Near-Infrared Light-Triggered Polymeric Nanomicelles for Cancer

Oct 30, 2017 - Polymeric nanomicelles (PNMs) are a class of 1–200 nm nanoparticles prepared by amphiphilic copolymers. ...... Zheng , N.; Dai , W. B...
1 downloads 13 Views 2MB Size
Subscriber access provided by READING UNIV

Review

Near-infrared light-triggered polymeric nanomicelles for cancer therapy and imaging LEI LI, Xin Pang, and Gang Liu ACS Biomater. Sci. Eng., Just Accepted Manuscript • DOI: 10.1021/acsbiomaterials.7b00648 • Publication Date (Web): 30 Oct 2017 Downloaded from http://pubs.acs.org on November 2, 2017

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

ACS Biomaterials Science & Engineering is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 45

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

Near-infrared Light-triggered Polymeric Nanomicelles for Cancer Therapy and Imaging Lei Li#, Xin Pang#, and Gang Liu* State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China #These authors are contributed equally to this work *Correspondence author E-mail: [email protected]

1

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Abstract: Early diagnosis and efficient therapy are very important for cancer management. Among the various intelligent delivery systems, near-infrared (NIR) light-triggered polymeric nanomicelles (PNMs) have emerged as a promising approach for imaging and photoactive delivery. This platform can be used for spatially and temporally controlled remote release of therapeutic payload molecules under NIR light irradiation. This review discusses the design and characteristics of PNMs, as well as their recent application in imaging, drug delivery, and theranostics in photothermal therapy and photodynamic therapy. The challenges and potential of PNMs in biomedical applications are also highlighted. Keywords: near-infrared light, polymer, nanomicelles, cancer therapy, cancer imaging

2

ACS Paragon Plus Environment

Page 2 of 45

Page 3 of 45

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

1.

Introduction Cancer is a major public health problem and is responsible for about 8.2 million

deaths around the world1. There have been considerable advances in knowledge about cancer in the past few decades. The conventional therapeutic options to combat cancer include surgery, radiotherapy, chemotherapy, and combinations of them2,3. Surgery is the most important approach to remove solid tumors but is highly invasive and can hardly eliminate all tumor cells, especially when metastasis has already happened4. Chemotherapy and radiotherapy also have significant concerns, such as severe toxic side effects, limited therapeutic efficacy, induced drug resistance, and long-term damage to the immune system5-8. Therefore, big challenges still remain for early diagnosis and highly targeted and controlled cancer treatments. In recent years, nanotechnology has become a promising alternative approach for cancer “theranostics”, which Funkhouser coined in 2002 to explain the integration of concurrent diagnosis and treatment modalities at the same dosage range9,10. Nanomaterials with high targeting ability and multifunctionality are a significant focus in theranostics. Nanomaterials can reach the site of disease by passive and/or active targeting11-15, and they play an important role in cancer diagnosis in reporting the location of the disease, its stage, or the response to treatment16-19. Nanomaterials can also deliver therapeutic agents to a targeted site at necessary concentrations in response to external stimuli or molecular signals. This reduces the risk of toxicity to the normal tissues around the lesion and other organs20-22. In addition, nanomaterials that absorb a specific wavelength of light have also been exploited in drug delivery 3

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 4 of 45

and molecular imaging for cancer theranositics. Near-infrared (NIR) light is defined as light with wavelengths of 700-1100 nm, which can be exploited to distinctively avoid autofluorescence in the range of 270-665 nm in vivo. This allows penetration for deep-tissue imaging with higher contrast23,24. NIR nanomaterials such as gold nanoparticles25 carbon nanotubes (CNTs)26, graphene oxide (GO)27, and upconversion nanocrystals (UNCs)28, have become efficient tools to visualize, detect, and treat cancer29,30. Such materials could realize the drug release with on-demand control or molecular imaging remotely triggered by exposure to NIR light. Furthermore, nanomaterials doped with organic NIR fluorescent molecules are also emerging as another promising candidate for in vivo imaging and cancer treatment, demonstrating many advantages over pristine molecular NIR fluorescent probes. Nanoparticle formulations could provide multimodal imaging as well as complementary anatomical and physiological information by co-doping with imaging agents of other modalities, such as magnetic resonance imaging (MRI)31 or PET/SPECT imaging

32,33

. Inert and biocompatible

polymers such as polyethylene glycol (PEG) could also be used easily to modify the surfaces of nanoparticles and allow them to evade capture and degradation by the reticuloendothelial

system

(RES)34,35.

Interestingly,

nanoparticles

could

be

preferentially taken up by the malignant tissues by passive targeting by virtue of the enhanced permeability and retention (EPR) effect36,37. Polymeric nanomicelles (PNMs) are a class of 1-200 nm nanoparticles prepared by amphiphilic copolymers. They contain two individual functional sections: an 4

ACS Paragon Plus Environment

Page 5 of 45

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

“inner core” and an “outer shell”. The outer shell is usually composed of a hydrophilic block like PEG, which controls the pharmacokinetic properties in vivo. The inner core is made up of a hydrophobic block, which is responsible for drug entrapment, stability, and drug-release characteristics38,39. The uniqueness of PNMs is that their copolymeric structures can encapsulate hydrophobic drugs in the inner core, and the outer shell can be easily modified or engineered to achieve desired properties. This creates promising opportunities for using PNMs in drug delivery and targeting. Polymeric micelles with high stability both in vitro and in vivo show good biocompatibility and can solubilize a broad variety of poorly soluble pharmaceuticals, as well as improve their biological behavior40. Micelle-incorporated anticancer drugs, such as doxorubicin (DOX) and paclitaxel, have better accumulation in tumors than in non-target tissues, which could minimize the toxicity of drugs towards normal tissue41-43. Such drugs can also be protected from biodegradation in the body and achieve prolonged half-lives in vivo, resulting in enhanced efficacy44. Recently, PNMs encapsulating NIR phosphorescent dye have been increasingly investigated as efficient candidates for optical bioimaging probes and drug delivery systems45-47. In this review, we mainly focus on the NIR polymeric nanomicelles and present strategies for their design and modification (Figure 1), as well as introduce their NIR response characteristics and theranostic applications in cancer. Their future prospects and new challenges for theranostic applications in cancer treatments are also discussed.

5

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Figure 1. Schematic illustration of multifunctional NIR agents for cancer imaging and phototherapy.

2.

Design and characteristic of PNMs Micelles are self-assembling nanosized colloidal particles with a hydrophobic

core and a hydrophilic shell, which have successfully been used as pharmaceutical carriers for water-insoluble drugs17. Some commonly used micellar carriers are shown in Figure 2. PEG with a molecular weight from 1 to 15 kDa is the polymer most commonly used for the hydrophilic shell because of its physicochemical characteristics, including high water solubility, significant chain mobility, and low toxicity. Poly(N-vinyl-2-pyrrolidone) (PVP) is considered as a primary alternative to PEG48. PVP is highly biocompatible and has been used in formulations of particulate drug carriers such as liposomes49 and diblock polymer micelles50. Another hydrophilic candidate is poly(vinyl alcohol) (PVA). Poly(vinyl alcohol-co-vinyloleate) copolymer 6

ACS Paragon Plus Environment

Page 6 of 45

Page 7 of 45

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

was used to prepare micelles to enhance the transcutaneous permeation of retinyl palmitate51 .

Figure 2. Chemical structures of commonly used polymers as micellar carrier. In the case of the hydrophobic core, the most commonly used polymers are polyesters, polyethers and polyamino acids. The main advantage of polyesters is that many have been approved by the Food and Drug Administration (FDA) for biomedical

applications

in

humans,

including

poly(lactic

acid)

(PLA),

poly(lactic-co-glycolic acid) (PLGA), poly(e-caprolactone) (PCL), poly(trimethylene carbonate) (PTMC), and poly(propyleneoxide) (PPO)52. Biodegradable hydrophobic 7

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 8 of 45

blocks of polyesters are usually covalently bonded to hydrophilic blocks (mainly PEG) and then self-assembled into polymeric micelles. Other biodegradable hydrophobic block copolymers are composed of polyanhydrides, such as poly sebacic anhydride (PSA). These are also covalently bonded to hydrophilic blocks of PEG and have advantages of biocompatibility and degradation into non-toxic diacid compounds in vivo

53

. The poly (L-amino acid) (PAA) family includes poly(L-histidine) (polyHis),

poly(L-asparticacid)

(polyAsp),

poly(L-glutamic

acid)

(polyGlut)

and

poly(L-lysine)(polyLis). These are also employed as nanosized vehicles that are covalently bonded to hydrophilic blocks of PEG. These vehicles are not only biodegradable and biocompatible53, but they also have pH-sensitivity and potential use in pH-dependent drug release at tumor sites54. The PEGylated phospholipid-based block

copolymer

family

includes

PEG-distearoylphos-phatidylethanolamine

(PEG-PE). This family can also self-assemble into nanostructured micelles. Their advantages are their simpler and more reproducible preparation, the avoidance of macrophage

phagocytosis system (MPS) uptake, longer circulation times,

biocompatibility, and almost no toxicity55 Recently micelle-forming di- and tri-block-copolymers have been of particular interest as a long-circulating pharmaceutical carrier56,57. Micelle-forming co-polymers are usually synthesized by anionic polymerization, ring-opening polymerization, or polymerization using poly (ethylene oxide)-based initiators58. These allow for synthesis of amphiphilic copolymers with different molecular mass and different hydrophobic-lipophilic

balance

by

controlling

the

8

ACS Paragon Plus Environment

lengths

of

the

Page 9 of 45

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

hydrophobic/hydrophilic

blocks.

Different

physicochemical

and

biological

characteristics can be realized by varying the molecular size of different blocks and their molar ratio in the copolymer. Larger hydrophobic blocks result in a bigger core size and greater ability to entrap hydrophobic drugs. Increasing the length of the hydrophilic block leads to an increase of the critical micelle concentration (CMC). These nanoparticulate drug delivery systems for solid tumor treatment usually undergo the steps of circulation, accumulation, penetration, internalization and release to deliver drugs into cancer cells, which is determined by surface properties, stability, targeting groups, shape and size of nanoparticles. Generally, smaller micelles (e.g., 30 nm) exhibits better tumor penetration and results in high anticancer activity59. Because of such attractive characteristics, a series of novel PNMs with different chemical structures and biological behaviors were recently developed, and increasingly applied to the field of NIR-triggered drug delivery systems. As a typical carrier material for PNMs, the biodegradable diblock copolymer has been developed from poly-(ethylene glycol)-block-poly (ε-caprolactone) (PEG-b-PCL) decorated by a fluorescent moiety (Cy7) on the surface. It could be simultaneously used for anticancer drug delivery and noninvasive optical imaging60. Additionally, efficient probes

have

been

obtained

from

NIR

phosphorescent

dye

[Pt

(II)-tetraphenyltetranaphthoporphyrin, Pt (TPNP)] encapsulated by polymeric nanomicelles61. They have a crucial advantage over conventional NIR-fluorescent probes due to the large spectral separation between the absorption and phosphorescence emission. This ensures a dramatic decrease in the level of 9

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

background autofluorescence and scattered excitation light in the spectral range where the signal from phosphorescent probe is observed. Recently, a novel acid-sensitive polymeric prodrug of PMPC-b-P(MEMA-hydrazide-DOX) was also introduced to NIR-triggered PNMs62. After loading with a cyanine dye IR-780, such polymeric prodrug micelles could effectively release DOX in acidic endosomal compartments and tumor mass while remaining pharmacologically inactive in blood circulation. Benefiting from the localized hyperthermia induced by co-delivered IR-780 upon NIR laser irradiation, the intracellular DOX accumulation and anticancer efficacy in vitro and in vivo were significantly increased. Widespread use of PNMs is expected in the field of drug and gene delivery. Therefore, much more work should be done to develop prototypes of novel polymer micelles with NIR fluorescence imaging capabilities. 3.

NIR-triggered PNMs for imaging Light in the NIR range (700-1000 nm) exhibits unique advantages, such as deep

tissue penetration, low fluorescence background, and limited photo-damage. NIR light is minimally absorbed by the major light absorbers in living systems, including water, lipids, and some intrinsic proteins, such as hemoglobin (Hb) and oxyhemoglobin (HbO2). Combined NIR fluorescence and molecular imaging techniques have promising capabilities for noninvasive visualization of biological structures63. Such techniques could provide a new route for NIR dye-encapsulated nanoparticles for cancer targeting and imaging. 3.1. NIR fluorescence imaging 10

ACS Paragon Plus Environment

Page 10 of 45

Page 11 of 45

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

An excellent NIR probe is necessary for satisfactory NIR fluorescence imaging of cancer. Compared to computed tomography (CT) and magnetic resonance imaging (MRI) which have been of great help in characterizing diseases but not useful for intraoperative assessment, NIR fluorescence imaging has exhibited great success as an optical technique for real-time imaging of surgical targets with high imaging resolution and distinct intraoperative sensitivity. Recently, the use of a photostable, biocompatible, bright, and tumor-specific NIR fluorescent agents has been approved for intraoperative imaging in clinical trials and has demonstrated importance. Examples include indocyanine green (ICG) and methylene blue64. However, their disadvantages include poor aqueous stability, low sensitivity, low specificity, and low temporal resolution in bioimaging. As a smart drug delivery system, PNMs can improve the physiochemical properties and biological behavior of NIR probes, and therefore show great potential in intraoperative surgical guidance and disease diagnosis. Wu et al. developed ICG-encapsulated micelles using PEG-PLL-PLLeu copolymer as a carrier skeleton. These self-assembled ICG micelles exhibited improved quantum yield and fluorescence stability, as well as high cellular uptake rates in vitro. They also showed excellent passive tumor targeting capability and long circulation time in animal experiments, indicating broad prospects of the fluoro-probe for tumor diagnosis and targeted imaging65. Heptamethine dyes feature simultaneous tumor targeting and NIR imaging capabilities and have shown great advantages in experiments and in a clinical setting. With low molecular weight (usually less than 1000 Da), these dyes show some 11

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 12 of 45

similarity to biomacromolecules and are well suited for imaging with superior pharmaceutical and dynamic properties 66. They also have great stability and persist in tumors for more than two weeks. This makes it possible for repeated imaging after a single administration of these dyes. IR-780 iodide and IR-783 are two prototypical dyes that reach the NIR region at around 780 nm. They can be detected conveniently by an NIR fluorescent detection system without significant autofluorescence interference67. Recently, amphiphilic micelles based on D-a-tocopheryl polyethylene glycol succinate (TPGS) and D-a-tocopheryl succinate (TOS) were developed for IR780 delivery. The IR780-loaded micelles have a narrow size range of 115 to 123 nm, and they present optimal characteristics for cellular uptake and imaging under NIR irradiation at 808 nm68. BODIPY

(borondipyrromethane)

dyes

have

a

general

structure

of

4,40-difluoro-4-bora-3a, 4a-diaza-s-indacene and are potential candidates for laser dyes, molecular photonic wires, fluorescent switches, and bioprobes. These dyes have advantages of sharp fluorescence with high quantum yield and excellent thermal and photochemical stability69. Recently, a novel BODIPY-Br2-loaded pH-responsive polypeptide micelle was developed by Liu’s group using N-carboxyanhydride (NCA) monomer via a ring-opening polymerization and click reaction70. Due to the inherent fluorescence emission, these BODIPY-Br2 PNMs enable the possibility for biomedical imaging. Therefore, their cellular internalization by cancer cells could be successfully traced by NIR fluorescence imaging. Upon light irradiation, the loaded BODIPY-Br2 could effectively generate reactive oxygen species and induce NIR 12

ACS Paragon Plus Environment

Page 13 of 45

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

imaging-guided photodynamic therapy (PDT). A variety of dye-encapsulated micelles have been prepared for cancer fluorescence imaging and treatment, but desirable biodegradability and biocompatibility are still urgent needs for the development of NIR naonoprobes in future research. 3.2. Photoacoustic imaging (PAI) PAI combines the high contrast of optical imaging with the spatiotemporal resolution of ultrasound imaging71. When laser pulses are applied, contrast agents or biological tissues absorb some of the delivered optical energy and convert it into heat. Then the heat-induced transient thermoelastic expansion leads to the generation of broadband ultrasonic emission, which can be detected by an acoustic detector and analyzed to reconstruct PA images72,73. Recently, a new PA organic contrast agent based on semiconducting polymer nanoparticles has been developed for PAI in both first NIR window (NIR-I, 650-950 nm) and second NIR window (NIR-II, 1000-1700 nm)(Figure 3). Due to the weaker background PA signals from biological tissues in NIR-II window, 1.4-times higher signal-to-noise ratio (SNR) of contrast-enhanced PA images at 1064 nm could be detected as compared with that at 750 nm at the imaging depth of 3 cm72.

13

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Figure 3.

Preparation of semiconducting polymer nanoparticles for NIR-II PA

imaging. (a) Synthetic route of poly(diketopyrrolopyrrole-alt-thiadiazoloquinoxaline). (b) Chemical structure of poly[diketopyrrolopyrrole-alt-thiophene]. (c) Schematic illustration for preparation of the polymer nanoparticles. Reproduced with permission from ref 72. Copyright 2017 American Chemical Society. PAI exhibits greater performance on functional information imaging comparing with positron emission tomography (PET) and single photon emission computed tomography (SPECT). These nuclear imaging modalities depend on the use of nuclear radioisotopes for gamma-ray detection but have poor spatial resolution. Optical imaging, another technique to obtain functional information, is limited by the strong light absorption/scattering of skin, tissue and blood, leading to poor tissue penetration. Acoustic scattering in tissue, however, is several orders of magnitude weaker than optical scattering74. A combination of labels for different imaging modalities has been widely used in applications ranging from diagnosis of vulnerable atherosclerotic plaques to the detection of cancer. Liang et al. prepared CT/PA imaginable PNMs through conjugating hyaluronic acid (HA) with NIR dyes as hyaluronidase–active 14

ACS Paragon Plus Environment

Page 14 of 45

Page 15 of 45

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

cancer theranostics75,76. Recently, a series of perylene diimide (PDI) nanoparticles have been synthesized as dual PET/PA probes and PTT agents. It has been demonstrated that PDI nanoparticles with a size of 60 nm appear to be the best for tumor imaging and photothermal cancer therapy due to the maximum tumor accumulation efficiency (Figure 4)73.

Figure 4 Schematic illustration of the application of PDI nanoparticles as PET and PA imaging probes. Reproduced with permission from ref 73. Copyright 2017 American Chemical Society. 4.

NIR-triggered PNMs for drug delivery Controlled drug delivery systems that can selectively release chemotherapy

agents at a target site have been actively developed for cancer treatment. Increasing attention has been drawn to the delivery of therapeutic payloads using delivery 15

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

systems based on engineered nanomaterials, such as polymeric micelles77-79. The development of various stimuli-responsive delivery platforms has made it possible to have greater control over the delivery and release of anticancer agents in a temporal or spatial manner. Some of the release strategies that have been developed are pH alterations, redox activities, enzymatic reactions, overexpression of biomolecules (e.g., adenosine triphosphate, glutathione),80 and external triggers such as thermal energy, ultrasound waves, light irradiation, mechanical forces, and electromagnetic fields81,82. Among these external stimuli, light activation offers unprecedented control with ease of production, noninvasive nature, controllable intensity, and spatially confined application for finely controlled durations83,84. Different mechanisms have been involved in the light-activated drug delivery/release systems, which include bond-cleavage, isomerization, cross-linking, electrostatic assembly/disassembly, reduction, oxidation, photocaging/uncaging, and

nonlinear photoconversion

mechanisms (shown in Figure 5)85. However, problems with inevitable cellular phototoxicity and poor tissue penetration depth have limited the clinical potential of ultraviolet (UV) (wavelength: 100−400 nm) and short visible light (400−750 nm) as external stimuli for some photoactive delivery platforms. Nanomicelles that use NIR provide advantages over quantum dots and organic fluorescent materials in biological applications, such as low photodamage,

enhanced tissue penetration86,87, minimal

autofluorescence background88, and improved resistance to photobleaching and blinking. Therefore, more and more attention has been given to NIR nanomicelles as targeted delivery systems. 16

ACS Paragon Plus Environment

Page 16 of 45

Page 17 of 45

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

Figure 5. Different light-activated mechanisms used for drug delivery systems. Reproduced with permission from ref 85. Copyright 2017 American Chemical Society. 4.1. NIR PNMs delivering anticancer drugs Chemotherapeutic drugs for cancer with low molecular weight generally have a short circulation time and form low concentrations in tumors and metastases when they are administrated intravenously. Recently, nanoscale drug delivery systems have been developed as indispensable platforms for modern cancer therapy, such as liposomes and polymeric micelles. These could allow administered anticancer drugs to achieve a proper circulation time and tumor concentration, as well as attenuate their accumulation in potentially endangered healthy organs and tissues89,90. However, acquired drug resistance and poor targeting have limited the application of liposomes and lipid-based drug delivery systems. In the early 1990s, Kataoka’s group reported DOX-conjugated block copolymer micelles as a drug delivery system, which has been used for the delivery of many anticancer agents in preclinical and clinical studies91. PNMs could overcome multiple drug resistance through absolute targeting using various approaches, such as passive targeting, folate-mediated drug delivery systems, 17

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

and pH-sensitive and thermo-sensitive drug delivery systems. Another important problem of anticancer agents is their poor water-solubility, which has great concerns in therapeutic applications such as limited absorption and bioavailability and drug aggregation-related complications like embolism92,93. PNMs could significantly increase the water solubility of anticancer drugs by 10- to 5000times94. and have an inner core made up of a hydrophobic block copolymer nanomicelles, in which poorly water-soluble anticancer drugs can be entrapped, They also have an outer shell of hydrophilic block copolymer that reduces the interactions of drugs with the aqueous environment and keeps them stable95. The release of chemotherapeutic drugs by the drug delivery system can be controlled by light irradiation96-98, pH99-101 temperature102-104 enzymatic hydrolysis,105-107 and redox reactions108,109. Among these strategies, light irradiation works as an efficient and readily operated external stimulus because of its precise temporal and spatial control. The high-energy light of UV or visible light, which are frequently needed, are limited by poor tissue penetration and damage to health tissue. Longer-wavelength NIR light irradiation exhibits great priority due to its capability of deeper penetration into tissue and minimal damage to living cells110. Therefore, the controlled-release of molecules irradiated by NIR provides an alternative method of drug delivery and targeted therapy, and they have gained significant attention in recent years111,112. Recently, a novel PEG and pyrene-oxabicycloheptenealkyne (POA) modified PPy nanomaterials were synthesized to covalently append hydrophobic anticancer molecules113. Under NIR irradiation, PPy could mediate photothermal effect, thereby 18

ACS Paragon Plus Environment

Page 18 of 45

Page 19 of 45

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

triggering retro D-A reaction for drug release. Li et al. developed an NIR responsive drug delivery system based on thermosensitive block copolymer micelles of poly(oligo(ethyleneglycol) (POEGMA-b-PFMA)

methacrylate)-block-poly(furfuryl

methacrylate)

114

. When encapsulated with NIR dye indocyaninegreen (ICG)

and DOX, the PNMs could deliver the drug efficiently without premature drug leakage and release. Upon NIR irradiation at 805 nm, ICG located in the inner core of micelles potently generated photothermal effect. Such local increased temperature of the micellar cores could induce fast drug diffusion, resulting in the effective release of DOX from micelles. Similarly, a novel galactose targeted pH-responsive amphiphilic multiblock copolymer conjugated with both drug and NIR probe has been designed and exerted a fast and enhanced endocytosis for HepG2 cells (Figure 6)115. Therefore, NIR-responsive PNMs provide a promising platform for anticancer drugs with the possibility of controllable drug release, which could maximize the therapeutic efficacy and minimize their adverse side effects.

19

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Figure 6.

Structure of ligand targeted copolymer conjugated with DOX and NIR

probe, micellization, selective accumulation in liver cancer cell, and imaging of the endocytosis of the micelle with subsequent pH triggered drug release. Reproduced with permission from ref 115. Copyright 2015 American Chemical Society. 4.2. NIR PNMs delivering therapeutic genes Recently, novel nucleic acid-based therapies such as plasmid DNA and siRNA have been developed as an innovative medicine. This has been enabled by the great progress in the understanding of biological mechanisms of life processes at molecular level116,117. However, they are unstable under physiological conditions and have low cellular uptake efficiency because of their large molecular weight and anionic nature, which hinder their application in clinical treatment. DNA or RNA is rapidly eliminated by DNase and RNase when directly administered into the blood, so it is necessary to create a nano-platform to encapsulate them to achieve effective accumulation within the targeted tissues and intracellular delivery to the nucleus for gene therapy. Viral vectors are commonly used as gene carriers, but there are problems associated with the immune response and possible oncogene effects due to their recombination with endogenous genes117-119. Due to their advanced and tunable characteristics, PNMs offer a superior alternative as a gene carrier in regard to safety, mass production and cost120,121. They can be formed by anionically charged DNA or RNA and a block copolymer with hydrophilic and cationic segments through polyion complexation. Studies have shown that DNA in the PNMs have prolonged circulation in blood, which allows for the 20

ACS Paragon Plus Environment

Page 20 of 45

Page 21 of 45

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

efficient gene expression in diseased site after intravenous injection into mouse tail veins122. The new biodegradable polyethylenimine derivatives of siRNA-PEG/PEI (polyethylene imine) polyplex micelles have been designed for siRNA delivery with raised hydrophilicity, decreased cytotoxicity, and increased transfection efficacy. PEG–PCL–PEI triblock copolymers and cationic micelles shaped from poly(ethylene glycol)- bl-poly(propylene sulfide)-bl-poly(ethylene imine) (PEG-b-PPS-b-PEI) were designed for plasmid DNA (pDNA) transfection as non-viral vectors in a tumor model in vitro and in vivo. Some polymeric micelles have shown a high potential for use as a delivery system for gene therapy applications123. One example is ERβ, which is a potent inhibitor of cell proliferation in the HCT-8 human colon cancer cell line, which occurs through the regulation of cell cycle components. The ERβ gene could therefore significantly inhibit tumor growth. SOC-ICG-Der-01 was formed by hydrophobic fluorescent dye ICG-Der-01 and amphipathic N-succinyl-N-octyl chitosan (SOC) micelles through hydrophobic association. The micelles were further combined with the ERβ gene, resulting in significantly inhibitory efficiency on tumor growth in a xenograft model of colon cancer124. Therefore, considering the DNA-based therapeutics used in gene therapy including plasmid DNA (pDNA), oligonucleotides, DNAzymes aptamers and small interfering RNA (siRNA), the PNMs show great promise in effectively delivering the therapeutic agents to the cytoplasm and further importing into the nucleus for pDNA. 4.3. NIR PNMs delivering therapeutic protein drugs With the development of biotechnology, protein drugs have come to play an 21

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

important role in cancer treatment. Efficient internalization of protein drugs into tumor cells is critical for gaining the desired therapeutic effect. However, the plasma membrane of the tumor cell is an effective barrier that limits the internalization of protein drugs with high molecular weight. The tumor microenvironment has distinctive characteristics with upregulated proteolytic enzymes such as matrix metalloproteinases, which are required for cancer progression. This decreases the efficiency of protein drugs in cancer therapy. Furthermore, the instability, immunogenicity and short half-life of protein drugs also restrict their wide application in routine therapies against cancer. Polymeric micelles with improved tumor extravasation and penetration offer substantial benefits as nanocarriers for delivering protein drugs. Benefiting from the surface coverage by commonly-used PEG strands, PNMs present high and versatile loading of bioactive molecules with controlled release, and exhibit prolonged blood circulation 125-128. Nanomicelles with small sizes of 10 to 100 nm have excellent tumor accumulation due to the EPR effect through atypical and leaky vasculatures, as well as poor lymphatic drainage in solid tumors129,130. Antibody fragments installed in polymeric micelles via maleimide-thiol conjugation have been developed to deliver platinum drugs selectively to pancreatic tumors, which achieved efficient drug delivery. A number of proteins are usually over-expressed in neovascular endothelial cells and some kind of tumor cells, such as vascular endothelial growth (VEGFR) and APN/CD13131. Therefore, it is promising to synthesize nanomicelles with double-targeting ability towards upregulated proteins in the endothelial cells of both angiogenic vessels and tumor cells. This would 22

ACS Paragon Plus Environment

Page 22 of 45

Page 23 of 45

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

maximize the utility of anticancer drugs by combining blood vessel destruction with conventional antitumor actions together132. NIR-triggered nanomicelles could provide a much more targeted and precise platform for protein drug delivery. Although much attention has been focused on the development of nanomicelles as a carrier for protein antitumor drugs, little work has been done on NIR-triggered nanomicelles for this purpose. Therefore, there is a promising opportunity for developing NIR nanomicelles as a protein drug carrier for cancer treatment. 5.

Theranostics Theranostics involves the simultaneous diagnosis and treatment of tumors. In

recent years, theranostics based on NIR nanomicelles have been emerging as a promising therapeutic paradigm because of their capabilities in diagnosis, drug delivery, and monitoring therapeutic responses133-141. Photothermal therapy (PTT) and PDT based on NIR nanomicelles could enable targeted cancer treatment and reduce damage to normal tissue142-145. The imaging properties of NIR PNMs also make it possible to detect and visualize tumor tissue while simultaneously ablating the tissues with the help of PTT and PDT. Thus, they may be advantageous in theranostics. 5.1. Photothermal therapy PTT is an emerging strategy in cancer therapy that involves directly converting light illumination into heat based on the strong absorption of thermally sensitive agents for targeted disease treatment. Photothermal conversion agents such as cyanine dyes can absorb high amounts of energy and release a great amount of heat upon irradiation with NIR light. This induces hyperthermia damage in the cancer cells and 23

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

tumor mass with deep tissue penetration146-148, eventually resulting in cell death. Hyperthermia is capable of killing tumor cells directly at > 45℃ or in combination with other therapies at 39-42℃149. The hyperthermia effect of nanoparticles could also enhance therapeutic efficacy by augmenting the cytotoxicity of some chemotherapeutic agents in vitro and in vivo for cancer treatment without increasing the drug dosage

150,151

. Effective photothermal conversion agents feature strong NIR

light absorption, great photothermal conversion efficiency, excellent biocompatibility and biodegradability, and the opportunity for real-time visualization for therapy 152,153. Some promising photothermal conversion agents (including organic nanomicelles) have been widely employed for effective light-triggered PTT treatment in vitro and in vivo. With the NIR laser irradiation, the temperature of IR-780 loaded PNMs increased by 20.6 ℃, which significantly changed the permeability and fluidity of the plasma membrane of MCF-7/ADR cells, enhanced the intracellular retention and accumulation of DOX, as well as reversed the drug resistance of tumor cells62. After being exposed to 805 nm light irradiation, the ICG and DOX co-loaded POEGMA-b-POMFMA PNMs showed significant temperature increase, achieving a synergistic effect of thermo-triggered drug release and PTT on Hela cells. Recently, Yuan’s group also developed a new photothermal therapeutic agent based on PEG-IR-780-C13 micelles with NIR absorption

154

. The micelles were stable in

aqueous conditions and exhibited no observable toxicity to cells and mice at therapeutic doses. PEG-IR-780-C13 micelles showed higher photostability than free IR-780 and conventional micelles containing IR-780. Furthermore, they exhibited 24

ACS Paragon Plus Environment

Page 24 of 45

Page 25 of 45

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

high tumor accumulation by the EPR effect and enabled fluorescence imaging of the tumor tissue. The micelles also acted as an excellent PTT agent for tumor ablation upon laser irradiation without rendering any appreciable toxicity. All these results clearly prove that NIR nanomicelles can be a safe theranostic agent for imaging-guided PTT. Future work is needed for optimization of the sizes, chemical treatments, sorting and assembly of nanostructures. Other topics that should be explored are the colloidal stability, tumor-homing capacity, and resistance to protein adsorption on NIR nanomicelles, which will improve their potential of the nanomicelles in cancer PTT. 5.2. Photodynamic therapy PDT is another efficient light-triggered cancer therapy method, which could induce the creation of singlet oxygen or other free radicals through photo-biochemical processes. This selectively causes acute microvascular injury, blood vessel blockage, and cell apoptosis tumors, thus achieving the purpose of local treatment155. Superior anticancer efficacy may be achieved from the synergistic effect between cyanine dye and therapeutic agent (Figure 7)156. The free radicals could be generated by the activation of a photosensitizer (PS) initiated by light in the NIR region. Due to the intrinsical non-invasiveness, safety, and highly spatiotemporal selectivity after light irradiation, PDT exhibits great advantages in cancer therapy compared to traditional therapeutic modalities. On the other hand, NIR light-responsive photosensitizer molecules have some unavoidable drawbacks, such as complicated synthesis procedures, low water solubility, and limited accumulation capabilities in targeted 25

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

areas. These drawbacks heavily restrict the clinical applications in deep-tissue therapy and imaging studies. Therefore, organic nanomaterials like polymetric nanomicelles with a large extinction coefficient in the NIR window have been extensively explored for PDT applications both in vitro and in vivo.

Figure 7. Mechanism illustration of cyanine-based micelles as a multimodal platform for synergistic cancer therapy. Reproduced with permission from ref 156. Copyright 2014 Ivyspring International. Micelles containing a new water-soluble amphiphilic two-photon absorption (2PA) chromophore have been developed, which were realized by using a block copolymer as a nanocarrier to encapsulate a hydrophobic porphyrin photo sensitizer inside the micelle core. With efficient energy transfer (as much as 96%), such 2PA PNMs performed effective singlet oxygen generation under irradiation of NIR light at 800 nm, and could be considered as an promising platform for cancer PDT. Although great progress has been achieved in the field of two-photon-activated photodynamic therapy (2P-PDT), the application of traditional photosensitizers in 2P-PDT remains a critical challenge in clinical practice due to their low 2PA cross sections. To address this issue,

26

ACS Paragon Plus Environment

Page 26 of 45

Page 27 of 45

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

Huang et al. developed a Ce6-conjugated core−shell unimolecular micelle using a large two-photon-absorbing hyperbranched conjugated polymer (HCP) as the inner core (HPE)

and a thermos-responsive hyperbranched polyether (HPE) as the outer shell (Figure 8)157. Under NIR irradiation, the local temperature increases and

HPE shell collapses, which shortens the distance between the HCP and photosensitizer. As a result, the fluorescence resonance energy transfer (FRET) between HCP core and Ce6 can be readily switched “on” to enhance the 2PA activity of Ce6 and, in turn, amplify its production of cytotoxic singlet oxygen for cancer PDT. Compared with free Ce6, the HCP@HPE-Ce6 micelles showed greater promise for enhanced photodynamic antitumor therapy with high stability in the blood and selective accumulation in tumors. As a typical carrier material for PNMs, PLA-PEG copolymer recently was extended to field of NIR-triggered PDT.

158

. After carrying a

pH-responsive fluorescent probe and a robust NIR photosensitizer of R16FP, the PLA-PEG PNMs under 808 nm irradiation could generate reactive oxygen species to mediate lysosomal destruction and subsequently trigger of lysosomal cell death. Besides, the significant inhibition on tumor growth and obvious necrosis of tumor tissue further demonstrated the potent PDT efficacy of PNMs for cancer therapy.

27

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Figure 8. Schematic of HCP@HPE-Ce6 micelles (A) and illustration of the combination of 2P-FRET and photothermal effect of NIR for cancer PDT (B). Reproduced with permission from ref 157. Copyright 2016 American Chemical Society. PDT has been approved by the FDA as a well-established and unique type of therapeutic modality for effective cancer treatment, and it is attracting more interest in cancer therapy. Great efforts should be made to develop improved PDT treatments and multimodality cancer imaging, including optical and PAI techniques for 28

ACS Paragon Plus Environment

Page 28 of 45

Page 29 of 45

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

theranostics in vivo. 6.

Conclusion and challenges In this review, we have summarized the current research progress on NIR PNMs

and their biomedical applications, such as imaging, PTT, PDT, and drug delivery. NIR PNMs have the following characteristics of absorbing NIR light and converting it to visible light or heat, thus serving as a drug carrier and enhancing tumor localization of therapeutic molecules. In spite of the promising characteristics of PMs, few clinical trials have been done. Thus far, there have been only a few PM-based formulations composed of anticancer agents in clinical trials, which are summarized in Table 1. Genexol-polymeric micelles are a formulation of paclitaxel encapsulated in monomethoxy-PEG-b-poly(D,L-lactide)

(MPEG-PDLLA),

which

is

currently

involved in 13 clinical trials and in a more advanced state of development. SP1049C, a Pluronic-based polymeric micelle formulation of DOX, is currently in phase III clinical studies and was designated as an orphan drug by the FDA in 2008. The clinical trials show highly promising results in these early stages of clinical development of the PNMs. Table 1 Examples of polymeric micelles in clinical trials Product name

Drug

Platform

Genexol-PM

Paclitaxel

mPEG-PLA

Statu s Phas e II/III/ IV Phas e II

Therapeutic References indication 159-162 Advanced, recurrent metastatic breast cancer advanced urothelial cancer, advanced head and neck cancer,

29

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Nanoxel-PM

Docetaxel

mPEG-PLA

NC 4016

Oxaliplatin

PEG-poly(gluta mic acid)

and advanced non-small-cel l lung cancer Phas ovarian e I/II cancer and advanced or metastatic pancreatic cancer phase lung, ovary I and breast cancer phase solid tumors I

Page 30 of 45

163

164

165 phase metastatic I/II pancreatic cancer NK105 Paclitaxel PEG and PAA phase Advanced or 166 II/III recurrent gastric cancer 167 NC-6004 Cisplatin PEG and PGA phase advanced I/II solid tumors 168 NK012 SN-38 PEG and PGA phase advanced I/II solid tumors 160 SP1049C Doxorubicin Pluronic L61 phase advanced and F 127 II/III adenocarcino ma of the esophagus and gastro esophageal junction Lipotecan Camptotheci Polymeric Phas Liver and 169 n analog micelle e I/II renal cancer mPEG: methoxy poly-ethylene glycol; PLA: poly-lactic acids; PEG: polyethylene glycol; PAA: poly-acrylic acid; PGA: poly-glutamic acid

NK911

Doxorubicin

PEG and PAA

There are still many challenges for NIR PNMs and their wide application in biomedicine. For NIR fluorescence imaging-guided cancer therapy, it is necessary to optimize the imaging systems and NIR fluorescence probes. There are many fluorescent dyes available in the NIR spectrum, which are usually conjugated with specific cancer-targeting ligands to enable highly selective fluorescence imaging of 30

ACS Paragon Plus Environment

Page 31 of 45

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

cancer. But ICG is the only NIR fluorescence dye approved by the FDA. It is urgent to find some other NIR fluorescence probes that are available clinically, as well as sophisticated imaging instrumentation for detection. There are still some limitations for PNMs in the diagnosis and treatment of cancer. The therapeutic effects of PNM methods such as PTT, PDT, and drug delivery can only be applied to limited types of cancer because of the limited penetration depth and ineffectiveness of therapy on large-mass tumors or metastasized cancer in PDT and PTT. Increasing evidence suggests that the physicochemical properties (e.g., size, charge, and shape) of PNMs play a vital role in controlling their biological performance, such as cellular internalization, blood circulation time, in vivo cancer targeting and biodistribution. Therefore, more efforts are desirable to deeply understand the correlation between them, thereby precisely regulate the characteristics of PNMs to improve their penetration depth and effectiveness. Moreover, combination of photo-induced therapies with other disease treatments like magnetic/sonodynamic therapy which is more potent in treating deeply located tumors will also be one of exciting tides in the future. Before testing in cancer patients, it is still a challenge to verify the metabolic processes related to PNMs, such as the pharmacodynamics and pharmacokinetics in human bodies. Their distribution, fate, translocation, long-term stability, and toxicity should also be addressed before applying them in clinical studies. Although PNMs have promising potential for cancer theranostics, it is still very challenging to develop PNMs commercially. Without abundant economic support, it is difficult to conduct the further toxicity research and synthesis according to current good manufacturing practices in a purely academic environment. Collaboration among highly specialized groups or laboratories is needed to accomplish clinical 31

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 32 of 45

studies of PNMs. The future of advanced probe design for NIR fluorescence imaging mainly depends on multidisciplinary cooperation between chemists, physicists, biologists, and clinicians. With continuous efforts using multidisciplinary approaches, we believe that NIR PNMs will open up new opportunities in cancer theranostics. Acknowledgements This work was supported by the Major State Basic Research Development Program

of

China

(Grant

Nos.

2017YFA0205201,

2014CB744503,

and

2013CB733802), the National Natural Science Foundation of China (NSFC) (Grant Nos. 81422023, 81371596, 51273165, and U1505221), the Science Foundation of Fujian Province (2014Y2004 and 2014J05098), the Program for New Century Excellent Talents in University (NCET-13-0502), and the Fundamental Research Funds for the Central Universities, China (20720150206 and 20720150141). References 1.

Torre, L.A.;Bray, F.;Siegel, R.L.;Ferlay, J.;Lortet-Tieulent, J.,Jemal, A. Global cancer statistics, 2012. CA: a cancer journal for clinicians. 2015, 65(2), 87-108. DOI:10.3322/caac.21262.

2.

Baguley, B.C. Multiple drug resistance mechanisms in cancer. Molecular biotechnology. 2010,

3.

Apetoh, L.;Ghiringhelli, F.;Tesniere, A.;Obeid, M.;Ortiz, C.;Criollo, A.,al., e. Tolllike receptor

46(3), 308-316. DOI:10.1007/s12033-010-9321-2. 4edependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat. Med. 2007, (13), 1050-1059. DOI:10.3410/f.1090489.543761. 4.

D. Peer;J.M. Karp;S. Hong;O.C. Farokhzad;R. Margalit,Langer, R. Nanocarriers as an emerging platform for cancer therapy. Nat. Nanotechnol. 2007,( 2), 751-760. DOI:10.1038/nnano.2007.387

5.

Howell, M.,Valle, J.W. The role of adjuvant chemotherapy and radiotherapy for cholangiocarcinoma. Best practice & research. Clinical gastroenterology. 2015, 29(2), 333-343. DOI:10.1016/j.bpg.2015.03.001.

6.

Bucci, M.K.;Bevan, A.,Roach, M., 3rd. Advances in radiation therapy: conventional to 3D, to IMRT, to 4D, and beyond. CA: a cancer journal for clinicians. 2005, 55(2), 117-134. DOI:10.3322/canjclin.55.2.117.

7.

Gupta, A.A.;Yao, X.;Verma, S.;Mackay, H.;Hopkins, L.;Grp, S.D.S.,Grp, G.C.D.S. Systematic Chemotherapy for Inoperable, Locally Advanced, Recurrent, or Metastatic Uterine Leiomyosarcoma: A Systematic Review. Clin Oncol-Uk. 2013, 25(6), 346-355. DOI:10.1016/j.clon.2012.11.008.

8.

Baxevanis, C.N.;Perez, S.A.,Papamichail, M. Combinatorial treatments including vaccines, chemotherapy and monoclonal antibodies for cancer therapy. Cancer Immunol Immun. 2009, 32

ACS Paragon Plus Environment

Page 33 of 45

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

58(3), 317-324. DOI:10.1007/s00262-008-0576-4. 9.

Funkhouser, J. Reinventing pharma: the theranostic revolution. Curr. Drug Discov. 2002, (2), 17–19. DOI:10.1007/s12274-014-0628-3.

10.

Kelkar, S.S.,Reineke, T.M. Theranostics: Combining Imaging and Therapy. Bioconjugate Chem.

11.

Lu, Y.;Chen, Y.;Gemeinhart, R.A.;Wu, W.,Li, T.L. Developing nanocrystals for cancer

2011, 22(10), 1879-1903. DOI:10.1021/bc200151q. treatment. Nanomedicine-Uk. 2015, 10(16), 2537-2552. DOI:10.2217/nnm.15.73. 12.

Mundra, V.;Li, W.,Mahato, R.I. Nanoparticle-mediated drug delivery for treating melanoma. Nanomedicine-Uk. 2015, 10(16), 2613-2633. DOI:10.2217/nnm.15.111.

13.

Drummond, D.C.;Meyer, O.;Hong, K.L.;Kirpotin, D.B.,Papahadjopoulos, D. Optimizing liposomes for delivery of chemotherapeutic agents to solid tumors. Pharmacol Rev. 1999, 51(4), 691-743. PMID: 10581328.

14.

Byrne, J.D.;Betancourt, T.,Brannon-Peppas, L. Active targeting schemes for nanoparticle systems in cancer therapeutics. Adv Drug Deliver Rev. 2008, 60(15), 1615-626. DOI:10.1016/j.addr.2008.08.005.

15.

Cho, K.J.;Wang, X.;Nie, S.M.;Chen, Z.,Shin, D.M. Therapeutic nanoparticles for drug delivery

16.

Toy, R.;Bauer, L.;Hoimes, C.;Ghaghada, K.B.,Karathanasis, E. Targeted nanotechnology for

in cancer. Clin Cancer Res. 2008, 14(5), 1310-316. DOI:10.1158/1078-0432.CCR-07-1441. cancer imaging. Adv Drug Deliver Rev. 2014, 76,79-97. DOI:10.1016/j.addr.2014.08.002. 17.

Zheng, N.;Dai, W.B.;Du, W.W. A Novel Lanreotide-Encoded Micelle System Targets Paclitaxel to the Tumors with Overexpression of Somatostatin Receptors. Mol Pharmaceut. 2012, 9(5), 1175-1188. DOI:10.1021/mp200464x.

18.

Alam, F.;Naim, M.;Aziz, M.,Yadav, N. Unique roles of nanotechnology in medicine and cancer-II. Indian J Cancer. 2015, 52(1). DOI:10.4103/0019-509X.175591.

19.

Liong, M.;Lu, J.;Kovochich, M.;Xia, T.;Ruehm, S.G.;Nel, A.E.;Tamanoi, F.,Zink, J.I. Multifunctional inorganic nanoparticles for imaging, targeting, and drug delivery. Acs Nano. 2008, 2(5), 889-896. DOI:10.1021/nn800072t.

20.

S.M. Moghimi;A.C. Hunter,Murray, J.C. Long-circulating and target-specificnanoparticles:

21.

M.E. Davis;Z.G. Chen,D.M. Shin. Nanoparticle therapeutics: an emergingtreatment modality

theory to practice. Pharmacol. Rev. 2001, (53), 283–318.WOS:000168846500005. for cancer. Nat. Rev. Drug Discov. 2008, (7), 771–782. DOI:10.1038/nrd2614. 22.

Lammers, T.;Hennink, W.E.,Storm, G. Tumour-targeted nanomedicines: principles and practice. Brit J Cancer. 2008, 99(3), 392-397. DOI:10.1038/sj.bjc.6604483.

23.

Huang, X.H.;Jain, P.K.;El-Sayed, I.H.,El-Sayed, M.A. Plasmonic photothermal therapy (PPTT) using gold nanoparticles. Laser Med Sci. 2008, 23(3), 217-228. DOI:10.1007/s10103-007-0470-x.

24.

Daniel, M.C.,Astruc, D. Gold nanoparticles: Assembly, supramolecular chemistry, quantum-size-related properties, and applications toward biology, catalysis, and nanotechnology. Chem Rev. 2004, 104(1), 293-346. DOI:10.1021/cr030698.

25.

Cao-Milan, R.,Liz-Marzan, L.M. Gold nanoparticle conjugates: recent advances toward clinical applications. Expert Opin Drug Del. 2014, 11(5), 741-752. DOI:10.1517/17425247.2014.891582.

26.

Liang, C.;Diao, S.;Wang, C. Tumor Metastasis Inhibition by Imaging-Guided Photothermal Therapy with Single-Walled Carbon Nanotubes. Adv Mater. 2014, 26(32), 5646-5650. 33

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

DOI:10.1002/adma.201401825. 27.

Yang, K.;Zhang, S.A.;Zhang, G.X.;Sun, X.M.;Lee, S.T.,Liu, Z.A. Graphene in Mice: Ultrahigh In Vivo Tumor Uptake and Efficient Photothermal Therapy. Nano Lett. 2010, 10(9), 3318-3323. DOI:10.1021/nl100996u.

28.

Su, L.T.;Karuturi, S.K.;Luo, J.S. Photon Upconversion in Hetero-nanostructured Photoanodes for Enhanced Near-Infrared Light Harvesting. Adv Mater. 2013, 25(11), 1603-1607. DOI:10.1002/adma.201204353.

29.

Yang, Y.;Aw, J.,Xing, B. Nanostructures for NIR light-controlled therapies. Nanoscale. 2017, 9(11), 3698-3718. DOI:10.1039/C6NR09177F.

30.

Min, Y.;Li, J.;Liu, F.;Yeow, E.K.,Xing, B. Near-infrared light-mediated photoactivation of a platinum antitumor prodrug and simultaneous cellular apoptosis imaging by upconversion-luminescent nanoparticles. Angewandte Chemie. 2014, 53(4), 1012-1016. DOI:10.1002/anie.201308834.

31.

Park, J.H.;von Maltzahn, G.;Ruoslahti, E.;Bhatia, S.N.,Sailor, M.J. Micellar hybrid nanoparticles for simultaneous magnetofluorescent imaging and drug delivery. Angewandte Chemie. 2008, 47(38), 7284-7288. DOI:10.1002/ange.200801810.

32.

Nahrendorf, M.;Zhang, H.;Hembrador, S. Nanoparticle PET-CT imaging of macrophages in inflammatory atherosclerosis. Circulation. 2008, 117(3), 379-387. DOI:10.1161/circulationaha.107.741181.

33.

Shokeen, M.;Fettig, N.M.,Rossin, R. Synthesis, in vitro and in vivo evaluation of radiolabeled nanoparticles. The quarterly journal of nuclear medicine and molecular imaging : official publication of the Italian Association of Nuclear Medicine. 2008, 52(3), 267-277. PMID: 18475250.

34.

Jain, T.K.;Roy, I.;De, T.K.,Maitra, A. Nanometer silica particles encapsulating active compounds: A novel ceramic drug carrier. J Am Chem Soc. 1998, 120(43), 11092-11095. DOI:10.1021/ja973849x.

35.

Esmaeili, F.;Ghahremani, M.H.;Esmaeili, B.;Khoshayand, M.R.;Atyabi, F.,Dinarvand, R. PLGA nanoparticles of different surface properties: preparation and evaluation of their body distribution. International journal of pharmaceutics. 2008, 349(2), 249-255. DOI:10.1016/j.ijpharm.2007.07.038.

36.

Lukyanov, A.N.;Gao, Z.;Mazzola, L.,Torchilin, V.P. Polyethylene glycol-diacyllipid micelles demonstrate increased acculumation in subcutaneous tumors in mice. Pharmaceutical research. 2002, 19(10), 1424-1429. DOI:10.1023/A:1020488012264.

37.

Maeda, H.;Wu, J.;Sawa, T.;Matsumura, Y.,Hori, K. Tumor vascular permeability and the EPR effect in macromolecular therapeutics: a review. Journal of controlled release : official journal of the Controlled Release Society. 2000, 65(2), 271-284. DOI:10.1016/s0168-3659(99)00248-5.

38.

Zhu, L.;Perche, F.;Wang, T.,Torchilin, V.P. Matrix metalloproteinase 2-sensitive multifunctional polymeric micelles for tumor-specific co-delivery of siRNA and hydrophobic drugs. Biomaterials. 2014, 35(13), 4213-4222. DOI:10.1016/j.biomaterials.2014.01.060.

39.

Howell, M.;Mallela, J.;Wang, C.;Ravi, S.;Dixit, S.;Garapati, U.,Mohapatra, S. Manganese-loaded lipid-micellar theranostics for simultaneous drug and gene delivery to lungs. Journal of controlled release : official journal of the Controlled Release Society. 2013, 167(2), 210-218. DOI:10.1016/j.jconrel.2013.01.029. 34

ACS Paragon Plus Environment

Page 34 of 45

Page 35 of 45

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

40.

Tan, C.;Wang, Y.,Fan, W. Exploring polymeric micelles for improved delivery of anticancer agents: recent developments in preclinical studies. Pharmaceutics. 2013, 5(1), 201-219. DOI:10.1016/j.addr.2010.05.008.

41.

Duan, J.;Mansour, H.M.;Zhang, Y.;Deng, X.;Chen, Y.;Wang, J.;Pan, Y.,Zhao, J. Reversion of multidrug resistance by co-encapsulation of doxorubicin and curcumin in chitosan/poly(butyl cyanoacrylate) nanoparticles. International journal of pharmaceutics. 2012, 426(2), 193-201. DOI:10.1016/j.ijpharm.2012.01.020.

42.

Opanasopit, P.;Yokoyama, M.;Watanabe, M.;Kawano, K.;Maitani, Y.,Okano, T. Block copolymer design for camptothecin incorporation into polymeric micelles for passive tumor targeting. Pharmaceutical research. 2004, 21(11), 2001-2008. DOI:10.1023/b:pham.0000048190.53439.eb.

43.

Gao J;Blanco E;Bey EA;Dong Y;Weinberg BD;Sutton DM,al, e. β-Lapachone-containing PEG–PLA polymer micelles as novel nanotherapeutics against NQO1-overexpressing tumor cells. Journal of controlled release : official journal of the Controlled Release Society. 2007, (122), 365–374. DOI:10.1016/j.jconrel.2007.04.014.

44.

Qin, L.;Zhang, F.;Lu, X. Polymeric micelles for enhanced lymphatic drug delivery to treat metastatic tumors. Journal of controlled release : official journal of the Controlled Release Society. 2013, 171(2), 133-142. DOI:10.1016/j.jconrel.2013.07.005.

45.

Zheng, N.;Dai, W.;Du, W. A novel lanreotide-encoded micelle system targets paclitaxel to the tumors with overexpression of somatostatin receptors. Mol Pharm. 2012, 9(5), 1175-1188. DOI:10.1021/mp200464x.

46.

Jones, M.,Leroux, J. Polymeric micelles - a new generation of colloidal drug carriers. European journal of pharmaceutics and biopharmaceutics : official journal of Arbeitsgemeinschaft fur Pharmazeutische Verfahrenstechnik e.V. 1999, 48(2), 101-111. DOI:10.1016/s0939-6411(99)00039-9.

47.

Kim, S.C.;Kim, D.W.;Shim, Y.H.;Bang, J.S.;Oh, H.S.;Kim, S.W.,Seo, M.H. In vivo evaluation of polymeric micellar paclitaxel formulation: toxicity and efficacy. Journal of Controlled Release. 2001, 72(3), 191-202. DOI:10.1016/s0168-3659(01)00275-9.

48.

Le Garrec, D.;Taillefer, J.;Van Lier, J.E.;Lenaerts, V.,Leroux, J.C. Optimizing pH-responsive polymeric micelles for drug delivery in a cancer photodynamic therapy model. J Drug Target. 2002, 10(5), 429-437. DOI:10.1080/1061186021000001887.

49.

Torchilin, V.P.;Shtilman, M.I.;Trubetskoy, V.S.;Whiteman, K.,Milstein, A.M. Amphiphilic vinyl polymers effectively prolong liposome circulation time in vivo. Biochimica et biophysica acta. 1994, 1195(1), 181-184. DOI:10.1016/0005-2736(94)90025-6.

50.

Benahmed, A.;Ranger, M.,Leroux, J.C. Novel polymeric micelles based on the amphiphilic diblock copolymer poly(N-vinyl-2-pyrrolidone)-block-poly(D,L-lactide). Pharmaceutical research. 2001, 18(3), 323-328. DOI:10.1023/A:1011054930439.

51.

Luppi, B.;Orienti, I.;Bigucci, F.;Cerchiara, T.;Zuccari, G.;Fazzi, S.,Zecchi, V. Poly(Vinylalcohol-Co-Vinyloleate) for the Preparation of Micelles Enhancing Retinyl Palmitate Transcutaneous Permeation. Drug Deliv. 2002, 9(3), 147-152. DOI:10.1080/10426500290095647.

52.

Kwon, G.S. Polymeric micelles for delivery of poorly water-soluble compounds. Crit Rev Ther Drug. 2003, 20(5), 357-403. DOI:10.1615/critrevtherdrugcarriersyst.v20.i5.20.

53.

Kumar, N.;Langer, R.S.,Domb, A.J. Polyanhydrides: an overview. Adv Drug Deliver Rev. 2002, 35

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

54(7), 889-910. DOI:10.1016/s0169-409x(02)00050-9. 54.

Ojugo, A.S.E.;McSheehy, P.M.J.;McIntyre, D.J.O.;McCoy, C.;Stubbs, M.;Leach, M.O.;Judson, I.R.,Griffiths, J.R. Measurement of the extracellular pH of solid tumours in mice by magnetic resonance spectroscopy: a comparison of exogenous F-19 and P-31 probes. Nmr Biomed. 1999, 12(8), 495-504. DOI:10.1002/(sici)1099-1492(199912).

55.

Koo, O.M.;Rubinstein, I.,Onyuksel, H. Role of nanotechnology in targeted drug delivery and imaging: a concise review. Nanomed-Nanotechnol. 2005, 1(3), 193-212. DOI:10.1016/j.nano.2005.06.004.

56.

Kataoka, K.;Kwon, G.S.;Yokoyama, M.;Okano, T.,Sakurai, Y. Block-Copolymer Micelles as Vehicles for Drug Delivery. Journal of Controlled Release. 1993, 24(3), 119-132. DOI:10.1016/0168-3659(93)90172-2.

57.

Gao, Z.S.,Eisenberg, A. A Model of Micellization for Block-Copolymers in Solutions. Macromolecules. 1993, 26(26), 7353-7360. DOI:10.1021/ma00078a035.

58.

Schmolka, I.R. A review of block polymer surfactants. J. Am. Oil Chem. Soc. 1977, 54,110-116. DOI:10.1007/bf02894385.

59.

JinqiangWang;Weiwei Mao;Jianbin Tang;Meihua Sui;Weilin Sun;Honggang Cu;Dong Xu,Shen, Y. The Role of Micelle Size in Tumor Accumulation, Penetration, and Treatment. ACS nano. 2015, 9(7), 7195-7206. DOI:10.1021/acsnano.5b02017.

60.

Zhang, Y.,Zhuo, R.X. Synthesis and in vitro drug release behavior of amphiphilic triblock copolymer nanoparticles based on poly (ethylene glycol) and polycaprolactone. Biomaterials. 2005, 26(33), 6736-6742. DOI:10.1016/j.biomaterials.2005.03.045.

61.

Kumar, R.;Ohulchanskyy, T.Y.;Roy, I.;Gupta, S.K.;Borek, C.;Thompson, M.E.,Prasad, P.N. Near-infrared phosphorescent polymeric nanomicelles: efficient optical probes for tumor imaging and detection. ACS applied materials & interfaces. 2009, 1(7), 1474-1481. DOI:10.1021/am9001293.

62.

Kockler, K.B.;Haehnel, A.P.;Junkers, T.,Barner-Kowollik, C. Determining Free-Radical Propagation Rate Coefficients with High-Frequency Lasers: Current Status and Future Perspectives. Macromolecular rapid communications. 2016, 37(2), 123-134. DOI:10.1002/marc.201500503.

63.

Luo, S.L.;Zhang, E.L.;Su, Y.P.;Cheng, T.M.,Shi, C.M. A review of NIR dyes in cancer targeting and imaging. Biomaterials. 2011, 32(29), 7127-7138. DOI:10.1016/j.biomaterials.2011.06.024.

64.

van Dam, G.M.;Themelis, G.;Crane, L.M. Intraoperative tumor-specific fluorescence imaging in ovarian cancer by folate receptor-alpha targeting: first in-human results. Nature medicine. 2011, 17(10), 1315-1319. DOI:10.1038/nm.2472.

65.

Wu, L.;Fang, S.;Shi, S.;Deng, J.;Liu, B.,Cai, L. Hybrid polypeptide micelles loading indocyanine green for tumor imaging and photothermal effect study. Biomacromolecules. 2013, 14(9), 3027-3033. DOI:10.1021/bm400839b.

66.

Shi, C.;Zhang, C.;Su, Y.,Cheng, T. Cyanine dyes in optical imaging of tumours. The Lancet. Oncology. 2010, 11(9), 815-816. DOI:10.1016/s1470-2045(10)70196-0.

67.

Zhang, C.;Wang, S.;Xiao, J.;Tan, X.;Zhu, Y.;Su, Y.;Cheng, T.,Shi, C. Sentinel lymph node mapping by a near-infrared fluorescent heptamethine dye. Biomaterials. 2010, 31(7), 1911-1917. DOI:10.1016/j.biomaterials.2009.11.061.

68.

Pais-Silva, C.;de Melo-Diogo, D.,Correia, I.J. IR780-loaded TPGS-TOS micelles for breast 36

ACS Paragon Plus Environment

Page 36 of 45

Page 37 of 45

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

cancer photodynamic therapy. European Journal of Pharmaceutics and Biopharmaceutics. 2017, 113,108-117. DOI:10.1016/j.ejpb.2017.01.002. 69.

Palma, A.;Tasior, M.;Frimannsson, D.O.;Vu, T.T.;Meallet-Renault, R.,O'Shea, D.F. New On-Bead Near-Infrared Fluorophores and Fluorescent Sensor Constructs. Org Lett. 2009, 11(16), 3638-3641. DOI:10.1021/ol901413u.

70.

Liu, L.;Fu, L.Y.;Jing, T.T.;Ruan, Z.,Yang, L.F. pH-Triggered Polypeptides Nanoparticles for Efficient BODIPY Imaging-Guided Near Infrared Photodynamic Therapy. ACS applied materials & interfaces. 2016, 8(14), 8980-8990. DOI:10.1021/acsami.6b01320.

71.

Fan, Q.L.;Cheng, K.;Yang, Z. Perylene-Diimide-Based Nanoparticles as Highly Efficient Photoacoustic Agents for Deep Brain Tumor Imaging in Living Mice. Adv Mater. 2015, 27(5), 843-847. DOI:10.1002/adma.201570028.

72.

Jiang, Y.;Upputuri, P.K.;Xie, C.;Lyu, Y.;Zhang, L.;Xiong, Q.;Pramanik, M.,Pu, K. Broadband Absorbing Semiconducting Polymer Nanoparticles for Photoacoustic Imaging in Second Near-Infrared Window. Nano letters. 2017, 17(8), 4964-4969. DOI:10.1021/acs.nanolett.7b02106.

73.

Yang, Z.;Tian, R.;Wu, J. Impact of Semiconducting Perylene Diimide Nanoparticle Size on Lymph Node Mapping and Cancer Imaging. ACS nano. 2017, 11(4), 4247-255. DOI:10.1021/acsnano.7b01261.

74.

Su, J.L.;Wang, B.;Wilson, K.E.;Bayer, C.L.;Chen, Y.S.;Kim, S.;Homan, K.A.,Emelianov, S.Y. Advances in Clinical and Biomedical Applications of Photoacoustic Imaging. Expert opinion on medical diagnostics. 2010, 4(6), 497-10. DOI:10.1517/17530059.2010.529127.

75.

Liang, X.L.;Fang, L.;Li, X.D.;Zhang, X.,Wang, F. Activatable near infrared dye conjugated hyaluronic acid based nanoparticles as a targeted theranostic agent for enhanced fluorescence/CT/photoacoustic imaging guided photothermal therapy. Biomaterials. 2017, 132,72-84. DOI:10.1016/j.biomaterials.2017.04.006.

76.

Scudiero, O.;Nigro, E.;Monaco, M.L. New synthetic AICAR derivatives with enhanced AMPK and ACC activation. Journal of enzyme inhibition and medicinal chemistry. 2016, 31(5), 748-753. DOI:10.3109/14756366.2015.1063622.

77.

Mura, S.;Nicolas, J.,Couvreur, P. Stimuli-responsive nanocarriers for drug delivery. Nature materials. 2013, 12(11), 991-1003. DOI:10.1038/nmat3776.

78.

J.H. Kim;Y. Li;Kim, M.S.;S.W. Kang;J.H. Jeong,D.S. Lee. Synthesis and evaluationof biotin-conjugated pH-responsive polymeric micelles as drug carriers. Int. J.Pharm. 2012, (427), 435–442. DOI:10.1016/j.ijpharm.2012.01.034.

79.

Chacko, R.T.;J. Ventura;J. Zhuang,Thayumanavan, S. Polymer nanogels: a ver-satile nanoscopic drug delivery platform. Adv. Drug Deliv. Rev. 2012, (64), 836–851. DOI:10.1016/j.addr.2012.02.002.

80.

Cao, C.;Wang, X.;Cai, Y. Targeted in vivo imaging of microscopic tumors with ferritin-based nanoprobes across biological barriers. Adv Mater. 2014, 26(16), 2566-2571. DOI:10.1002/adma.201304544.

81.

Karimi, M.;Sahandi Zangabad, P.;Ghasemi, A. Temperature-Responsive Smart Nanocarriers for Delivery Of Therapeutic Agents: Applications and Recent Advances. ACS applied materials & interfaces. 2016, 8(33), 21107-21133. DOI:10.1021/acsami.6b00371.

82.

Lieber, C.M. Nanoscale science and technology: Building a big future from small things. Mrs Bull. 2003, 28(7), 486-491. DOI:10.1557/mrs2003.144. 37

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

83.

Fomina, N.;Sankaranarayanan, J.,Almutairi, A. Photochemical mechanisms of light-triggered release from nanocarriers. Adv Drug Deliver Rev. 2012, 64(11), 1005-1020. DOI:10.1016/j.addr.2012.02.006.

84.

Gohy, J.F.,Zhao, Y. Photo-responsive block copolymer micelles: design and behavior. Chem

85.

Karimi, M.;Zangabad, P.S.;Baghaee-Ravari, S.;Ghazadeh, M.;Mirshekari, H.,Hamblin, M.R.

Soc Rev. 2013, 42(17), 7117-7129. DOI:10.1039/c3cs35469e. Smart Nanostructures for Cargo Delivery: Uncaging and Activating by Light. J Am Chem Soc. 2017, 139(13), 4584-4610. DOI:10.1021/jacs.6b08313. 86.

Sahandi Zangabad, P.;Karimi, M.;Mehdizadeh, F. Nanocaged platforms: modification, drug delivery and nanotoxicity. Opening synthetic cages to release the tiger. Nanoscale. 2017, 9(4), 1356-1392. DOI:10.1039/c6nr07315h.

87.

Karimi, M.;Avci, P.;Ahi, M.;Gazori, T.;Hamblin, M.R.,Naderi-Manesh, H. Evaluation of Chitosan-Tripolyphosphate Nanoparticles as a p-shRNA Delivery Vector: Formulation, Optimization and Cellular Uptake Study. Journal of nanopharmaceutics and drug delivery. 2013, 1(3), 266-278. DOI:10.1166/jnd.2013.1027.

88.

Karimi, M.;Bahrami, S.;Ravari, S.B. Albumin nanostructures as advanced drug delivery systems. Expert Opin Drug Deliv. 2016, 13(11), 1609-1623. DOI:10.1080/17425247.2016.1193149.

89.

Petros, R.A.,DeSimone, J.M. Strategies in the design of nanoparticles for therapeutic

90.

Paleos, C.M.;Tsiourvas, D.;Sideratou, Z.,Tziveleka, L.A. Drug delivery using multifunctional

applications. Nat Rev Drug Discov. 2010, 9(8), 615-627. DOI:10.1038/nrd2591. dendrimers and hyperbranched polymers. Expert Opin Drug Del. 2010, 7(12), 1387-1398. DOI:10.1517/17425247.2010.534981. 91.

Yokoyama, M.;Kwon, G.S.;Okano, T.;Sakurai, Y.;Seto, T.,Kataoka, K. Preparation of Micelle-Forming Polymer Drug Conjugates. Bioconjugate Chem. 1992, 3(4), 295-301. DOI:10.1021/bc00016a007.

92.

Fernandez AM;Van DK;Dasnois L;Lebtah;i K Dubois V,Lobl TJ, e.a. N-succinyl-(β-alanyl-L-leucyl-l-alanyl-l-leucyl) doxorubicin: an extracellularly tumor-activated prodrug devoid of intravenous acute toxicity. J Med Chem 2001, (44), 3750-3753. DOI:10.1021/jm0108754.

93.

Lipinski, C.A.;Lombardo, F.;Dominy, B.W.,Feeney, P.J. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliver Rev. 2001, 46(3), 3-26. DOI:10.1016/j.addr.2012.09.019.

94.

Savic, R.;Eisenberg, A.,Maysinger, D. Block copolymer micelles as delivery vehicles of hydrophobic drugs: Micelle-cell interactions. J Drug Target. 2006, 14(6), 343-355. DOI:10.1080/10611860600874538.

95.

Kedar, U.;Phutane, P.;Shidhaye, S.,Kadam, V. Advances in polymeric micelles for drug delivery and tumor targeting. Nanomed-Nanotechnol. 2010, 6(6), 714-729. DOI:10.1016/j.nano.2010.05.005.

96.

Lin, Q.N.;Huang, Q.;Li, C.Y.;Bao, C.Y.;Liu, Z.Z.;Li, F.Y.,Zhu, L.Y. Anticancer Drug Release from a Mesoporous Silica Based Nanophotocage Regulated by Either a One- or Two-Photon Process. J Am Chem Soc. 2010, 132(31), 10645-10647. DOI:10.1021/ja103415t.

97.

Yan, B.;Boyer, J.C.;Habault, D.;Branda, N.R.,Zhao, Y. Near Infrared Light Triggered Release of Biomacromolecules from Hydrogels Loaded with Upconversion Nanoparticles. J Am Chem 38

ACS Paragon Plus Environment

Page 38 of 45

Page 39 of 45

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

Soc. 2012, 134(40), 16558-16561. DOI:10.1021/ja308876j. 98.

Mal, N.K.;Fujiwara, M.,Tanaka, Y. Photocontrolled reversible release of guest molecules from coumarin-modified mesoporous silica. Nature. 2003, 421(6921), 350-353. DOI:10.1038/nature01362.

99.

Murthy, N.;Xu, M.C.;Schuck, S.;Kunisawa, J.;Shastri, N.,Frechet, J.M.J. A macromolecular delivery vehicle for protein-based vaccines: Acid-degradable protein-loaded microgels. P Natl Acad Sci USA. 2003, 100(9), 4995-5000. DOI:10.1073/pnas.0930644100.

100.

Bulmus, V.;Chan, Y.;Nguyen, Q.,Tran, H.L. Synthesis and characterization of degradable p(HEMA) microgels: Use of acid-labile crosslinkers. Macromol Biosci. 2007, 7(4), 446-455. DOI:10.1002/mabi.200600258.

101.

Yoshikawa, H.Y.;Rossetti, F.F.;Kaufmann, S. Quantitative Evaluation of Mechanosensing of Cells on Dynamically Tunable Hydrogels. J Am Chem Soc. 2011, 133(5), 1367-1374. DOI:10.1021/ja1060615.

102.

Li, Y.;Lokitz, B.S.,McCormick, C.L. Thermally responsive vesicles and their structural "locking" through polyelectrolyte complex formation. Angew Chem Int Edit. 2006, 45(35), 5792-5795. DOI:10.1002/ange.200602168.

103.

Pasparakis, G.,Alexander, C. Sweet talking double hydrophilic block copolymer vesicles. Angew Chem Int Edit. 2008, 47(26), 4847-4850. DOI:10.1002/anie.200801098.

104.

Wang, C.;Stewart, R.J.,Kopecek, J. Hybrid hydrogels assembled from synthetic polymers and coiled-coil protein domains. Nature. 1999, 397(6718), 417-420. DOI:10.1038/17092.

105.

Xiong, M.H.;Bao, Y.;Yang, X.Z.;Wang, Y.C.;Sun, B.L.,Wang, J. Lipase-Sensitive Polymeric Triple-Layered Nanogel for "On-Demand" Drug Delivery. J Am Chem Soc. 2012, 134(9), 4355-4362. DOI:10.1021/ja211279u.

106.

Miyata, T.;Asami, N.,Uragami, T. A reversibly antigen-responsive hydrogel. Nature. 1999, 399(6738), 766-769. DOI:10.1038/21619.

107.

Raghupathi, K.R.;Azagarsamy, M.A.,Thayumanavan, S. Guest-Release Control in Enzyme-Sensitive, Amphiphilic-Dendrimer-Based Nanoparticles through Photochemical Crosslinking. Chem-Eur J. 2011, 17(42), 11752-11760. DOI:10.1002/chem.201101066.

108.

Dunn, S.S.;Tian, S.M.;Blake, S. Reductively Responsive siRNA-Conjugated Hydrogel Nanoparticles for Gene Silencing. J Am Chem Soc. 2012, 134(17), 7423-7430. DOI:10.1021/ja300174v.

109.

Verheyen, E.;van der Wal, S.;Deschout, H.;Braeckmans, K.;de Smedt, S.;Barendregt, A.;Hennink, W.E.,van Nostrum, C.F. Protein macromonomers containing reduction-sensitive linkers for covalent immobilization and glutathione triggered release from dextran hydrogels. Journal of Controlled Release. 2011, 156(3), 329-336. DOI:10.1016/j.jconrel.2011.08.040.

110.

Weissleder, R. A clearer vision for in vivo imaging. Nat Biotechnol. 2001, 19(4), 316-317. DOI:10.1038/86684.

111.

Lee, S.E.;Liu, G.L.;Kim, F.,Lee, L.P. Remote optical switch for localized and selective control of gene interference. Nano Lett. 2009, 9(2), 562-570. DOI:10.1021/nl802689k.

112.

Yang, Y.M.;Liu, F.;Liu, X.G.,Xing, B.G. NIR light controlled photorelease of siRNA and its targeted intracellular delivery based on upconversion nanoparticles. Nanoscale. 2013, 5(1), 231-238. DOI:10.1039/c2nr32835f.

113.

Zhang, H.;Xiong, L.F.;Liao, X.J.,Huang, K. Controlled-Release System of Small Molecules Triggered by the Photothermal Effect of Polypyrrole. Macromol Rapid Comm. 2016, 37(2), 39

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

149-154. DOI:10.1002/marc.201500523. 114.

Li, H.;Li, J.J.;Ke, W.D.,Ge, Z.S. A Near-Infrared Photothermal Effect-Responsive Drug Delivery System Based on Indocyanine Green and Doxorubicin-Loaded Polymeric Micelles Mediated by Reversible Diels-Alder Reaction. Macromol Rapid Comm. 2015, 36(20), 1841-1849. DOI:10.1002/marc.201500337.

115.

Fu, L.Y.;Sun, C.Y.,Yan, L.F. Galactose Targeted pH-Responsive Copolymer Conjugated with Near Infrared Fluorescence Probe for Imaging of Intelligent Drug Delivery. Acs Appl Mater Inter. 2015, 7(3), 2104-2115. DOI:10.1016/j.jconrel.2015.05.120.

116.

Akagi, D.;Oba, M.;Koyama, H.;Nishiyama, N.;Fukushima, S.;Miyata, T.;Nagawa, H.,Kataoka, K. Biocompatible micellar nanovectors achieve efficient gene transfer to vascular lesions without cytotoxicity and thrombus formation. Gene Ther. 2007, 14(13), 1029-1038. DOI:10.1038/sj.gt.3302945.

117.

Hacein-Bey-Abina, S.;Von Kalle, C.;Schmidt, M. LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1. Science. 2003, 302(5644), 415-419. DOI:10.1126/science.1088547.

118.

Aiuti, A.;Slavin, S.;Aker, M. Correction of ADA-SCID by stem cell gene therapy combined with nonmyeloablative conditioning. Science. 2002, 296(5577), 2410-2413. DOI:10.1126/science.1070104.

119.

Cavazzana-Calvo, M.;Hacein-Bey, S.;Basile, C.D. Gene therapy of human severe combined immunodeficiency (SCID)-X1 disease. Science. 2000, 288(5466), 669-672. DOI:10.1126/science.288.5466.669.

120.

Glover, D.J.;Lipps, H.J.,Jans, D.A. Towards safe, non-viral therapeutic gene expression in

121.

Merdan, T.;Kopecek, J.,Kissel, T. Prospects for cationic polymers in gene and oligonucleotide

humans. Nat Rev Genet. 2005, 6(4), 299-U229. DOI:10.1038/nrg1577. therapy against cancer. Adv Drug Deliver Rev. 2002, 54(5), 715-758. DOI:10.1016/S0169-409X(02)00046-7. 122.

Harada-Shiba, M.;Yamauchi, K.;Harada, A.;Takamisawa, I.;Shimokado, K.,Kataoka, K. Polyion complex micelles as vectors in gene therapy - pharmacokinetics and in vivo gene transfer. Gene Ther. 2002, 9(6), 407-414. DOI:10.1038/sj.gt.3301665.

123.

Yousefpour Marzbali, M.,Yari Khosroushahi, A. Polymeric micelles as mighty nanocarriers for cancer gene therapy: a review. Cancer chemotherapy and pharmacology. 2017, 79(4), 637-649. DOI:10.1007/s00280-017-3273-1.

124.

Tu, Z.;Ma, Y.;Akers, W.;Achilefu, S.,Gu, Y. Therapeutic effect of the treatment for colorectal cancer with adenoviral vectors mediated estrogen receptor beta gene therapy combined with thermotherapy. Journal of cancer research and clinical oncology. 2014, 140(4), 623-632. DOI:10.1007/s00432-014-1611-9.

125.

Aliabadi, H.M.,Lavasanifar, A. Polymeric micelles for drug delivery. Expert Opin Drug Deliv.

126.

Kataoka, K.;Harada, A.,Nagasaki, Y. Block copolymer micelles for drug delivery: design,

2006, 3(1), 139-162. DOI:10.1517/17425247.3.1.139. characterization and biological significance. Adv Drug Deliver Rev. 2001, 47(1), 113-131. DOI:10.1016/j.addr.2012.09.013. 127.

Nishiyama, N.,Kataoka, K. Current state, achievements, and future prospects of polymeric micelles as nanocarriers for drug and gene delivery. Pharmacol Therapeut. 2006, 112(3), 630-648. DOI:10.1016/j.pharmthera.2006.05.006. 40

ACS Paragon Plus Environment

Page 40 of 45

Page 41 of 45

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

128.

Osada, K.;Christie, R.J.,Kataoka, K. Polymeric micelles from poly(ethylene glycol)-poly(amino acid) block copolymer for drug and gene delivery. J R Soc Interface. 2009, 6,325-339. DOI:10.1098/rsif.2008.0547.focus.

129.

K., G. Enhanced permeability and retention (EPR) effect for anticancer nanomedicine drug targeting. Methods Mol Biol. 2010, (624), 25-37. DOI:10.1007/978-1-60761-609-23.

130.

Matsumura, Y.,Maeda, H. A New Concept for Macromolecular Therapeutics in Cancer-Chemotherapy - Mechanism of Tumoritropic Accumulation of Proteins and the Antitumor Agent Smancs. Cancer Res. 1986, 46(12), 6387-6392. PMID:2946403.

131.

Wang, R.S.E.;Niu, Y.H.;Wu, H.F.;Hu, Y.G.,Cai, J.F. Development of NGR-Based Anti-Cancer Agents for Targeted Therapeutics and Imaging. Anti-Cancer Agent Me. 2012, 12(1), 76-86. DOI:10.2174/187152012798764714.

132.

F, P.;Brignole C;Marimpietri D;Cilli M;Gambini C,Ribatti D, e.a. Vascular damage and anti-angiogenic effects of tumor vesseltargeted liposomal chemotherapy. Cancer Res. 2003, (63), 7400–7409. DOI:10.1016/S0006-8993(03)02939-1.

133.

Han, H.S.;Choi, K.Y.;Ko, H.;Jeon, J.;Saravanakumar, G.;Suh, Y.D.;Lee, D.S.,Park, J.H. Bioreducible core-crosslinked hyaluronic acid micelle for targeted cancer therapy. Journal of controlled release : official journal of the Controlled Release Society. 2015, 200,158-166. DOI:10.1016/j.jconrel.2014.12.032.

134.

Upadhyay, K.K.;Bhatt, A.N.;Castro, E. In vitro and in vivo evaluation of docetaxel loaded biodegradable polymersomes. Macromol Biosci. 2010, 10(5), 503-512. DOI:10.1002/mabi.200900415.

135.

Upadhyay, K.K.;Bhatt, A.N.;Mishra, A.K. The intracellular drug delivery and anti tumor activity of doxorubicin loaded poly(gamma-benzyl L-glutamate)-b-hyaluronan polymersomes. Biomaterials. 2010, 31(10), 2882-2892. DOI:10.1016/j.biomaterials.2009.12.043.

136.

Jin, Q.;Zhu, W.;Jiang, D. Ultra-small iron-gallic acid coordination polymer nanoparticles for chelator-free labeling of 64Cu and multimodal imaging-guided photothermal therapy. Nanoscale. 2017, 9(34), 12609-12617. DOI:10.1039/c7nr03086j.

137.

Chen, Y.;Cheng, L.;Dong, Z.;Chao, Y.;Lei, H.;Zhao, H.;Wang, J.,Liu, Z. Degradable Vanadium Disulfide Nanostructures with Unique Optical and Magnetic Functions for Cancer Theranostics. Angewandte Chemie. 2017, 56(42), 12991-12996. DOI:10.1002/ange.201707128.

138.

Wang, J.;Tao, W.;Chen, X.;Farokhzad, O.C.,Liu, G. Emerging Advances in Nanotheranostics with Intelligent Bioresponsive Systems. Theranostics. 2017, 7(16), 3915-3919. DOI:10.7150/thno.21317.

139.

Wei, J.P.;Chen, X.L.;Wang, X.Y.;Li, J.C.;Shi, S.G.;Liu, G.,Zheng, N.F. Polyethylene glycol phospholipids encapsulated silicon 2,3-naphthalocyanine dihydroxide nanoparticles (SiNcOH-DSPE-PEG(NH2) NPs) for single NIR laser induced cancer combination therapy. Chinese Chem Lett. 2017, 28(6), 1290-1299. DOI:10.1016/j.cclet.2017.01.007.

140.

Chu, C.;Lin, H.;Liu, H. Tumor Microenvironment-Triggered Supramolecular System as an In Situ Nanotheranostic Generator for Cancer Phototherapy. Advanced materials. 2017, 29(23), DOI:10.1002/adma.201770164.

141.

Ren, E.;Wang, J.,Liu, G. Cell-surface cascaded landing location for nanotheranostics. Chinese Chem Lett. 2017, 28(9), 1799-1800. DOI:10.1016/j.cclet.2017.07.015.

142.

Guo, M.;Mao, H.J.;Li, Y.L. Dual imaging-guided photothermal/photodynamic therapy using 41

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

micelles. Biomaterials. 2014, 35(16), 4656-4666. DOI:10.1016/j.biomaterials.2014.02.018. 143.

Chao, Y.;Wang, G.;Liang, C. Rhenium-188 Labeled Tungsten Disulfide Nanoflakes for Self-Sensitized, Near-Infrared Enhanced Radioisotope Therapy. Small. 2016, 12(29), 3967-3975. DOI:10.1002/smll.201601375.

144.

Zhang, J.F.;Yang, C.X.;Zhang, R. Biocompatible D-A Semiconducting Polymer Nanoparticle with Light-Harvesting Unit for Highly Effective Photoacoustic Imaging Guided Photothermal Therapy. Adv Funct Mater. 2017, 27(13), DOI:10.1002/adfm.201605094.

145.

Huang, C.;Chu, C.C.;Wang, X.Y. Ultra-high loading of sinoporphyrin sodium in ferritin for single-wave motivated photothermal and photodynamic co-therapy. Biomater Sci-Uk. 2017, 5(8), 1512-1516. DOI:10.1039/C7BM00302A.

146.

Guo, Z.Q.;Park, S.;Yoon, J.,Shin, I. Recent progress in the development of near-infrared fluorescent probes for bioimaging applications. Chem Soc Rev. 2014, 43(1), 16-29. DOI:10.1002/chin.201415298.

147.

Yuan, L.;Lin, W.Y.;Zheng, K.B.;He, L.W.,Huang, W.M. Far-red to near infrared analyte-responsive fluorescent probes based on organic fluorophore platforms for fluorescence imaging. Chem Soc Rev. 2013, 42(2), 622-661. DOI:10.1002/chin.201320264.

148.

Sheng, Z.H.;Hu, D.H.;Xue, M.M.;He, M.;Gong, P.,Cai, L.T. Indocyanine Green Nanoparticles for Theranostic Applications. Nano-Micro Lett. 2013, 5(3), 145-150. DOI:10.1039/c2cs35313j.

149.

May, J.P.,Li, S.D. Hyperthermia-induced drug targeting. Expert Opin Drug Deliv. 2013, 10(4), 511-527. DOI:10.1517/17425247.2013.758631.

150.

Huo, D.;He, J.;Li, H.;Huang, A.J.;Zhao, H.Y.;Ding, Y.;Zhou, Z.Y.,Hu, Y. X-ray CT guided fault-free photothermal ablation of metastatic lymph nodes with ultrafine HER-2 targeting W18O49 nanoparticles. Biomaterials. 2014, 35(33), 9155-9166. DOI:10.1016/j.biomaterials.2014.07.034.

151.

Jiang, B.-P.;Zhang, L.;Zhu, Y.;Shen, X.-C.;Ji, S.-C.;Tan, X.-Y.;Cheng, L.,Liang, H. Water-soluble hyaluronic acid–hybridized polyaniline nanoparticles for effectively targeted photothermal therapy. J. Mater. Chem. B. 2015, 3(18), 3767-3776. DOI:10.1039/c4tb01738b.

152.

Melancon, M.P.;Zhou, M.,Li, C. Cancer theranostics with near-infrared light-activatable multimodal nanoparticles. Accounts of chemical research. 2011, 44(10), 947-956. DOI:10.1021/ar200022e .

153.

Fazal, S.;Jayasree, A.;Sasidharan, S.;Koyakutty, M.;Nair, S.V.,Menon, D. Green synthesis of anisotropic gold nanoparticles for photothermal therapy of cancer. ACS applied materials & interfaces. 2014, 6(11), 8080-8089. DOI:10.1021/am500302t .

154.

Yuan, A.;Qiu, X.;Tang, X.;Liu, W.;Wu, J.,Hu, Y. Self-assembled PEG-IR-780-C13 micelle as a targeting, safe and highly-effective photothermal agent for in vivo imaging and cancer therapy. Biomaterials. 2015, 51(184-193. DOI:10.1016/j.biomaterials.2015.01.069.

155.

Wilson, B.C.,Patterson, M.S. The physics, biophysics and technology of photodynamic

156.

Wan, Z.;Mao, H.;Guo, M. Highly efficient hierarchical micelles integrating photothermal

therapy. Phys Med Biol. 2008, 53(9), R61-R109. DOI:10.1088/0031-9155/53/9/r01. therapy and singlet oxygen-synergized chemotherapy for cancer eradication. Theranostics. 2014, 4(4), 399-411. DOI:10.7150/thno.8171. 157.

Huang, Y.;Qiu, F.;Shen, L. Combining Two-Photon-Activated Fluorescence Resonance Energy Transfer and Near-Infrared Photothermal Effect of Unimolecular Micelles for Enhanced Photodynamic Therapy. Acs Nano. 2016, 10(11), 10489-10499. 42

ACS Paragon Plus Environment

Page 42 of 45

Page 43 of 45

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

DOI:10.1021/acsnano.6b06450. 158.

Tian, J.W.;Ding, L.;Ju, H.X. A Multifunctional Nanomicelle for Real-Time Targeted Imaging and Precise Near-Infrared Cancer Therapy. Angew Chem Int Edit. 2014, 53(36), 9544-9549. DOI:10.1002/ange.201405490.

159.

Lammers, T.;Kiessling, F.;Hennink, W.E.,Storm, G. Drug targeting to tumors: principles, pitfalls and (pre-) clinical progress. Journal of controlled release : official journal of the Controlled Release Society. 2012, 161(2), 175-187. DOI:10.1016/j.jconrel.2011.09.063.

160.

Lu, Y.,Park, K. Polymeric micelles and alternative nanonized delivery vehicles for poorly soluble drugs. International Journal of Pharmaceutics. 2013, 453(1), 198-214. DOI:10.1016/j.ijpharm.2012.08.042.

161.

Lee, K.S.;Chung, H.C.;Im, S.A. Multicenter phase II trial of Genexol-PM, a Cremophor-free, polymeric micelle formulation of paclitaxel, in patients with metastatic breast cancer. Breast cancer research and treatment. 2008, 108(2), 241-250. DOI:10.1007/s10549-007-9591-y.

162.

Kim, D.W.;Kim, S.Y.;Kim, H.K. Multicenter phase II trial of Genexol-PM, a novel Cremophor-free, polymeric micelle formulation of paclitaxel, with cisplatin in patients with advanced non-small-cell lung cancer. Annals of oncology : official journal of the European Society for Medical Oncology. 2007, 18(12), 2009-2014.DOI:10.1093/annonc/mdm374.

163.

Lee, S.W.;Yun, M.H.;Jeong, S.W.;In, C.H.;Kim, J.Y.;Seo, M.H.;Pai, C.M.,Kim, S.O. Development of docetaxel-loaded intravenous formulation, Nanoxel-PM using polymer-based delivery system. Journal of controlled release : official journal of the Controlled Release Society. 2011, 155(2), 262-271. DOI:10.1016/j.jconrel.2011.06.012.

164.

Schutz, C.A.;Juillerat-Jeanneret, L.;Mueller, H.;Lynch, I.,Riediker, M. Therapeutic nanoparticles in clinics and under clinical evaluation. Nanomedicine. 2013, 8(3), 449–467.DOI:10.2217/nnm.13.8.

165.

Matsumura, Y.;Hamaguchi, T.;Ura, T. Phase I clinical trial and pharmacokinetic evaluation of NK911, a micelle-encapsulated doxorubicin. British journal of cancer. 2004, 91(10), 1775-1781. DOI:10.1038/sj.bjc.6602204.

166.

Ohnogi, H.;Hayami, S.;Kudo, Y. Angelica keiskei extract improves insulin resistance and hypertriglyceridemia in rats fed a high-fructose drink. Bioscience, biotechnology, and biochemistry. 2012, 76(5), 928-932. DOI:10.1271/bbb.110927.

167.

Plummer, R.;Wilson, R.H.;Calvert, H. A Phase I clinical study of cisplatin-incorporated polymeric micelles (NC-6004) in patients with solid tumours. British journal of cancer. 2011, 104(4), 593-598. DOI:10.1038/bjc.2011.6

168.

Hamaguchi, T.;Doi, T.;Eguchi-Nakajima, T. Phase I study of NK012, a novel SN-38-incorporating micellar nanoparticle, in adult patients with solid tumors. Clinical cancer research : an official journal of the American Association for Cancer Research. 2010, 16(20), 5058-5066. DOI:10.1158/1078-0432.CCR-10-0387.

169.

S, G.;C-C, L.;DC, C. A phase I study of the novel DNA topoisomerase-1 inhibitor, TLC388 (Lipotecan®), administered intravenously to patients with advanced solid tumors. Mol Cancer Ther 2011, 10(1), 89. DOI:10.1158/1535-7163.targ-11-a89.

43

ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

44

ACS Paragon Plus Environment

Page 44 of 45

Page 45 of 45

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Biomaterials Science & Engineering

For Table of Contents Use Only.

Schematic illustration of multifunctional NIR agents for cancer imaging and phototherapy.

Near-infrared Light-triggered Polymeric Nanomicelles for Cancer Therapy and Imaging Lei Li, Xin Pang, and Gang Liu

45

ACS Paragon Plus Environment