, J. Zhang, Z. Zhang, Preparation of graphene quantum dots for bioimaging application, J. Nanosci. Nanotechnol., 12 (2012) 2924-2928. [193] Y. Chong, Y. Ma, H. Shen, X. Tu, X. Zhou, J. Xu, J. Dai, S. Fan, Z. Zhang, The in vitro and in vivo

PT

toxicity of graphene quantum dots, Biomaterials, 35 (2014) 5041-5048. [194] N. Abdullah Al, J.E. Lee, I. In, H. Lee, K.D. Lee, J.H. Jeong, S.Y. Park, Target delivery and cell

RI

imaging using hyaluronic acid-functionalized graphene quantum dots, Mol. Pharm., 10 (2013) 37363744.

SC

[195] X. Wang, X. Sun, J. Lao, H. He, T. Cheng, M. Wang, S. Wang, F. Huang, Multifunctional graphene quantum dots for simultaneous targeted cellular imaging and drug delivery, Colloids Surf. B

NU

Biointerfaces, 122 (2014) 638-644.

[196] P. Roy, A.P. Periasamy, C.Y. Lin, G.M. Her, W.J. Chiu, C.L. Li, C.L. Shu, C.C. Huang, C.T.

MA

Liang, H.T. Chang, Photoluminescent graphene quantum dots for in vivo imaging of apoptotic cells, Nanoscale, 7 (2015) 2504-2510.

[197] L. Deng, L. Liu, C. Zhu, D. Li, S. Dong, Hybrid gold nanocube@silica@graphene-quantum-dot

D

superstructures: synthesis and specific cell surface protein imaging applications, Chem. Commun.

TE

(Camb), 49 (2013) 2503-2505.

[198] T. Wen, B. Yang, Y. Guo, J. Sun, C. Zhao, S. Zhang, M. Zhang, Y. Wang, Organosilanefunctionalized graphene quantum dots and their encapsulation into bi-layer hollow silica spheres for

AC CE P

bioimaging applications, Phys. Chem. Chem. Phys., 16 (2014) 23188-23195. [199] L. Zhou, L. Zhou, X. Ge, J. Zhou, S. Wei, J. Shen, Multicolor imaging and the anticancer effect of a bifunctional silica nanosystem based on the complex of graphene quantum dots and hypocrellin A, Chem. Commun. (Camb), 51 (2015) 421-424. [200] G. Lalwani, X. Cai, L. Nie, L.V. Wang, B. Sitharaman, Graphene-based contrast agents for photoacoustic and thermoacoustic tomography, Photoacoustics, 1 (2013) 62-67. [201] M.A. Patel, H. Yang, P.L. Chiu, D.D. Mastrogiovanni, C.R. Flach, K. Savaram, L. Gomez, A. Hemnarine, R. Mendelsohn, E. Garfunkel, H. Jiang, H. He, Direct production of graphene nanosheets for near infrared photoacoustic imaging, ACS Nano, 7 (2013) 8147-8157. [202] Z. Sheng, L. Song, J. Zheng, D. Hu, M. He, M. Zheng, G. Gao, P. Gong, P. Zhang, Y. Ma, L. Cai, Protein-assisted fabrication of nano-reduced graphene oxide for combined in vivo photoacoustic imaging and photothermal therapy, Biomaterials, 34 (2013) 5236-5243. [203] X. Ma, H. Tao, K. Yang, L. Feng, L. Cheng, X. Shi, Y. Li, L. Guo, Z. Liu, A functionalized graphene oxide-iron oxide nanocomposite for magnetically targeted drug delivery, photothermal therapy, and magnetic resonance imaging, Nano Res., 5 (2012) 199-212.

56

ACCEPTED MANUSCRIPT [204] M. Zhang, Y. Cao, Y. Chong, Y. Ma, H. Zhang, Z. Deng, C. Hu, Z. Zhang, Graphene oxide based theranostic platform for T1-weighted magnetic resonance imaging and drug delivery, ACS Appl. Mater. Interfaces, 5 (2013) 13325-13332.

PT

[205] J. Shi, L. Wang, J. Zhang, R. Ma, J. Gao, Y. Liu, C. Zhang, Z. Zhang, A tumor-targeting nearinfrared laser-triggered drug delivery system based on GO@Ag nanoparticles for chemo-photothermal

RI

therapy and X-ray imaging, Biomaterials, 35 (2014) 5847-5861.

[206] L. Nie, P. Huang, W. Li, X. Yan, A. Jin, Z. Wang, Y. Tang, S. Wang, X. Zhang, G. Niu, X. Chen,

SC

Early-stage imaging of nanocarrier-enhanced chemotherapy response in living subjects by scalable photoacoustic microscopy, ACS Nano, 8 (2014) 12141-12150.

NU

[207] D. Joseph, S. Seo, D.R. Williams, K.E. Geckeler, Double-stranded DNA-graphene hybrid: preparation and anti-proliferative activity, ACS Appl. Mater. Interfaces, 6 (2014) 3347-3356.

MA

[208] L. Zhang, Z. Lu, Q. Zhao, J. Huang, H. Shen, Z. Zhang, Enhanced chemotherapy efficacy by sequential delivery of siRNA and anticancer drugs using PEI-grafted graphene oxide, Small, 7 (2011) 460-464.

D

[209] H. Kim, R. Namgung, K. Singha, I.K. Oh, W.J. Kim, Graphene oxide-polyethylenimine

TE

nanoconstruct as a gene delivery vector and bioimaging tool, Bioconjug. Chem., 22 (2011) 2558-2567. [210] H. Bao, Y. Pan, Y. Ping, N.G. Sahoo, T. Wu, L. Li, J. Li, L.H. Gan, Chitosan-functionalized graphene oxide as a nanocarrier for drug and gene delivery, Small, 7 (2011) 1569-1578.

AC CE P

[211] L. Feng, X. Yang, X. Shi, X. Tan, R. Peng, J. Wang, Z. Liu, Polyethylene glycol and polyethylenimine dual-functionalized nano-graphene oxide for photothermally enhanced gene delivery, Small, 9 (2013) 1989-1997.

[212] D. Yin, Y. Li, H. Lin, B. Guo, Y. Du, X. Li, H. Jia, X. Zhao, J. Tang, L. Zhang, Functional graphene oxide as a plasmid-based Stat3 siRNA carrier inhibits mouse malignant melanoma growth in vivo, Nanotechnology, 24 (2013) 105102. [213] Z.M. Markovic, L.M. Harhaji-Trajkovic, B.M. Todorovic-Markovic, D.P. Kepic, K.M. Arsikin, S.P. Jovanovic, A.C. Pantovic, M.D. Dramicanin, V.S. Trajkovic, In vitro comparison of the photothermal anticancer activity of graphene nanoparticles and carbon nanotubes, Biomaterials, 32 (2011) 1121-1129. [214] Q. Liang, W. Ma, Y. Shi, Z. Li, X. Yang, Easy synthesis of highly fluorescent carbon quantum dots from gelatin and their luminescent properties and applications, Carbon, 60 (2013) 421-428. [215] G. Huang, X. Zhu, H. Li, L. Wang, X. Chi, J. Chen, X. Wang, Z. Chen, J. Gao, Facile integration of multiple magnetite nanoparticles for theranostics combining efficient MRI and thermal therapy, Nanoscale, 7 (2015) 2667-2675.

57

ACCEPTED MANUSCRIPT [216] H. Zhang, H. Wu, J. Wang, Y. Yang, D. Wu, Y. Zhang, Y. Zhang, Z. Zhou, S. Yang, Graphene oxide-BaGdF5 nanocomposites for multi-modal imaging and photothermal therapy, Biomaterials, 42 (2015) 66-77.

PT

[217] S. Wang, Q. Zhang, X.F. Luo, J. Li, H. He, F. Yang, Y. Di, C. Jin, X.G. Jiang, S. Shen, L. Fu de, Magnetic graphene-based nanotheranostic agent for dual-modality mapping guided photothermal therapy

RI

in regional lymph nodal metastasis of pancreatic cancer, Biomaterials, 35 (2014) 9473-9483. [218] J.T. Robinson, S.M. Tabakman, Y. Liang, H. Wang, H.S. Casalongue, D. Vinh, H. Dai, Ultrasmall

SC

reduced graphene oxide with high near-infrared absorbance for photothermal therapy, J. Am. Chem. Soc., 133 (2011) 6825-6831.

NU

[219] K. Yang, J. Wan, S. Zhang, B. Tian, Y. Zhang, Z. Liu, The influence of surface chemistry and size of nanoscale graphene oxide on photothermal therapy of cancer using ultra-low laser power, Biomaterials,

MA

33 (2012) 2206-2214.

[220] Y. Tahara, M. Nakamura, M. Yang, M. Zhang, S. Iijima, M. Yudasaka, Lysosomal membrane destabilization induced by high accumulation of single-walled carbon nanohorns in murine macrophage

D

RAW 264.7, Biomaterials, 33 (2012) 2762-2769.

TE

[221] O. Akhavan, E. Ghaderi, Graphene nanomesh promises extremely efficient in vivo photothermal therapy, Small, 9 (2013) 3593-3601.

[222] H. Dong, Z. Zhao, H. Wen, Y.l.t.e.c. Li, F. Guo, A. Shen, F. Pilger, C. Lin, D. Shi, Poly(ethylene

2271.

AC CE P

glycol) conjugated nano-graphene oxide for photodynamic therapy, Sci. China Chem., 53 (2010) 2265-

[223] P. Huang, C. Xu, J. Lin, C. Wang, X. Wang, C. Zhang, X. Zhou, S. Guo, D. Cui, Folic acidconjugated graphene oxide loaded with photosensitizers for targeting photodynamic therapy, Theranostics, 1 (2011) 240-250.

[224] J. Ge, M. Lan, B. Zhou, W. Liu, L. Guo, H. Wang, Q. Jia, G. Niu, X. Huang, H. Zhou, X. Meng, P. Wang, C.S. Lee, W. Zhang, X. Han, A graphene quantum dot photodynamic therapy agent with high singlet oxygen generation, Nat. Commun., 5 (2014) 4596. [225] X. Yan, G. Niu, J. Lin, A.J. Jin, H. Hu, Y. Tang, Y. Zhang, A. Wu, J. Lu, S. Zhang, P. Huang, B. Shen, X. Chen, Enhanced fluorescence imaging guided photodynamic therapy of sinoporphyrin sodium loaded graphene oxide, Biomaterials, 42 (2015) 94-102. [226] P. Rong, K. Yang, A. Srivastan, D.O. Kiesewetter, X. Yue, F. Wang, L. Nie, A. Bhirde, Z. Wang, Z. Liu, G. Niu, W. Wang, X. Chen, Photosensitizer loaded nano-graphene for multimodality imaging guided tumor photodynamic therapy, Theranostics, 4 (2014) 229-239. [227] M. Nurunnabi, Z. Khatun, G.R. Reeck, D.Y. Lee, Y.K. Lee, Photoluminescent graphene nanoparticles for cancer phototherapy and imaging, ACS Appl. Mater. Interfaces, 6 (2014) 12413-12421. 58

ACCEPTED MANUSCRIPT [228] X. Yan, H. Hu, J. Lin, A.J. Jin, G. Niu, S. Zhang, P. Huang, B. Shen, X. Chen, Optical and photoacoustic dual-modality imaging guided synergistic photodynamic/photothermal therapies, Nanoscale, 7 (2015) 2520-2526.

PT

[229] H. Luo, L. Lu, F. Yang, L. Wang, X. Yang, Q. Luo, Z. Zhang, Nasopharyngeal cancer-specific therapy based on fusion peptide-functionalized lipid nanoparticles, ACS Nano, 8 (2014) 4334-4347.

RI

[230] L.P. Wu, D. Wang, L. Parhamifar, A. Hall, G.Q. Chen, S.M. Moghimi, Poly(3-hydroxybutyrate-coR-3-hydroxyhexanoate) nanoparticles with polyethylenimine coat as simple, safe, and versatile vehicles

SC

for cell targeting: population characteristics, cell uptake, and intracellular trafficking, Adv. Healthc. Mater., 3 (2014) 817-824.

NU

[231] C. Wang, S. Ravi, U.S. Garapati, M. Das, M. Howell, J. MallelaMallela, S. Alwarapan, S.S. Mohapatra, S. Mohapatra, Multifunctional chitosan magnetic-graphene (CMG) nanoparticles: a

MA

theranostic Platform for tumor-targeted co-delivery of drugs, genes and MRI contrast agents, J. Mater. Chem. B, 1 (2013) 4396-4405.

[232] H.W. Yang, C.Y. Huang, C.W. Lin, H.L. Liu, C.W. Huang, S.S. Liao, P.Y. Chen, Y.J. Lu, K.C.

D

Wei, C.C. Ma, Gadolinium-functionalized nanographene oxide for combined drug and microRNA

TE

delivery and magnetic resonance imaging, Biomaterials, 35 (2014) 6534-6542. [233] M. Ghosh, S.K. Sonkar, M. Saxena, S. Sarkar, Carbon nano-onions for imaging the life cycle of Drosophila melanogaster, Small, 7 (2011) 3170-3177.

AC CE P

[234] J. Bartelmess, S.J. Quinn, S. Giordani, Carbon nanomaterials: multi-functional agents for biomedical fluorescence and Raman imaging, Chem. Soc. Rev., 2014, Epub. [235] Y. Xiao, H. Hong, A. Javadi, J.W. Engle, W. Xu, Y. Yang, Y. Zhang, T.E. Barnhart, W. Cai, S. Gong, Multifunctional unimolecular micelles for cancer-targeted drug delivery and positron emission tomography imaging, Biomaterials, 33 (2012) 3071-3082. [236] Y. Zhang, D. Petibone, Y. Xu, M. Mahmood, A. Karmakar, D. Casciano, S. Ali, A.S. Biris, Toxicity and efficacy of carbon nanotubes and graphene: the utility of carbon-based nanoparticles in nanomedicine, Drug Metab. Rev., 46 (2014) 232-246. [237] A.B. Seabra, A.J. Paula, R. de Lima, O.L. Alves, N. Duran, Nanotoxicity of graphene and graphene oxide, Chem. Res. Toxicol., 27 (2014) 159-168. [238] M. Yang, M. Zhang, Y. Tahara, S. Chechetka, E. Miyako, S. Iijima, M. Yudasaka, Lysosomal membrane permeabilization: carbon nanohorn-induced reactive oxygen species generation and toxicity by this neglected mechanism, Toxicol. Appl. Pharmacol., 280 (2014) 117-126. [239] P. Miao, K. Han, Y. Tang, B. Wang, T. Lin, W. Cheng, Recent advances in carbon nanodots: synthesis, properties and biomedical applications, Nanoscale, 7 (2015) 1586-1595.

59

ACCEPTED MANUSCRIPT [240] K. Bhattacharya, C. Sacchetti, R. El-Sayed, A. Fornara, G.P. Kotchey, J.A. Gaugler, A. Star, M. Bottini, B. Fadeel, Enzymatic 'stripping' and degradation of PEGylated carbon nanotubes, Nanoscale, 6 (2014) 14686-14690.

PT

[241] Y. Li, L. Feng, X. Shi, X. Wang, Y. Yang, K. Yang, T. Liu, G. Yang, Z. Liu, Surface coatingdependent cytotoxicity and degradation of graphene derivatives: towards the design of non-toxic,

RI

degradable nano-graphene, Small, 10 (2014) 1544-1554.

[242] S. Matsumura, S. Sato, M. Yudasaka, A. Tomida, T. Tsuruo, S. Iijima, K. Shiba, Prevention of

SC

carbon nanohorn agglomeration using a conjugate composed of comb-shaped polyethylene glycol and a peptide aptamer, Mol. Pharm., 6 (2009) 441-447.

NU

[243] Y. Li, J. Liu, H. Dong, G. Liu, H. Hu, Engineering of a Pluronic F127 functionalized

AC CE P

TE

D

MA

magnetite/graphene nanohybrid for chemophototherapy, Nanotechnology, 25 (2014) 065602.

60

MA

NU

SC

RI

PT

ACCEPTED MANUSCRIPT

AC CE P

TE

D

Graphical abstract

61

Theranostic applications of carbon nanomaterials in cancer: Focus on imaging and cargo delivery.

Carbon based nanomaterials have attracted significant attention over the past decades due to their unique physical properties, versatile functionaliza...
1MB Sizes 12 Downloads 35 Views