701
|
Jiang Y, Pu K. Molecular Fluorescence and Photoacoustic Imaging in the Second Near‐Infrared Optical Window Using Organic Contrast Agents. ACTA ACUST UNITED AC 2018; 2:e1700262. [DOI: 10.1002/adbi.201700262] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 02/10/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Yuyan Jiang
- School of Chemical and Biomedical Engineering Nanyang Technological University 70 Nanyang Drive Singapore 637457 Singapore
| | - Kanyi Pu
- School of Chemical and Biomedical Engineering Nanyang Technological University 70 Nanyang Drive Singapore 637457 Singapore
| |
Collapse
|
702
|
Ye H, Shen Z, Yu L, Wei M, Li Y. Manipulating nanoparticle transport within blood flow through external forces: an exemplar of mechanics in nanomedicine. Proc Math Phys Eng Sci 2018; 474:20170845. [PMID: 29662344 DOI: 10.1098/rspa.2017.0845] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 02/16/2018] [Indexed: 02/05/2023] Open
Abstract
A large number of nanoparticles (NPs) have been raised for diverse biomedical applications and some of them have shown great potential in treatment and imaging of diseases. Design of NPs is essential for delivery efficacy due to a number of biophysical barriers, which prevents the circulation of NPs in vascular flow and their accumulation at tumour sites. The physiochemical properties of NPs, so-called '4S' parameters, such as size, shape, stiffness and surface functionalization, play crucial roles in their life journey to be delivered to tumour sites. NPs can be modified in various ways to extend their blood circulation time and avoid their clearance by phagocytosis, and efficiently diffuse into tumour cells. However, it is difficult to overcome these barriers simultaneously by a simple combination of '4S' parameters for NPs. At this moment, external triggerings are necessary to guide the movement of NPs, which include light, ultrasound, magnetic field, electrical field and chemical interaction. The delivery system can be constructed to be sensitive to these external stimuli which can reduce the non-specific toxicity and improve the efficacy of the drug-delivery system. From a mechanics point of view, we discuss how different forces play their roles in the margination of NPs in blood flow and tumour microvasculature.
Collapse
Affiliation(s)
- Huilin Ye
- Department of Mechanical Engineering, University of Connecticut, 191 Auditorium Road, Unit 3139, Storrs, CT 06269, USA
| | - Zhiqiang Shen
- Department of Mechanical Engineering, University of Connecticut, 191 Auditorium Road, Unit 3139, Storrs, CT 06269, USA
| | - Le Yu
- Department of Materials Science and Engineering, University of Connecticut, 97 North Eagleville Road, Unit 3136, Storrs, CT 06269, USA
| | - Mei Wei
- Department of Materials Science and Engineering, University of Connecticut, 97 North Eagleville Road, Unit 3136, Storrs, CT 06269, USA.,Institute of Materials Science, University of Connecticut, 97 North Eagleville Road, Unit 3136, Storrs, CT 06269, USA
| | - Ying Li
- Department of Mechanical Engineering, University of Connecticut, 191 Auditorium Road, Unit 3139, Storrs, CT 06269, USA.,Institute of Materials Science, University of Connecticut, 97 North Eagleville Road, Unit 3136, Storrs, CT 06269, USA
| |
Collapse
|
703
|
Chen W, Qin M, Chen X, Wang Q, Zhang Z, Sun X. Combining photothermal therapy and immunotherapy against melanoma by polydopamine-coated Al 2O 3 nanoparticles. Am J Cancer Res 2018; 8:2229-2241. [PMID: 29721075 PMCID: PMC5928883 DOI: 10.7150/thno.24073] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 02/04/2018] [Indexed: 12/14/2022] Open
Abstract
Photothermal therapy (PTT) can be an effective antitumor therapy, but it may not completely eliminate tumor cells, leading to the risk of recurrence or metastasis. Here we describe nanocarriers that allow combination therapy involving PTT and immunotherapy. Nanocarriers are prepared by coating Al2O3 nanoparticles with non-toxic, biodegradable polydopamine, which shows high photothermal efficiency. A near-infrared laser irradiation can kill the majority of tumor tissues, resulting in the release of tumor-associated antigens. The Al2O3 within the nanoparticles, together with CpG, acts as an adjuvant to trigger robust cell-mediated immune responses that can help eliminate the residual tumor cells and reduce the risk of tumor recurrence. Methods: The characteristics and photothermal performance of polydopamine-coated Al2O3 nanoparticles were examined after one-step preparation. Then we studied their internalization, photothermal toxicity and immunostimulatory activity in vitro. For in vivo experiments, these nanocarriers were injected directly into B16F10 melanoma allografts in mice to ensure specific localization. After photothermal irradiation on day 0, mice were subcutaneously injected with CpG adjuvant on day 1, 3 and 5. Tumor volumes and number of living mice were recorded every two days. Moreover, various immune responses induced by our combined therapy were tested for mechanism research. Results: 50% of mice after our combined treatment successfully achieved the goal of tumor eradication, and survived for 120 days, which was the end point of the experiment. Mechanism studies demonstrated the combined therapy efficiently led to dendritic cell maturation, resulting in the secretion of antibodies and cytokines as well as the proliferation of splenocytes and lymphocytes for anti-tumor immunotherapy. Conclusion: Taken together, these results demonstrated the promise of our combined photothermal therapy and immunotherapy for tumor shrinkage, which merited further research.
Collapse
|
704
|
|
705
|
Zhang M, Song CC, Su S, Du FS, Li ZC. ROS-Activated Ratiometric Fluorescent Polymeric Nanoparticles for Self-Reporting Drug Delivery. ACS APPLIED MATERIALS & INTERFACES 2018; 10:7798-7810. [PMID: 29424527 DOI: 10.1021/acsami.7b18438] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Reactive oxygen species (ROS)-responsive theranostic nanomedicines have attracted wide interest in recent years because ROS stress is implicated in some pathological disorders such as inflammatory diseases and cancers. In this article, we report a kind of innovative ROS-responsive theranostic polymeric nanoparticles that are able to load hydrophobic drugs and to fluorescently self-report the in vitro or intracellular drug release under ROS triggering. The fluorescent nanoparticles were formed by amphiphilic block copolymers consisting of a poly(ethylene glycol) (PEG) segment and an oxidation-responsive hydrophobic block. The copolymers with different hydrophobic block lengths were synthesized by the atom transfer radical polymerization of a phenylboronic ester-containing acrylic monomer with a small fraction of a ROS-activatable 1,8-naphthalimide-based fluorescent monomer, using PEG-Br as the macroinitiator. The copolymer nanoparticles were stable in neutral phosphate buffer but degraded upon H2O2 triggering, with the degradation rate depending on the hydrophobic block length and the concentration of H2O2. The degradation of nanoparticles was accompanied by a colorimetric change of the fluorophore from blue to green, which affords the nanoparticles the ability to detecting H2O2 by a ratiometric fluorescent approach. Moreover, the nanoparticles could encapsulate doxorubicin (DOX) and the H2O2-triggered DOX release was well associated with the change in ratiometric fluorescence. Confocal laser scanning microscope results reveal that the fluorescent nanoparticles were internalized into A549 cells through the endocytosis pathway. The ROS-stimulated degradation of the nanoparticles and intracellular DOX release and the fate of the degraded polymers could be monitored by ratiometric fluorescent imaging. Finally, the naked nanoparticles and the degradation products are cytocompatible, whereas the DOX-loaded ones exhibit concentration-dependent cytotoxicity. Of importance, the stimulation with exogenous H2O2 or lipopolysaccharide enhanced obviously the cell-killing capability of the DOX-loaded nanoparticles because of the ROS-enhanced intracellular DOX release.
Collapse
Affiliation(s)
- Mei Zhang
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Polymer Chemistry & Physics of Ministry of Education, Department of Polymer Science & Engineering, College of Chemistry and Molecular Engineering , Peking University , Beijing 100871 , China
| | - Cheng-Cheng Song
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Polymer Chemistry & Physics of Ministry of Education, Department of Polymer Science & Engineering, College of Chemistry and Molecular Engineering , Peking University , Beijing 100871 , China
| | - Shan Su
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Polymer Chemistry & Physics of Ministry of Education, Department of Polymer Science & Engineering, College of Chemistry and Molecular Engineering , Peking University , Beijing 100871 , China
| | - Fu-Sheng Du
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Polymer Chemistry & Physics of Ministry of Education, Department of Polymer Science & Engineering, College of Chemistry and Molecular Engineering , Peking University , Beijing 100871 , China
| | - Zi-Chen Li
- Beijing National Laboratory for Molecular Sciences (BNLMS), Key Laboratory of Polymer Chemistry & Physics of Ministry of Education, Department of Polymer Science & Engineering, College of Chemistry and Molecular Engineering , Peking University , Beijing 100871 , China
| |
Collapse
|
706
|
Yang B, Yin J, Chen Y, Pan S, Yao H, Gao Y, Shi J. 2D-Black-Phosphorus-Reinforced 3D-Printed Scaffolds:A Stepwise Countermeasure for Osteosarcoma. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:1705611. [PMID: 29333689 DOI: 10.1002/adma.201705611] [Citation(s) in RCA: 230] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 11/15/2017] [Indexed: 05/21/2023]
Abstract
With the ever-deeper understanding of nano-bio interactions and the development of fabrication methodologies of nanomaterials, various therapeutic platforms based on nanomaterials have been developed for next-generation oncological applications, such as osteosarcoma therapy. In this work, a black phosphorus (BP) reinforced 3D-printed scaffold is designed and prepared to provide a feasible countermeasure for the efficient localized treatment of osteosarcoma. The in situ phosphorus-driven, calcium-extracted biomineralization of the intra-scaffold BP nanosheets enables both photothermal ablation of osteosarcoma and the subsequent material-guided bone regeneration in physiological microenvironment, and in the meantime endows the scaffolds with unique physicochemical properties favoring the whole stepwise therapeutic process. Additionally, a corrugated structure analogous to Haversian canals is found on newborn cranial bone tissue of Sprague-Dawley rats, which may provide much inspiration for the future research of bone-tissue engineering.
Collapse
Affiliation(s)
- Bowen Yang
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Junhui Yin
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, P. R. China
| | - Yu Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Shanshan Pan
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Heliang Yao
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| | - Youshui Gao
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, P. R. China
| | - Jianlin Shi
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| |
Collapse
|
707
|
Jia Q, Ge J, Liu W, Zheng X, Chen S, Wen Y, Zhang H, Wang P. A Magnetofluorescent Carbon Dot Assembly as an Acidic H 2 O 2 -Driven Oxygenerator to Regulate Tumor Hypoxia for Simultaneous Bimodal Imaging and Enhanced Photodynamic Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1706090. [PMID: 29436031 DOI: 10.1002/adma.201706090] [Citation(s) in RCA: 298] [Impact Index Per Article: 42.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 12/15/2017] [Indexed: 05/17/2023]
Abstract
Recent studies indicate that carbon dots (CDs) can efficiently generate singlet oxygen (1 O2 ) for photodynamic therapy (PDT) of cancer. However, the hypoxic tumor microenvironment and rapid consumption of oxygen in the PDT process will severely limit therapeutic effects of CDs due to the oxygen-dependent PDT. Thus, it is becoming particularly important to develop a novel CD as an in situ tumor oxygenerator for overcoming hypoxia and substantially enhancing the PDT efficacy. Herein, for the first time, magnetofluorescent Mn-CDs are successfully prepared using manganese(II) phthalocyanine as a precursor. After cooperative self-assembly with DSPE-PEG, the obtained Mn-CD assembly can be applied as a smart contrast agent for both near-infrared fluorescence (FL) (maximum peak at 745 nm) and T1 -weighted magnetic resonance (MR) (relaxivity value of 6.97 mM-1 s-1 ) imaging. More interestingly, the Mn-CD assembly can not only effectively produce 1 O2 (quantum yield of 0.40) but also highly catalyze H2 O2 to generate oxygen. These collective properties of the Mn-CD assembly enable it to be utilized as an acidic H2 O2 -driven oxygenerator to increase the oxygen concentration in hypoxic solid tumors for simultaneous bimodal FL/MR imaging and enhanced PDT. This work explores a new biomedical use of CDs and provides a versatile carbon nanomaterial candidate for multifunctional nanotheranostic applications.
Collapse
Affiliation(s)
- Qingyan Jia
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jiechao Ge
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Weimin Liu
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiuli Zheng
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shiqing Chen
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yongmei Wen
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hongyan Zhang
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Pengfei Wang
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials and CityU-CAS Joint Laboratory of Functional Materials and Devices, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
708
|
Chen H, Chen Y, Wu H, Xu JF, Sun Z, Zhang X. Supramolecular polymeric chemotherapy based on cucurbit[7]uril-PEG copolymer. Biomaterials 2018; 178:697-705. [PMID: 29545011 DOI: 10.1016/j.biomaterials.2018.02.051] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 02/08/2018] [Accepted: 02/27/2018] [Indexed: 02/06/2023]
Abstract
We develop a strategy of supramolecular polymeric chemotherapy based on a new kind of water-soluble polymer that bears cucurbit[7]uril (CB[7]) in the main-chain. To this end, we synthesized a bis-alkynyl functionalized CB[7] and polymerized it with α,ω-diazide-PEG through click reaction to form the desired CB[7] based main-chain polymer (poly-CB[7]). Anticancer drug, oxaliplatin, could be encapsulated into the cavity of poly-CB[7] to form a supramolecular polymeric complex, which displayed low cytotoxicity to normal cells. In addition, the cytotoxicity of the oxaliplatin was recovered when the complex met cancer cells that could overexpress spermine, e.g. colorectal cancer cell, through competitive replacement of oxaliplatin from CB[7] cavity by spermine. Interestingly, the cytotoxicity of the supramolecular polymeric complex to cancer cells is higher than oxaliplatin itself. The enhanced cytotoxicity should result from a combined effect by combining the release of oxaliplatin from the supramolecular polymeric complex and decrease of spermine in the micro-environment of the cancer cells, as spermine is needed for cell growth and proliferation. One more advantage of the supramolecular polymeric complex is its long circulation performance in vivo compared with the supramolecular complex between oxaliplatin and CB[7]. Therefore, this line of research may open new horizons for supramolecular polymeric chemotherapy.
Collapse
Affiliation(s)
- Hao Chen
- Key Lab of Organic Optoelectronics & Molecular Engineering, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Yueyue Chen
- Department of Toxicology and Sanitary Chemistry, School of Public Health, and Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Han Wu
- Key Lab of Organic Optoelectronics & Molecular Engineering, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Jiang-Fei Xu
- Key Lab of Organic Optoelectronics & Molecular Engineering, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, and Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China.
| | - Xi Zhang
- Key Lab of Organic Optoelectronics & Molecular Engineering, Department of Chemistry, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
709
|
Perfluorooctyl bromide & indocyanine green co-loaded nanoliposomes for enhanced multimodal imaging-guided phototherapy. Biomaterials 2018; 165:1-13. [PMID: 29500978 DOI: 10.1016/j.biomaterials.2018.02.041] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 01/31/2018] [Accepted: 02/21/2018] [Indexed: 01/08/2023]
Abstract
As a highly biocompatible NIR dye, indocyanine green (ICG) has been widely explored for cancer treatment due to its various energy level transition pathways upon NIR light excitation simultaneously, which leads to different theranostic effects (eg. Photoacoustic (PA) and fluorescence imaging (FL), photodynamic and photothermal therapy (PDT&PTT)). However, the theranostic efficiency of ICG is restricted intrinsically, owing to the competitive relationship of its co-existing imaging and therapeutic effect. Moreover, the extrinsic hypoxia nature of tumor further limits its therapeutic effect, especially for the oxygen-dependent PDT. Herein, perfluorooctyl bromide (PFOB), another biocompatible chemical, was integrated with ICG in a nanoliposome structure via a facile two-step emulsion method. Such an ICG&PFOB co-loaded nanoliposomes (LIP-PFOB-ICG) realized computed tomography (CT) contrast imaging in vivo, providing better anatomical information of tumor in comparison to ICG enabled PA and FL imaging. More importantly, LIP-PFOB-ICG inhibited MDA-MB-231 tumor growth completely via intravenous injection through enhanced PDT&PTT synergistic therapy due to the excellent oxygen carrying ability of PFOB, which effectively attenuated tumor hypoxia, improved the efficiency of collisional energy transfer between ICG and oxygen and reduced the expression of heat shock protein (HSP). As expected, the introduction of PFOB within nanoliposomes with ICG has augmented the theranostic effect of ICG comprehensively, which makes this simple biocompatible liposome-based nanoagent a potential candidate for clinical imaging guided phototherapy of cancer.
Collapse
|
710
|
Zhang L, Sheng D, Wang D, Yao Y, Yang K, Wang Z, Deng L, Chen Y. Bioinspired Multifunctional Melanin-Based Nanoliposome for Photoacoustic/Magnetic Resonance Imaging-Guided Efficient Photothermal Ablation of Cancer. Theranostics 2018; 8:1591-1606. [PMID: 29556343 PMCID: PMC5858169 DOI: 10.7150/thno.22430] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 12/08/2017] [Indexed: 12/15/2022] Open
Abstract
Background: The construction of theranostic nanosystems with concurrently high biosafety and therapeutic performance is a challenge but has great significance for the clinical translation of nanomedicine for combating cancer. Methods: Bio-inspired melanin-based nanoliposomes (Lip-Mel) as theranostic agents were constructed for simultaneous photoacoustic (PA) imaging- and T1-weighted magnetic resonance (MR) imaging-guided photothermal ablation of tumors, which was demonstrated both in vitro and in vivo. The high biosafety of Lip-Mel was also systematically evaluated. Results: The achieved Lip-Mel nanoliposomes demonstrated their imaging capability for both PA and T1-weighted MR imaging (r1 = 0.25 mM-1·s-1) both in vitro and in vivo, providing the potential for therapeutic guidance and monitoring. Importantly, the desirable photothermal-conversion efficiency of the as-prepared Lip-Mel achieved complete eradication of tumors in breast cancer-bearing mice, exhibiting remarkable photothermal-based therapeutic performance. In particular, the efficient encapsulation of melanin into the PEGylated liposome mitigated the potential toxicity of melanin and improved the photothermal performance of the loaded melanin. Systematic in vivo biosafety evaluations demonstrated the high biocompatibility of Lip-Mel at a high dose of 100 mg/kg. Conclusion: In this work, we reported a bioinspired strategy where melanin, a natural product in the human body, is encapsulated into PEGylated nanoliposomes for efficient theranostics with high biocompatibility. This work provides a new strategy for creating desirable theranostic agents with concurrent high biocompatibility and satisfactory theranostic performance through the use of materials that totally originate from biosystems.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P. R. China
| | - Danli Sheng
- Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Dong Wang
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P. R. China
| | - Yuanzhi Yao
- Department of Ultrasound, Chongqing Cancer Hospital, Chongqing Cancer Institute, Chongqing Cancer Center, Chongqing 400030, P. R. China
| | - Ke Yang
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P. R. China
| | - Zhigang Wang
- Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Liming Deng
- Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Yu Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| |
Collapse
|
711
|
Bose RJ, Paulmurugan R, Moon J, Lee SH, Park H. Cell membrane-coated nanocarriers: the emerging targeted delivery system for cancer theranostics. Drug Discov Today 2018; 23:891-899. [PMID: 29426004 DOI: 10.1016/j.drudis.2018.02.001] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 01/06/2018] [Accepted: 02/01/2018] [Indexed: 01/12/2023]
Abstract
Cancer is a leading cause of death worldwide. The use of nanocarriers (NCs) has generated significant interest to improve cancer therapy by targeted delivery. However, conventional NCs in general lack specificity and have poor biodistribution, resulting in low efficacy in cancer therapy. To circumvent this problem, there has been an increasing focus on cancer cell membrane-coated NCs (CCMCNCs), which can deliver therapeutics directly to tumor cells. CCMCNCs comprise active cancer cell surface adhesive molecules combined with other functional proteins, and offer extended blood circulation with robust cell-specific targeting, ensuring enhanced intratumoral penetration and higher tumor-specific accumulation of NCs. In this review, we discuss the preparation, homologous targeting mechanisms, and application of CCMCNCs in targeted cancer therapy.
Collapse
Affiliation(s)
- Rajendran Jc Bose
- School of Integrative Engineering, Chung-Ang University, Seoul, Republic of Korea; Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Republic of Korea; Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ramasamy Paulmurugan
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - James Moon
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Soo-Hong Lee
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, Republic of Korea.
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul, Republic of Korea.
| |
Collapse
|
712
|
Yi Y, Lin G, Chen S, Liu J, Zhang H, Mi P. Polyester micelles for drug delivery and cancer theranostics: Current achievements, progresses and future perspectives. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 83:218-232. [DOI: 10.1016/j.msec.2017.10.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 10/03/2017] [Accepted: 10/04/2017] [Indexed: 12/14/2022]
|
713
|
Gu Z, Yan S, Cheong S, Cao Z, Zuo H, Thomas AC, Rolfe BE, Xu ZP. Layered double hydroxide nanoparticles: Impact on vascular cells, blood cells and the complement system. J Colloid Interface Sci 2018; 512:404-410. [DOI: 10.1016/j.jcis.2017.10.069] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 10/17/2017] [Accepted: 10/17/2017] [Indexed: 10/18/2022]
|
714
|
Chatterjee B, Raza A, Ghosh SS. Developing single-entity theranostic: drug-based fluorescent nanoclusters with augmented cytotoxicity. Nanomedicine (Lond) 2018; 13:283-295. [PMID: 29345211 DOI: 10.2217/nnm-2017-0275] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM To develop methotrexate (MTX) templated luminescent gold nanoclusters (NCs) as a single unit nanotheranostic for cancer therapy and to assess its potential as an alternative to the parent drug, for drug delivery vehicles (DDVs). METHODS Theranostics were synthesized and extensively characterized. The stability of the theranostic and its bioimaging aptitude were evaluated. The antiproliferative propensity of the theranostic was gauged with cell viability assays and was supplemented with cytometry-based assays. Feasibility of delivering the MTX NCs instead of parent drug on a DDV was also checked. RESULTS MTX NCs displayed remarkable physical characteristics and augmented cytotoxicity with a robust stability in phosphate-buffered saline and serum. MTX NCs also demonstrated their amenability to being loaded on a DDV (chitosan folic acid nanoparticles) while retaining their physical and cytotoxic profile. CONCLUSION Generation of next level drug-based theranostics with the potential of replacing the free drug in drug delivery platforms.
Collapse
Affiliation(s)
- Bandhan Chatterjee
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Asif Raza
- Department of Biosciences & Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Siddhartha Sankar Ghosh
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.,Department of Biosciences & Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| |
Collapse
|
715
|
Su L, Li R, Khan S, Clanton R, Zhang F, Lin YN, Song Y, Wang H, Fan J, Hernandez S, Butters AS, Akabani G, MacLoughlin R, Smolen J, Wooley KL. Chemical Design of Both a Glutathione-Sensitive Dimeric Drug Guest and a Glucose-Derived Nanocarrier Host to Achieve Enhanced Osteosarcoma Lung Metastatic Anticancer Selectivity. J Am Chem Soc 2018; 140:1438-1446. [PMID: 29350522 DOI: 10.1021/jacs.7b11462] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Although nanomedicines have been pursued for nearly 20 years, fundamental chemical strategies that seek to optimize both the drug and drug carrier together in a concerted effort remain uncommon yet may be powerful. In this work, two block polymers and one dimeric prodrug molecule were designed to be coassembled into degradable, functional nanocarriers, where the chemistry of each component was defined to accomplish important tasks. The result is a poly(ethylene glycol) (PEG)-protected redox-responsive dimeric paclitaxel (diPTX)-loaded cationic poly(d-glucose carbonate) micelle (diPTX@CPGC). These nanostructures showed tunable sizes and surface charges and displayed controlled PTX drug release profiles in the presence of reducing agents, such as glutathione (GSH) and dithiothreitol (DTT), thereby resulting in significant selectivity for killing cancer cells over healthy cells. Compared to free PTX and diPTX, diPTX@CPGC exhibited improved tumor penetration and significant inhibition of tumor cell growth toward osteosarcoma (OS) lung metastases with minimal side effects both in vitro and in vivo, indicating the promise of diPTX@CPGC as optimized anticancer therapeutic agents for treatment of OS lung metastases.
Collapse
Affiliation(s)
- Lu Su
- Departments of Chemistry, Chemical Engineering, and Materials Science & Engineering, Texas A&M University , College Station, Texas 77842, United States
| | - Richen Li
- Departments of Chemistry, Chemical Engineering, and Materials Science & Engineering, Texas A&M University , College Station, Texas 77842, United States
| | - Sarosh Khan
- Departments of Chemistry, Chemical Engineering, and Materials Science & Engineering, Texas A&M University , College Station, Texas 77842, United States
| | - Ryan Clanton
- Departments of Nuclear Engineering and Veterinary Integrative Biosciences and Texas A&M Institute for Preclinical Studies, Texas A&M University , College Station, Texas 77842, United States
| | - Fuwu Zhang
- Departments of Chemistry, Chemical Engineering, and Materials Science & Engineering, Texas A&M University , College Station, Texas 77842, United States
| | - Yen-Nan Lin
- Departments of Chemistry, Chemical Engineering, and Materials Science & Engineering, Texas A&M University , College Station, Texas 77842, United States.,College of Medicine, Texas A&M University , Bryan, Texas 77807, United States
| | - Yue Song
- Departments of Chemistry, Chemical Engineering, and Materials Science & Engineering, Texas A&M University , College Station, Texas 77842, United States
| | - Hai Wang
- Departments of Chemistry, Chemical Engineering, and Materials Science & Engineering, Texas A&M University , College Station, Texas 77842, United States
| | - Jingwei Fan
- Departments of Chemistry, Chemical Engineering, and Materials Science & Engineering, Texas A&M University , College Station, Texas 77842, United States
| | - Soleil Hernandez
- Departments of Nuclear Engineering and Veterinary Integrative Biosciences and Texas A&M Institute for Preclinical Studies, Texas A&M University , College Station, Texas 77842, United States
| | - Andrew S Butters
- Departments of Nuclear Engineering and Veterinary Integrative Biosciences and Texas A&M Institute for Preclinical Studies, Texas A&M University , College Station, Texas 77842, United States
| | - Gamal Akabani
- Departments of Nuclear Engineering and Veterinary Integrative Biosciences and Texas A&M Institute for Preclinical Studies, Texas A&M University , College Station, Texas 77842, United States
| | - Ronan MacLoughlin
- Aerogen , IDA Business Park, Dangan, Galway, Ireland .,School of Pharmacy, Royal College of Surgeons , Dublin, Ireland .,School of Pharmacy and Pharmaceutical Sciences, Trinity College , Dublin, Ireland
| | - Justin Smolen
- Departments of Chemistry, Chemical Engineering, and Materials Science & Engineering, Texas A&M University , College Station, Texas 77842, United States
| | - Karen L Wooley
- Departments of Chemistry, Chemical Engineering, and Materials Science & Engineering, Texas A&M University , College Station, Texas 77842, United States
| |
Collapse
|
716
|
Abstract
Multifunctional nanoparticles with superior imaging properties and therapeutic effects have been extensively developed for the nanomedicine. However, tumor-intrinsic barriers and tumor heterogeneity have resulted in low in vivo therapeutic efficacy. The poor in vivo targeting efficiency in passive and active targeting of nanotherapeutics along with the toxicity of nanoparticles has been a major problem in nanomedicine. Recently, image-guided nanomedicine, which can deliver nanoparticles locally using non-invasive imaging and interventional oncology techniques, has been paid attention as a new opportunity of nanomedicine. This short review will discuss the existing challenges in nanomedicine and describe the prospects for future image-guided nanomedicine.
Collapse
Affiliation(s)
- Dong-Hyun Kim
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Robert H. Lurie Comprehensive Cancer Center, Chicago, IL 60611, USA
| |
Collapse
|
717
|
Wang Q, Li JM, Yu H, Deng K, Zhou W, Wang CX, Zhang Y, Li KH, Zhuo RX, Huang SW. Fluorinated polymeric micelles to overcome hypoxia and enhance photodynamic cancer therapy. Biomater Sci 2018; 6:3096-3107. [DOI: 10.1039/c8bm00852c] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Perfluoroalkyl groups-containing polymeric micelles were constructed to transport oxygen, overcome the hypoxia of tumours and enhance photodynamic cancer therapy.
Collapse
|
718
|
Wu L, Wen X, Wang X, Wang C, Sun X, Wang K, Zhang H, Williams T, Stacy AJ, Chen J, Schmieder AH, Lanza GM, Shen B. Local Intratracheal Delivery of Perfluorocarbon Nanoparticles to Lung Cancer Demonstrated with Magnetic Resonance Multimodal Imaging. Am J Cancer Res 2018; 8:563-574. [PMID: 29290827 PMCID: PMC5743567 DOI: 10.7150/thno.21466] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 10/05/2017] [Indexed: 12/15/2022] Open
Abstract
Eighty percent of lung cancers originate as subtle premalignant changes in the airway mucosal epithelial layer of bronchi and alveoli, which evolve and penetrate deeper into the parenchyma. Liquid-ventilation, with perfluorocarbons (PFC) was first demonstrated in rodents in 1966 then subsequently applied as lipid-encapsulated PFC emulsions to improve pulmonary function in neonatal infants suffering with respiratory distress syndrome in 1996. Subsequently, PFC nanoparticles (NP) were extensively studied as intravenous (IV) vascular-constrained nanotechnologies for diagnostic imaging and targeted drug delivery applications. Methods: This proof-of-concept study compared intratumoral localization of fluorescent paramagnetic (M) PFC NP in the Vx2 rabbit model using proton (1H) and fluorine (19F) magnetic resonance (MR) imaging (3T) following intratracheal (IT) or IV administration. MRI results were corroborated by fluorescence microscopy. Results: Dynamic 1H-MR and 19F-MR images (3T) obtained over 72 h demonstrated marked and progressive accumulation of M-PFC NP within primary lung Vx2 tumors during the first 12 h post IT administration. Marked 1H and 19F MR signal persisted for over 72 h. In contradistinction, IV M-PFC NP produced a modest transient signal during the initial 2 h post-injection that was consistent circumferential blood pool tumor enhancement. Fluorescence microscopy of excised tumors corroborated the MR results and revealed enormous intratumor NP deposition on day 3 after IT but not IV treatment. Rhodamine-phospholipid incorporated into the PFC nanoparticle surfactant was distributed widely within the tumor on day 3, which is consistent with a hemifusion-based contact drug delivery mechanism previously reported. Fluorescence microscopy also revealed similar high concentrations of M-PFC NP given IT for metastatic Vx2 lung tumors. Biodistribution studies in mice revealed that M-PFC NP given IV distributed into the reticuloendothelial organs, whereas, the same dosage given IT was basically not detected beyond the lung itself. PFC NP given IT did not impact rabbit behavior or impair respiratory function. PFC NP effects on cells in culture were negligible and when given IV or IT no changes in rabbit hematology nor serum clinical chemistry parameters were measured. Conclusion: IT delivery of PFC NP offered unique opportunity to locally deliver PFC NP in high concentrations into lung cancers with minimal extratumor systemic exposure.
Collapse
|
719
|
Zhao Z, Zhang Y, Tian C, Yin T, Zhang C. Facile dynamic one-step modular assembly based on boronic acid-diol for construction of a micellar drug delivery system. Biomater Sci 2018; 6:2605-2618. [DOI: 10.1039/c8bm00712h] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
This study reports a facile and dynamic one-step modular assembly strategy based on boronic acid-diol for constructing focus-responsive micellar drug delivery systems.
Collapse
Affiliation(s)
- Zekai Zhao
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Center of Advanced Pharmaceuticals and Biomaterials
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Ying Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Center of Advanced Pharmaceuticals and Biomaterials
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Chunli Tian
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Center of Advanced Pharmaceuticals and Biomaterials
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Tingjie Yin
- State Key Laboratory of Natural Medicines
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Can Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Center of Advanced Pharmaceuticals and Biomaterials
- China Pharmaceutical University
- Nanjing 210009
- China
| |
Collapse
|
720
|
Almeida-Marrero V, van de Winckel E, Anaya-Plaza E, Torres T, de la Escosura A. Porphyrinoid biohybrid materials as an emerging toolbox for biomedical light management. Chem Soc Rev 2018; 47:7369-7400. [DOI: 10.1039/c7cs00554g] [Citation(s) in RCA: 136] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The present article reviews the most important developing strategies in light-induced nanomedicine, based on the combination of porphyrinoid photosensitizers with a wide variety of biomolecules and biomolecular assemblies.
Collapse
Affiliation(s)
| | | | - Eduardo Anaya-Plaza
- Departamento de Química Orgánica
- Universidad Autónoma de Madrid
- Cantoblanco 28049
- Spain
| | - Tomás Torres
- Departamento de Química Orgánica
- Universidad Autónoma de Madrid
- Cantoblanco 28049
- Spain
- Institute for Advanced Research in Chemistry (IAdChem)
| | - Andrés de la Escosura
- Departamento de Química Orgánica
- Universidad Autónoma de Madrid
- Cantoblanco 28049
- Spain
- Institute for Advanced Research in Chemistry (IAdChem)
| |
Collapse
|
721
|
Liu H, Mai J, Shen J, Wolfram J, Li Z, Zhang G, Xu R, Li Y, Mu C, Zu Y, Li X, Lokesh GL, Thiviyanathan V, Volk DE, Gorenstein DG, Ferrari M, Hu Z, Shen H. A Novel DNA Aptamer for Dual Targeting of Polymorphonuclear Myeloid-derived Suppressor Cells and Tumor Cells. Am J Cancer Res 2018; 8:31-44. [PMID: 29290791 PMCID: PMC5743458 DOI: 10.7150/thno.21342] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 09/28/2017] [Indexed: 12/17/2022] Open
Abstract
Aptamers have the potential to be used as targeting ligands for cancer treatment as they form unique spatial structures. Methods: In this study, a DNA aptamer (T1) that accumulates in the tumor microenvironment was identified through in vivo selection and validation in breast cancer models. The use of T1 as a targeting ligand was evaluated by conjugating the aptamer to liposomal doxorubicin. Results: T1 exhibited a high affinity for both tumor cells and polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs). Treatment with T1 targeted doxorubicin liposomes triggered apoptosis of breast cancer cells and PMN-MDSCs. Suppression of PMN-MDSCs, which serve an immunosuppressive function, leads to increased intratumoral infiltration of cytotoxic T cells. Conclusion: The cytotoxic and immunomodulatory effects of T1-liposomes resulted in superior therapeutic efficacy compared to treatment with untargeted liposomes, highlighting the promise of T1 as a targeting ligand in cancer therapy.
Collapse
|
722
|
Fan J, He Q, Jin Z, Chen W, Huang W. A novel phosphoester-based cationic co-polymer nanocarrier delivers chimeric antigen receptor plasmid and exhibits anti-tumor effect. RSC Adv 2018; 8:14975-14982. [PMID: 35541346 PMCID: PMC9080078 DOI: 10.1039/c8ra02133c] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 04/16/2018] [Indexed: 12/30/2022] Open
Abstract
Chimeric antigen receptor T cells (CAR-T cells) targeting of CD19 antigen has been proven to be effective and successful in B cell acute lymphoblastic leukemia. The traditional CAR delivery systems have several problems such as poor biosafety, low loading capacity, and low transfection efficiency. Utilization of nanocarriers for CAR delivery offers new possibilities for CAR-T treatment. In the present study, an anti-CD19 CAR expression lentivirus plasmid was constructed for CAR delivery and immunotherapy. In addition, a three-segment amphiphilic co-polymer, methoxy polyethylene glycol-branched polyethyleneimine-poly(2-ethylbutyl phospholane) (mPEG-bPEI-PEBP) was synthesized via click reaction as the carrier with cationic PEI, capable of delivering the CAR and packaging plasmids to co-transfect Jurkat cells and undergo expression. The PEBP and mPEG parts of the co-polymer provide hydrophobic and hydrophilic interfaces and lead to the co-polymer self-assembly into micelles in water and encapsulation of the DNA plasmids. The mPEG-bPEI-PEBP-DNA composites with different N/P ratios were incubated with the CD19 overexpression K562 cells to identify the CAR functions. The obtained CAR-Jurkat cells had the ability to secrete interferon-γ and interleukin-2. The cytotoxic effects to CD19-K562 cells suggest that the induced CAR-Jurkat cells have an excellent targeted antitumor activity. A three-segment amphiphilic co-polymer mPEG-bPEI-PEBP was synthesized as the nanocarrier with cationic PEI, capable of delivering the CAR and packaging plasmids into Jurkat cells to generate the CAR-T cells for anti-CD19 immunotherapy study.![]()
Collapse
Affiliation(s)
- Jing Fan
- Key Laboratory of Medical Reprogramming Technology
- Shenzhen Second People's Hospital
- First Affiliated Hospital of Shenzhen University
- Shenzhen 518039
- China
| | - Qianjun He
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging
- School of Biomedical Engineering
- Health Science Center
- Shenzhen University
- Shenzhen 518060
| | - Zhaokui Jin
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging
- School of Biomedical Engineering
- Health Science Center
- Shenzhen University
- Shenzhen 518060
| | - Wei Chen
- Key Laboratory of Medical Reprogramming Technology
- Shenzhen Second People's Hospital
- First Affiliated Hospital of Shenzhen University
- Shenzhen 518039
- China
| | - Weiren Huang
- Key Laboratory of Medical Reprogramming Technology
- Shenzhen Second People's Hospital
- First Affiliated Hospital of Shenzhen University
- Shenzhen 518039
- China
| |
Collapse
|
723
|
Dehvari K, Lin PT, Chang JY. Fluorescence-guided magnetic nanocarriers for enhanced tumor targeting photodynamic therapy. J Mater Chem B 2018; 6:4676-4686. [DOI: 10.1039/c8tb00734a] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Fe3O4-HA-Ce6 nanotheranostic agents demonstrated specific targeting ability toward cancer cells with subsequent improvement in dual modal MR/NIR imaging and photodynamic therapeutic effects.
Collapse
Affiliation(s)
- Khalilalrahman Dehvari
- Department of Chemical Engineering
- National Taiwan University of Science and Technology
- Taiwan
- Republic of China
| | - Po-Ting Lin
- Department of Chemical Engineering
- National Taiwan University of Science and Technology
- Taiwan
- Republic of China
| | - Jia-Yaw Chang
- Department of Chemical Engineering
- National Taiwan University of Science and Technology
- Taiwan
- Republic of China
- Taiwan Building Technology Center
| |
Collapse
|
724
|
Zhang W, Deng G, Li B, Zhao X, Ji T, Song G, Xiao Z, Cao Q, Xiao J, Huang X, Guan G, Zou R, Lu X, Hu J. Degradable rhenium trioxide nanocubes with high localized surface plasmon resonance absorbance like gold for photothermal theranostics. Biomaterials 2017; 159:68-81. [PMID: 29316453 DOI: 10.1016/j.biomaterials.2017.12.021] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 12/21/2017] [Accepted: 12/24/2017] [Indexed: 01/08/2023]
Abstract
The applications of inorganic theranostic agents in clinical trials are generally limited to their innate non-biodegradability and potential long-term biotoxicity. To address this problem, herein via a straightforward and tailored space-confined on-substrate route, we obtained rhenium trioxide (ReO3) nanocubes (NCs) that display a good biocompatibility and biosafety. Importantly, their aqueous dispersion has high localized surface plasmon resonance (LSPR) absorbance in near-infrared (NIR) region different from previous report, which possibly associates with the charge transfer and structural distortion in hydrogen rhenium bronze (HxReO3), as well as ReO3's cubic shape. Such a high LSPR absorbance in the NIR region endows them with photoacoustic (PA)/infrared (IR) thermal imaging, and high photothermal conversion efficiency (∼57.0%) for efficient ablation of cancer cells. Also, ReO3 NCs show X-ray computed tomography (CT) imaging derived from the high-Z element Re. More attractively, those ReO3 NCs, with pH-dependent oxidized degradation behaviors, are revealed to be relatively stable in hypoxic and weakly acidic microenvironment of tumor for imaging and treatment whilst degradable in normal physiological environments of organs to enable effective clearance. In spite of their degradability, ReO3 NCs still possess tumor targeting capabilities. We thus develop a simple but powerful, safe and biodegradable inorganic theranostic platform to achieve PA/CT/IR imaging-guided cancer photothermal therapy (PTT) for improved therapeutic efficacy and decreased toxic side effects.
Collapse
Affiliation(s)
- Wenlong Zhang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| | - Guoying Deng
- Trauma Center, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201620, China
| | - Bo Li
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Xinxin Zhao
- School of Mathmatics, Physics and Statistics, Shanghai University of Engineering Science, Shanghai, 201620, China
| | - Tao Ji
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| | - Guosheng Song
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University School of Medicine, 1201 Welch Road, Stanford, CA, 94305-5484, USA
| | - Zhiyin Xiao
- College of Biological, Chemical Sciences and Engineering, Jiaxing University, Jiaxing, 314001, China
| | - Qing Cao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| | - Jingbo Xiao
- Shanghai Key Laboratory of Pancreatic Diseases & Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201620, China
| | - Xiaojuan Huang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| | - Guoqiang Guan
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China
| | - Rujia Zou
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China.
| | - Xinwu Lu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200011, China.
| | - Junqing Hu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Materials Science and Engineering, Donghua University, Shanghai, 201620, China.
| |
Collapse
|
725
|
Vinhas R, Mendes R, Fernandes AR, Baptista PV. Nanoparticles-Emerging Potential for Managing Leukemia and Lymphoma. Front Bioeng Biotechnol 2017; 5:79. [PMID: 29326927 PMCID: PMC5741836 DOI: 10.3389/fbioe.2017.00079] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Accepted: 12/05/2017] [Indexed: 12/31/2022] Open
Abstract
Nanotechnology has become a powerful approach to improve the way we diagnose and treat cancer. In particular, nanoparticles (NPs) possess unique features for enhanced sensitivity and selectivity for earlier detection of circulating cancer biomarkers. In vivo, NPs enhance the therapeutic efficacy of anticancer agents when compared with conventional chemotherapy, improving vectorization and delivery, and helping to overcome drug resistance. Nanomedicine has been mostly focused on solid cancers due to take advantage from the enhanced permeability and retention (EPR) effect experienced by tissues in the close vicinity of tumors, which enhance nanomedicine's accumulation and, consequently, improve efficacy. Nanomedicines for leukemia and lymphoma, where EPR effect is not a factor, are addressed differently from solid tumors. Nevertheless, NPs have provided innovative approaches to simple and non-invasive methodologies for diagnosis and treatment in liquid tumors. In this review, we consider the state of the art on different types of nanoconstructs for the management of liquid tumors, from preclinical studies to clinical trials. We also discuss the advantages of nanoplatforms for theranostics and the central role played by NPs in this combined strategy.
Collapse
Affiliation(s)
- Raquel Vinhas
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Rita Mendes
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Alexandra R Fernandes
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Pedro V Baptista
- UCIBIO, Departamento de Ciências da Vida, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| |
Collapse
|
726
|
Chen G, Ma B, Wang Y, Xie R, Li C, Dou K, Gong S. CuS-Based Theranostic Micelles for NIR-Controlled Combination Chemotherapy and Photothermal Therapy and Photoacoustic Imaging. ACS APPLIED MATERIALS & INTERFACES 2017; 9:41700-41711. [PMID: 29154532 PMCID: PMC5915677 DOI: 10.1021/acsami.7b14083] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Cancer remains a major threat to human health due to low therapeutic efficacies of currently available cancer treatment options. Nanotheranostics, capable of simultaneous therapy and diagnosis/monitoring of diseases, has attracted increasing amounts of attention, particularly for cancer treatment. In this study, CuS-based theranostic micelles capable of simultaneous combination chemotherapy and photothermal therapy (PTT), as well as photoacoustic imaging, were developed for targeted cancer therapy. The micelle was formed by a CuS nanoparticle (NP) functionalized by thermosensitive amphiphilic poly(acrylamide-acrylonitrile)-poly(ethylene glycol) block copolymers. CuS NPs under near-infrared (NIR) irradiation induced a significant temperature elevation, thereby enabling NIR-triggered PTT. Moreover, the hydrophobic core formed by poly(acrylamide-acrylonitrile) segments used for drug encapsulation exhibited an upper critical solution temperature (UCST; ∼38 °C), which underwent a hydrophobic-to-hydrophilic transition once the temperature rose above the UCST induced by NIR-irradiated CuS NPs, thereby triggering a rapid drug release and enabling NIR-controlled chemotherapy. The CuS-based micelles conjugated with GE11 peptides were tested in an epidermal growth factor receptor-overexpressing triple-negative breast cancer model. In both two-dimensional monolayer cell and three-dimensional multicellular tumor spheroid models, GE11-tagged CuS-based micelles under NIR irradiation, enabling the combination chemotherapy and PTT, exhibited the best therapeutic outcome due to a synergistic effect. These CuS-based micelles also displayed a good photoacoustic imaging ability under NIR illumination. Taken together, this multifunctional CuS-based micelle could be a promising nanoplatform for targeted cancer nanotheranostics.
Collapse
Affiliation(s)
- Guojun Chen
- Department of Materials Science and Engineering, University of Wisconsin—Madison, Madison, Wisconsin 53715, United States
- Wisconsin Institute for Discovery and Department of Biomedical Engineering, University of Wisconsin—Madison, Madison, Wisconsin 53715, United States
| | - Ben Ma
- Wisconsin Institute for Discovery and Department of Biomedical Engineering, University of Wisconsin—Madison, Madison, Wisconsin 53715, United States
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Yuyuan Wang
- Department of Materials Science and Engineering, University of Wisconsin—Madison, Madison, Wisconsin 53715, United States
- Wisconsin Institute for Discovery and Department of Biomedical Engineering, University of Wisconsin—Madison, Madison, Wisconsin 53715, United States
| | - Ruosen Xie
- Department of Materials Science and Engineering, University of Wisconsin—Madison, Madison, Wisconsin 53715, United States
- Wisconsin Institute for Discovery and Department of Biomedical Engineering, University of Wisconsin—Madison, Madison, Wisconsin 53715, United States
| | - Chun Li
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Kefeng Dou
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, Shaanxi 710032, China
| | - Shaoqin Gong
- Department of Materials Science and Engineering, University of Wisconsin—Madison, Madison, Wisconsin 53715, United States
- Department of Chemistry, University of Wisconsin—Madison, Madison, Wisconsin 53715, United States
| |
Collapse
|
727
|
Zhu G, Lynn GM, Jacobson O, Chen K, Liu Y, Zhang H, Ma Y, Zhang F, Tian R, Ni Q, Cheng S, Wang Z, Lu N, Yung BC, Wang Z, Lang L, Fu X, Jin A, Weiss ID, Vishwasrao H, Niu G, Shroff H, Klinman DM, Seder RA, Chen X. Albumin/vaccine nanocomplexes that assemble in vivo for combination cancer immunotherapy. Nat Commun 2017; 8:1954. [PMID: 29203865 PMCID: PMC5715147 DOI: 10.1038/s41467-017-02191-y] [Citation(s) in RCA: 234] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 11/13/2017] [Indexed: 01/09/2023] Open
Abstract
Subunit vaccines have been investigated in over 1000 clinical trials of cancer immunotherapy, but have shown limited efficacy. Nanovaccines may improve efficacy but have rarely been clinically translated. By conjugating molecular vaccines with Evans blue (EB) into albumin-binding vaccines (AlbiVax), here we develop clinically promising albumin/AlbiVax nanocomplexes that self-assemble in vivo from AlbiVax and endogenous albumin for efficient vaccine delivery and potent cancer immunotherapy. PET pharmacoimaging, super-resolution microscopies, and flow cytometry reveal almost 100-fold more efficient co-delivery of CpG and antigens (Ags) to lymph nodes (LNs) by albumin/AlbiVax than benchmark incomplete Freund's adjuvant (IFA). Albumin/AlbiVax elicits ~10 times more frequent peripheral antigen-specific CD8+ cytotoxic T lymphocytes with immune memory than IFA-emulsifying vaccines. Albumin/AlbiVax specifically inhibits progression of established primary or metastatic EG7.OVA, B16F10, and MC38 tumors; combination with anti-PD-1 and/or Abraxane further potentiates immunotherapy and eradicates most MC38 tumors. Albumin/AlbiVax nanocomplexes are thus a robust platform for combination cancer immunotherapy.
Collapse
Affiliation(s)
- Guizhi Zhu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Geoffrey M Lynn
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, 20892, USA
| | - Orit Jacobson
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Kai Chen
- Molecular Imaging Center, Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Yi Liu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA.,School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Huimin Zhang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Ying Ma
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Fuwu Zhang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Rui Tian
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Qianqian Ni
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Siyuan Cheng
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Zhantong Wang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA.,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
| | - Nan Lu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Bryant C Yung
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Zhe Wang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Lixin Lang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Xiao Fu
- Laboratory of Cellular Imaging and Macromolecular Biophysics, NIBIB, NIH, Bethesda, MD, 20892, USA
| | - Albert Jin
- Laboratory of Cellular Imaging and Macromolecular Biophysics, NIBIB, NIH, Bethesda, MD, 20892, USA
| | - Ido D Weiss
- Laboratory of Molecular Immunology, NIAID, NIH, Bethesda, MD, 20892, USA
| | - Harshad Vishwasrao
- Advanced Imaging and Microscopy Resource, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Gang Niu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA
| | - Hari Shroff
- Advanced Imaging and Microscopy Resource, National Institutes of Health, Bethesda, 20892, MD, USA.,Section on High Resolution Optical Imaging, NIBIB, NIH, Bethesda, MD, 20892, USA
| | - Dennis M Klinman
- Cancer and Inflammation Program, National Cancer Institute, Frederick, MD, 21702, USA
| | - Robert A Seder
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Bethesda, MD, 20892, USA
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD, 20892, USA.
| |
Collapse
|
728
|
Gutiérrez-Lovera C, Vázquez-Ríos AJ, Guerra-Varela J, Sánchez L, de la Fuente M. The Potential of Zebrafish as a Model Organism for Improving the Translation of Genetic Anticancer Nanomedicines. Genes (Basel) 2017; 8:E349. [PMID: 29182542 PMCID: PMC5748667 DOI: 10.3390/genes8120349] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 11/06/2017] [Accepted: 11/21/2017] [Indexed: 12/21/2022] Open
Abstract
In the last few decades, the field of nanomedicine applied to cancer has revolutionized cancer treatment: several nanoformulations have already reached the market and are routinely being used in the clinical practice. In the case of genetic nanomedicines, i.e., designed to deliver gene therapies to cancer cells for therapeutic purposes, advances have been less impressive. This is because of the many barriers that limit the access of the therapeutic nucleic acids to their target site, and the lack of models that would allow for an improvement in the understanding of how nanocarriers can be tailored to overcome them. Zebrafish has important advantages as a model species for the study of anticancer therapies, and have a lot to offer regarding the rational development of efficient delivery of genetic nanomedicines, and hence increasing the chances of their successful translation. This review aims to provide an overview of the recent advances in the development of genetic anticancer nanomedicines, and of the zebrafish models that stand as promising tools to shed light on their mechanisms of action and overall potential in oncology.
Collapse
Affiliation(s)
- C Gutiérrez-Lovera
- Zoology, Genetics and Physical Anthropology Department Veterinary Faculty, Universidade de Santiago de Compostela, Lugo 27002, Spain.
- Nano-Oncology Unit, Translational Medical Oncology Group, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), CIBERONC, Santiago de Compostela 15706, Spain.
| | - A J Vázquez-Ríos
- Nano-Oncology Unit, Translational Medical Oncology Group, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), CIBERONC, Santiago de Compostela 15706, Spain.
| | - J Guerra-Varela
- Zoology, Genetics and Physical Anthropology Department Veterinary Faculty, Universidade de Santiago de Compostela, Lugo 27002, Spain.
- Geneaqua S.L., Lugo 27002, Spain.
| | - L Sánchez
- Zoology, Genetics and Physical Anthropology Department Veterinary Faculty, Universidade de Santiago de Compostela, Lugo 27002, Spain.
| | - M de la Fuente
- Nano-Oncology Unit, Translational Medical Oncology Group, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital of Santiago de Compostela (CHUS), CIBERONC, Santiago de Compostela 15706, Spain.
| |
Collapse
|
729
|
Meir R, Shamalov K, Sadan T, Motiei M, Yaari G, Cohen CJ, Popovtzer R. Fast Image-Guided Stratification Using Anti-Programmed Death Ligand 1 Gold Nanoparticles for Cancer Immunotherapy. ACS NANO 2017; 11:11127-11134. [PMID: 29028305 DOI: 10.1021/acsnano.7b05299] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Cancer immunotherapy has made enormous progress in offering safer and more effective treatments for the disease. Specifically, programmed death ligand 1 antibody (αPDL1), designed to perform immune checkpoint blockade (ICB), is now considered a pillar in cancer immunotherapy. However, due to the complexity and heterogeneity of tumors, as well as the diversity in patient response, ICB therapy only has a 30% success rate, at most; moreover, the efficacy of ICB can be evaluated only two months after start of treatment. Therefore, early identification of potential responders and nonresponders to therapy, using noninvasive means, is crucial for improving treatment decisions. Here, we report a straightforward approach for fast, image-guided prediction of therapeutic response to ICB. In a colon cancer mouse model, we demonstrate that the combination of computed tomography imaging and gold nanoparticles conjugated to αPDL1 allowed prediction of therapeutic response, as early as 48 h after treatment. This was achieved by noninvasive measurement of nanoparticle accumulation levels within the tumors. Moreover, we show that the nanoparticles efficiently prevented tumor growth with only a fifth of the standard dosage of clinical care. This technology may be developed into a powerful tool for early and noninvasive patient stratification as responders or nonresponders.
Collapse
Affiliation(s)
- Rinat Meir
- Faculty of Engineering, ‡The Institute of Nanotechnology and Advanced Materials, and §Laboratory of Tumor Immunology and Immunotherapy, Goodman Faculty of Life Sciences, Bar-Ilan University , Ramat Gan 52900, Israel
| | - Katerina Shamalov
- Faculty of Engineering, ‡The Institute of Nanotechnology and Advanced Materials, and §Laboratory of Tumor Immunology and Immunotherapy, Goodman Faculty of Life Sciences, Bar-Ilan University , Ramat Gan 52900, Israel
| | - Tamar Sadan
- Faculty of Engineering, ‡The Institute of Nanotechnology and Advanced Materials, and §Laboratory of Tumor Immunology and Immunotherapy, Goodman Faculty of Life Sciences, Bar-Ilan University , Ramat Gan 52900, Israel
| | - Menachem Motiei
- Faculty of Engineering, ‡The Institute of Nanotechnology and Advanced Materials, and §Laboratory of Tumor Immunology and Immunotherapy, Goodman Faculty of Life Sciences, Bar-Ilan University , Ramat Gan 52900, Israel
| | - Gur Yaari
- Faculty of Engineering, ‡The Institute of Nanotechnology and Advanced Materials, and §Laboratory of Tumor Immunology and Immunotherapy, Goodman Faculty of Life Sciences, Bar-Ilan University , Ramat Gan 52900, Israel
| | - Cyrille J Cohen
- Faculty of Engineering, ‡The Institute of Nanotechnology and Advanced Materials, and §Laboratory of Tumor Immunology and Immunotherapy, Goodman Faculty of Life Sciences, Bar-Ilan University , Ramat Gan 52900, Israel
| | - Rachela Popovtzer
- Faculty of Engineering, ‡The Institute of Nanotechnology and Advanced Materials, and §Laboratory of Tumor Immunology and Immunotherapy, Goodman Faculty of Life Sciences, Bar-Ilan University , Ramat Gan 52900, Israel
| |
Collapse
|
730
|
Ni D, Bu W, Ehlerding EB, Cai W, Shi J. Engineering of inorganic nanoparticles as magnetic resonance imaging contrast agents. Chem Soc Rev 2017; 46:7438-7468. [PMID: 29071327 PMCID: PMC5705441 DOI: 10.1039/c7cs00316a] [Citation(s) in RCA: 292] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Magnetic resonance imaging (MRI) is a highly valuable non-invasive imaging tool owing to its exquisite soft tissue contrast, high spatial resolution, lack of ionizing radiation, and wide clinical applicability. Contrast agents (CAs) can be used to further enhance the sensitivity of MRI to obtain information-rich images. Recently, extensive research efforts have been focused on the design and synthesis of high-performance inorganic nanoparticle-based CAs to improve the quality and specificity of MRI. Herein, the basic rules, including the choice of metal ions, effect of electron motion on water relaxation, and involved mechanisms, of CAs for MRI have been elucidated in detail. In particular, various design principles, including size control, surface modification (e.g. organic ligand, silica shell, and inorganic nanolayers), and shape regulation, to impact relaxation of water molecules have been discussed in detail. Comprehensive understanding of how these factors work can guide the engineering of future inorganic nanoparticles with high relaxivity. Finally, we have summarized the currently available strategies and their mechanism for obtaining high-performance CAs and discussed the challenges and future developments of nanoparticulate CAs for clinical translation in MRI.
Collapse
Affiliation(s)
- Dalong Ni
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China.
| | | | | | | | | |
Collapse
|
731
|
Mansur AAP, Mansur HS, Carvalho SM, Caires AJ. One-Pot Aqueous Synthesis of Fluorescent Ag-In-Zn-S Quantum Dot/Polymer Bioconjugates for Multiplex Optical Bioimaging of Glioblastoma Cells. CONTRAST MEDIA & MOLECULAR IMAGING 2017; 2017:3896107. [PMID: 29259535 PMCID: PMC5702976 DOI: 10.1155/2017/3896107] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 09/04/2017] [Accepted: 09/13/2017] [Indexed: 11/17/2022]
Abstract
Cancer research has experienced astonishing advances recently, but cancer remains a major threat because it is one of the leading causes of death worldwide. Glioblastoma (GBM) is the most malignant brain tumor, where the early diagnosis is vital for longer survival. Thus, this study reports the synthesis of novel water-dispersible ternary AgInS2 (AIS) and quaternary AgInS2-ZnS (ZAIS) fluorescent quantum dots using carboxymethylcellulose (CMC) as ligand for multiplexed bioimaging of malignant glioma cells (U-87 MG). Firstly, AgInS2 core was prepared using a one-pot aqueous synthesis stabilized by CMC at room temperature and physiological pH. Then, an outer layer of ZnS was grown and thermally annealed to improve their optical properties and split the emission range, leading to core-shell alloyed nanostructures. Their physicochemical and optical properties were characterized, demonstrating that luminescent monodispersed AIS and ZAIS QDs were produced with average sizes of 2.2 nm and 4.3 nm, respectively. Moreover, the results evidenced that they were cytocompatible using in vitro cell viability assays towards human embryonic kidney cell line (HEK 293T) and U-87 MG cells. These AIS and ZAIS successfully behaved as fluorescent nanoprobes (red and green, resp.) allowing multiplexed bioimaging and biolabeling of costained glioma cells using confocal microscopy.
Collapse
Affiliation(s)
- Alexandra A. P. Mansur
- Center of Nanoscience, Nanotechnology and Innovation (CeNanoI), Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Herman S. Mansur
- Center of Nanoscience, Nanotechnology and Innovation (CeNanoI), Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Sandhra M. Carvalho
- Center of Nanoscience, Nanotechnology and Innovation (CeNanoI), Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
- Department of Preventive Veterinary Medicine, Veterinary School, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
- Department of Physiology and Biophysics, ICB, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Anderson J. Caires
- Center of Nanoscience, Nanotechnology and Innovation (CeNanoI), Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| |
Collapse
|
732
|
Yu G, Zhang M, Saha ML, Mao Z, Chen J, Yao Y, Zhou Z, Liu Y, Gao C, Huang F, Chen X, Stang PJ. Antitumor Activity of a Unique Polymer That Incorporates a Fluorescent Self-Assembled Metallacycle. J Am Chem Soc 2017; 139:15940-15949. [PMID: 29019660 PMCID: PMC5827967 DOI: 10.1021/jacs.7b09224] [Citation(s) in RCA: 167] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Despite the well-known anticancer activity of mono- and multinuclear platinum complexes, studies of the antitumor performances of platinum-based supramolecular coordination complexes are rare. Herein, we report on the synthesis of a four-armed amphiphilic copolymer, Pt-PAZMB-b-POEGMA, containing a metallacycle M, in which the tetraphenylethene derivative acts as an aggregation-induced emissive fluorescent probe for live cell imaging and the 3,6-bis[trans-Pt(PEt3)2]phenanthrene (PhenPt) is an anticancer drug. This copolymer was further self-assembled into nanoparticles of different sizes and vesicles depending upon the experimental conditions. The impacts of the morphology and size of the assemblies on their endocytic pathways, uptake rates, internalization amounts, and cytotoxicities were investigated. The self-assemblies were further employed to encapsulate doxorubicin (DOX) to achieve a synergistic anticancer effect. Controlled drug release was also realized via amphiphilicity changes and was driven by a glutathione-induced cascade elimination reaction. The DOX-loaded nanoparticles of around 50 nm in size exhibited an excellent antitumor performance as well as a low systemic toxicity, due to an enhanced permeability and retention effect.
Collapse
Affiliation(s)
- Guocan Yu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Mingming Zhang
- Department of Chemistry, University of Utah, 315 South 1400 East, Room 2020, Salt Lake City, Utah 84112, United States
| | - Manik Lal Saha
- Department of Chemistry, University of Utah, 315 South 1400 East, Room 2020, Salt Lake City, Utah 84112, United States
| | - Zhengwei Mao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, P. R. China
| | - Jin Chen
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, P. R. China
| | - Yong Yao
- Department of Chemistry, University of Utah, 315 South 1400 East, Room 2020, Salt Lake City, Utah 84112, United States
| | - Zijian Zhou
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Yijing Liu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, P. R. China
| | - Feihe Huang
- State Key Laboratory of Chemical Engineering, Center for Chemistry of High-Performance & Novel Materials, Department of Chemistry, Zhejiang University, Hangzhou 310027, P. R. China
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland 20892, United States
| | - Peter J. Stang
- Department of Chemistry, University of Utah, 315 South 1400 East, Room 2020, Salt Lake City, Utah 84112, United States
| |
Collapse
|
733
|
Zhang F, Khan S, Li R, Smolen JA, Zhang S, Zhu G, Su L, Jahnke AA, Elsabahy M, Chen X, Wooley KL. Design and development of multifunctional polyphosphoester-based nanoparticles for ultrahigh paclitaxel dual loading. NANOSCALE 2017; 9:15773-15777. [PMID: 29034932 DOI: 10.1039/c7nr05935c] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Multifunctional polyphosphoester-based nanoparticles capable of loading paclitaxel (PTX) both chemically and physically were prepared, achieving an ultrahigh equivalent PTX aqueous concentration of 25.30 mg mL-1. The dual-loaded nanoparticles were effective in killing cancer cells, which has the potential to minimize the amount of nanocarriers needed for clinical applications, due to their ultrahigh loading capacity.
Collapse
Affiliation(s)
- Fuwu Zhang
- Departments of Chemistry, Chemical Engineering, and Materials Science and Engineering, Laboratory for Synthetic-Biologic Interactions, and Texas A&M Institute for Preclinical Studies, Texas A&M University, College Station, Texas 77842, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
734
|
Björnmalm M, Thurecht KJ, Michael M, Scott AM, Caruso F. Bridging Bio-Nano Science and Cancer Nanomedicine. ACS NANO 2017; 11:9594-9613. [PMID: 28926225 DOI: 10.1021/acsnano.7b04855] [Citation(s) in RCA: 262] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
The interface of bio-nano science and cancer medicine is an area experiencing much progress but also beset with controversy. Core concepts of the field-e.g., the enhanced permeability and retention (EPR) effect, tumor targeting and accumulation, and even the purpose of "nano" in cancer medicine-are hotly debated. In parallel, considerable advances in neighboring fields are occurring rapidly, including the recent progress of "immuno-oncology" and the fundamental impact it is having on our understanding and the clinical treatment of the group of diseases collectively known as cancer. Herein, we (i) revisit how cancer is commonly treated in the clinic and how this relates to nanomedicine; (ii) examine the ongoing debate on the relevance of the EPR effect and tumor targeting; (iii) highlight ways to improve the next-generation of nanomedicines; and (iv) discuss the emerging concept of working with (and not against) biology. While discussing these controversies, challenges, emerging concepts, and opportunities, we explore new directions for the field of cancer nanomedicine.
Collapse
Affiliation(s)
- Mattias Björnmalm
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering, The University of Melbourne , Parkville, Victoria 3010, Australia
| | - Kristofer J Thurecht
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, The Australian Institute for Bioengineering and Nanotechnology and The Centre for Advanced Imaging, The University of Queensland , Brisbane, Queensland 4072, Australia
| | - Michael Michael
- Division of Cancer Medicine, Peter MacCallum Cancer Centre , Melbourne, Victoria 3000, Australia
- The Peter MacCallum Department of Oncology, The University of Melbourne , Parkville, Victoria 3010, Australia
| | - Andrew M Scott
- Olivia Newton-John Cancer Research Institute, and School of Cancer Medicine, La Trobe University , Melbourne, Victoria 3084, Australia
- Department of Molecular Imaging and Therapy, Austin Hospital , Heidelberg, Victoria 3084, Australia
| | - Frank Caruso
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical and Biomolecular Engineering, The University of Melbourne , Parkville, Victoria 3010, Australia
| |
Collapse
|
735
|
Wang D, Zhou J, Shi R, Wu H, Chen R, Duan B, Xia G, Xu P, Wang H, Zhou S, Wang C, Wang H, Guo Z, Chen Q. Biodegradable Core-shell Dual-Metal-Organic-Frameworks Nanotheranostic Agent for Multiple Imaging Guided Combination Cancer Therapy. Theranostics 2017; 7:4605-4617. [PMID: 29158848 PMCID: PMC5695152 DOI: 10.7150/thno.20363] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Accepted: 09/29/2017] [Indexed: 12/16/2022] Open
Abstract
Metal-organic-frameworks (MOFs) possess high porosity, large surface area, and tunable functionality are promising candidates for synchronous diagnosis and therapy in cancer treatment. Although large number of MOFs has been discovered, conventional MOF-based nanoplatforms are mainly limited to the sole MOF source with sole functionality. In this study, surfactant modified Prussian blue (PB) core coated by compact ZIF-8 shell (core-shell dual-MOFs, CSD-MOFs) has been reported through a versatile stepwise approach. With Prussian blue as core, CSD-MOFs are able to serve as both magnetic resonance imaging (MRI) and fluorescence optical imaging (FOI) agents. We show that CSD-MOFs crystals loading the anticancer drug doxorubicin (DOX) are efficient pH and near-infrared (NIR) dual-stimuli responsive drug delivery vehicles. After the degradation of ZIF-8, simultaneous NIR irradiation to the inner PB MOFs continuously generate heat that kill cancer cells. Their efficacy on HeLa cancer cell lines is higher compared with the respective single treatment modality, achieving synergistic chemo-thermal therapy efficacy. In vivo results indicate that the anti-tumor efficacy of CSD-MOFs@DOX+NIR was 7.16 and 5.07 times enhanced compared to single chemo-therapy and single thermal-therapy respectively. Our strategy opens new possibilities to construct multifunctional theranostic systems through integration of two different MOFs.
Collapse
Affiliation(s)
- Dongdong Wang
- Hefei National Laboratory for Physical Sciences at Microscale, Department of Materials Science & Engineering & Collaborative Innovation Center of Suzhou Nano Science and Technology, University of Science and Technology of China, Hefei, 230026, China
| | - Jiajia Zhou
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Ruohong Shi
- Hefei National Laboratory for Physical Sciences at Microscale, Department of Materials Science & Engineering & Collaborative Innovation Center of Suzhou Nano Science and Technology, University of Science and Technology of China, Hefei, 230026, China
| | - Huihui Wu
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Ruhui Chen
- Hefei National Laboratory for Physical Sciences at Microscale, Department of Materials Science & Engineering & Collaborative Innovation Center of Suzhou Nano Science and Technology, University of Science and Technology of China, Hefei, 230026, China
| | - Beichen Duan
- Hefei National Laboratory for Physical Sciences at Microscale, Department of Materials Science & Engineering & Collaborative Innovation Center of Suzhou Nano Science and Technology, University of Science and Technology of China, Hefei, 230026, China
| | - Guoliang Xia
- Hefei National Laboratory for Physical Sciences at Microscale, Department of Materials Science & Engineering & Collaborative Innovation Center of Suzhou Nano Science and Technology, University of Science and Technology of China, Hefei, 230026, China
| | - Pengping Xu
- Hefei National Laboratory for Physical Sciences at Microscale, Department of Materials Science & Engineering & Collaborative Innovation Center of Suzhou Nano Science and Technology, University of Science and Technology of China, Hefei, 230026, China
| | - Hui Wang
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Shu Zhou
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Chengming Wang
- Hefei National Laboratory for Physical Sciences at Microscale, Department of Materials Science & Engineering & Collaborative Innovation Center of Suzhou Nano Science and Technology, University of Science and Technology of China, Hefei, 230026, China
| | - Haibao Wang
- Radiology Department of the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Zhen Guo
- Anhui Key Laboratory for Cellular Dynamics and Chemical Biology, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Qianwang Chen
- Hefei National Laboratory for Physical Sciences at Microscale, Department of Materials Science & Engineering & Collaborative Innovation Center of Suzhou Nano Science and Technology, University of Science and Technology of China, Hefei, 230026, China
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| |
Collapse
|
736
|
Tao W, Ji X, Xu X, Islam MA, Li Z, Chen S, Saw PE, Zhang H, Bharwani Z, Guo Z, Shi J, Farokhzad OC. Antimonene Quantum Dots: Synthesis and Application as Near-Infrared Photothermal Agents for Effective Cancer Therapy. Angew Chem Int Ed Engl 2017; 56:11896-11900. [PMID: 28640986 PMCID: PMC5608550 DOI: 10.1002/anie.201703657] [Citation(s) in RCA: 299] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 05/19/2017] [Indexed: 12/17/2022]
Abstract
Photothermal therapy (PTT) has shown significant potential for cancer therapy. However, developing nanomaterials (NMs)-based photothermal agents (PTAs) with satisfactory photothermal conversion efficacy (PTCE) and biocompatibility remains a key challenge. Herein, a new generation of PTAs based on two-dimensional (2D) antimonene quantum dots (AMQDs) was developed by a novel liquid exfoliation method. Surface modification of AMQDs with polyethylene glycol (PEG) significantly enhanced both biocompatibility and stability in physiological medium. The PEG-coated AMQDs showed a PTCE of 45.5 %, which is higher than many other NMs-based PTAs such as graphene, Au, MoS2 , and black phosphorus (BP). The AMQDs-based PTAs also exhibited a unique feature of NIR-induced rapid degradability. Through both in vitro and in vivo studies, the PEG-coated AMQDs demonstrated notable NIR-induced tumor ablation ability. This work is expected to expand the utility of 2D antimonene (AM) to biomedical applications through the development of an entirely novel PTA platform.
Collapse
Affiliation(s)
- Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Xiaoyuan Ji
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoding Xu
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Mohammad Ariful Islam
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Zhongjun Li
- Shenzhen Engineering Laboratory of Phosphorene and Optoelectronics, SZU-NUS Collaborative Innovation Center for Optoelectronic Science and Technology, and, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, 518060, China
| | - Si Chen
- Shenzhen Engineering Laboratory of Phosphorene and Optoelectronics, SZU-NUS Collaborative Innovation Center for Optoelectronic Science and Technology, and, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, 518060, China
| | - Phei Er Saw
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Han Zhang
- Shenzhen Engineering Laboratory of Phosphorene and Optoelectronics, SZU-NUS Collaborative Innovation Center for Optoelectronic Science and Technology, and, Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, 518060, China
| | - Zameer Bharwani
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Zilei Guo
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Omid C Farokhzad
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| |
Collapse
|
737
|
Fernandes RS, de Aguiar Ferreira C, Soares DCF, Maffione AM, Townsend DM, Rubello D, de Barros ALB. The role of radionuclide probes for monitoring anti-tumor drugs efficacy: A brief review. Biomed Pharmacother 2017; 95:469-476. [PMID: 28865367 DOI: 10.1016/j.biopha.2017.08.079] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 08/17/2017] [Accepted: 08/20/2017] [Indexed: 02/06/2023] Open
Abstract
Despite recent advances in the development of new therapeutic agents and diagnostic imaging modalities, cancer is still one of the main causes of death worldwide. A better understanding of the molecular signature of cancer has promoted the development of a new generation of anti-cancer drugs and diagnostic agents that specifically target molecular components such as genes, ligands, receptors and signaling pathways. However, intrinsic heterogeneity of tumors has hampered the overall success of target therapies even among patients with similar tumor types but unpredictable different responses to therapy. In this sense, post-treatment response monitoring becomes indispensable and nuclear medicine imaging modalities could provide the tools for an early indication of therapeutic efficacy. Herein, we briefly discuss the current role of PET and SPECT imaging in monitoring cancer therapy together with an update on the current radiolabeled probes that are currently investigated for tumor therapy response assessment.
Collapse
Affiliation(s)
- Renata Salgado Fernandes
- Laboratório de radioisótopos, Departamento de análises Clinicas, Universidade Federal de Minas Gerais (UFMG), Avenida Presidente Antônio Carlos, 6627, Belo Horizonte, Minas Gerais, Brazil
| | | | - Daniel Cristian Ferreira Soares
- Laboratório de Bioengenharia, Universidade Federal de Itajubá (UNIFEI), Rua Irmã Ivone Drumond, 200, Itabira, Minas Gerais, Brazil
| | - Anna Margherita Maffione
- Department of Nuclear Medicine, Radiology, Medical Physics and Clinical Pathology, Santa Maria della Misericordia Hospital, Rovigo, Italy
| | - Danyelle M Townsend
- Department of Drug Discovery and Pharmaceutical Sciences, Medical University of South Carolina, USA
| | - Domenico Rubello
- Department of Nuclear Medicine, Radiology, Medical Physics and Clinical Pathology, Santa Maria della Misericordia Hospital, Rovigo, Italy.
| | - André Luís Branco de Barros
- Laboratório de radioisótopos, Departamento de análises Clinicas, Universidade Federal de Minas Gerais (UFMG), Avenida Presidente Antônio Carlos, 6627, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
738
|
Wang J, Tao W, Chen X, Farokhzad OC, Liu G. Emerging Advances in Nanotheranostics with Intelligent Bioresponsive Systems. Theranostics 2017; 7:3915-3919. [PMID: 29109787 PMCID: PMC5667414 DOI: 10.7150/thno.21317] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Accepted: 07/09/2017] [Indexed: 12/29/2022] Open
Abstract
The confluence of therapeutics and diagnostics based on the advantages of nanotechnology offers significant opportunities for personalized and precision medicine. Intelligent nanotheranostics based on bioresponsive systems have recently emerged and offer the promise of high specificity and efficiency via "on-demand" activation of both therapeutic and diagnostic capabilities.
Collapse
Affiliation(s)
- Junqing Wang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, Fujian 361102, China
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland 20892, United States
| | - Omid C. Farokhzad
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - 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, Fujian 361102, China
| |
Collapse
|
739
|
Jiang Y, Cui D, Fang Y, Zhen X, Upputuri PK, Pramanik M, Ding D, Pu K. Amphiphilic semiconducting polymer as multifunctional nanocarrier for fluorescence/photoacoustic imaging guided chemo-photothermal therapy. Biomaterials 2017; 145:168-177. [PMID: 28866477 DOI: 10.1016/j.biomaterials.2017.08.037] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 08/16/2017] [Accepted: 08/18/2017] [Indexed: 12/11/2022]
Abstract
Chemo-photothermal nanotheranostics has the advantage of synergistic therapeutic effect, providing opportunities for optimized cancer therapy. However, current chemo-photothermal nanotheranostic systems generally comprise more than three components, encountering the potential issues of unstable nanostructures and unexpected conflicts in optical and biophysical properties among different components. We herein synthesize an amphiphilic semiconducting polymer (PEG-PCB) and utilize it as a multifunctional nanocarrier to simplify chemo-photothermal nanotheranostics. PEG-PCB has a semiconducting backbone that not only serves as the diagnostic component for near-infrared (NIR) fluorescence and photoacoustic (PA) imaging, but also acts as the therapeutic agent for photothermal therapy. In addition, the hydrophobic backbone of PEG-PCB provides strong hydrophobic and π-π interactions with the aromatic anticancer drug such as doxorubicin for drug encapsulation and delivery. Such a trifunctionality of PEG-PCB eventually results in a greatly simplified nanotheranostic system with only two components but multimodal imaging and therapeutic capacities, permitting effective NIR fluorescence/PA imaging guided chemo-photothermal therapy of cancer in living mice. Our study thus provides a molecular engineering approach to integrate essential properties into one polymer for multimodal nanotheranostics.
Collapse
Affiliation(s)
- Yuyan Jiang
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, 637459, Singapore
| | - Dong Cui
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, 637459, Singapore
| | - Yuan Fang
- State of Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xu Zhen
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, 637459, Singapore
| | - Paul Kumar Upputuri
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, 637459, Singapore
| | - Manojit Pramanik
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, 637459, Singapore
| | - Dan Ding
- State of Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Kanyi Pu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, 637459, Singapore.
| |
Collapse
|
740
|
Li X, Kim CY, Lee S, Lee D, Chung HM, Kim G, Heo SH, Kim C, Hong KS, Yoon J. Nanostructured Phthalocyanine Assemblies with Protein-Driven Switchable Photoactivities for Biophotonic Imaging and Therapy. J Am Chem Soc 2017; 139:10880-10886. [DOI: 10.1021/jacs.7b05916] [Citation(s) in RCA: 253] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Xingshu Li
- Department
of Chemistry and Nano Science, Ewha Womans University, Seoul 120-750, Korea
| | - C-yoon Kim
- Department
of Medicine, School of Medicine, Konkuk University, Seoul 143-701, Korea
| | - Seunghyun Lee
- Department
of Electrical Engineering and Creative IT Engineering, POSTECH, Pohang 37673, Korea
| | - Dayoung Lee
- Department
of Chemistry and Nano Science, Ewha Womans University, Seoul 120-750, Korea
| | - Hyung-Min Chung
- Department
of Medicine, School of Medicine, Konkuk University, Seoul 143-701, Korea
| | - Gyoungmi Kim
- Department
of Chemistry and Nano Science, Ewha Womans University, Seoul 120-750, Korea
| | - Si-Hyun Heo
- Department
of Medicine, School of Medicine, Konkuk University, Seoul 143-701, Korea
| | - Chulhong Kim
- Department
of Electrical Engineering and Creative IT Engineering, POSTECH, Pohang 37673, Korea
| | - Ki-Sung Hong
- Department
of Medicine, School of Medicine, Konkuk University, Seoul 143-701, Korea
| | - Juyoung Yoon
- Department
of Chemistry and Nano Science, Ewha Womans University, Seoul 120-750, Korea
| |
Collapse
|
741
|
Tao W, Ji X, Xu X, Islam MA, Li Z, Chen S, Saw PE, Zhang H, Bharwani Z, Guo Z, Shi J, Farokhzad OC. Antimonene Quantum Dots: Synthesis and Application as Near-Infrared Photothermal Agents for Effective Cancer Therapy. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201703657] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Wei Tao
- Center for Nanomedicine and Department of Anesthesiology; Brigham and Women's Hospital; Harvard Medical School; Boston MA 02115 USA
| | - Xiaoyuan Ji
- Center for Nanomedicine and Department of Anesthesiology; Brigham and Women's Hospital; Harvard Medical School; Boston MA 02115 USA
- University of Chinese Academy of Sciences; Beijing 100049 China
| | - Xiaoding Xu
- Center for Nanomedicine and Department of Anesthesiology; Brigham and Women's Hospital; Harvard Medical School; Boston MA 02115 USA
| | - Mohammad Ariful Islam
- Center for Nanomedicine and Department of Anesthesiology; Brigham and Women's Hospital; Harvard Medical School; Boston MA 02115 USA
| | - Zhongjun Li
- Shenzhen Engineering Laboratory of Phosphorene and Optoelectronics; SZU-NUS Collaborative Innovation Center for Optoelectronic Science and Technology, and; Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province; Shenzhen University; Shenzhen 518060 China
| | - Si Chen
- Shenzhen Engineering Laboratory of Phosphorene and Optoelectronics; SZU-NUS Collaborative Innovation Center for Optoelectronic Science and Technology, and; Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province; Shenzhen University; Shenzhen 518060 China
| | - Phei Er Saw
- Center for Nanomedicine and Department of Anesthesiology; Brigham and Women's Hospital; Harvard Medical School; Boston MA 02115 USA
| | - Han Zhang
- Shenzhen Engineering Laboratory of Phosphorene and Optoelectronics; SZU-NUS Collaborative Innovation Center for Optoelectronic Science and Technology, and; Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province; Shenzhen University; Shenzhen 518060 China
| | - Zameer Bharwani
- Center for Nanomedicine and Department of Anesthesiology; Brigham and Women's Hospital; Harvard Medical School; Boston MA 02115 USA
| | - Zilei Guo
- Center for Nanomedicine and Department of Anesthesiology; Brigham and Women's Hospital; Harvard Medical School; Boston MA 02115 USA
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology; Brigham and Women's Hospital; Harvard Medical School; Boston MA 02115 USA
| | - Omid C. Farokhzad
- Center for Nanomedicine and Department of Anesthesiology; Brigham and Women's Hospital; Harvard Medical School; Boston MA 02115 USA
- King Abdulaziz University; Jeddah 21589 Saudi Arabia
| |
Collapse
|
742
|
Li R, Zheng K, Yuan C, Chen Z, Huang M. Be Active or Not: the Relative Contribution of Active and Passive Tumor Targeting of Nanomaterials. Nanotheranostics 2017; 1:346-357. [PMID: 29071198 PMCID: PMC5646738 DOI: 10.7150/ntno.19380] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 06/21/2017] [Indexed: 12/23/2022] Open
Abstract
Malignant tumor (cancer) remains as one of the deadliest diseases throughout the world, despite its overall mortality drops. Nanomaterials (NMs) have been widely studied as diagnostic and/or therapeutic agents for tumors. A feature of NMs, compared to small molecules, is that NMs can be concentrated passively in tumors through enhanced permeability and retention (EPR) effect. In the meantime, NMs can be engineered to target toward tumor specific markers in an active manner, e.g., receptor-mediated targeting. The relative contribution of the EPR effect and the receptor-mediated targeting to NM accumulation in tumor tissues has not been clearly defined yet. Here, we tackle this fundamental issue by reviewing previous studies. First, we summarize the current knowledge on these two tumor targeting strategies of NMs, and on how NMs arrive to tumors from blood circulation. We then demonstrate that contribution of the active and passive effects to total accumulation of NMs in tumors varies with time. Over time, the receptor-mediated targeting contributes more than the EPR effect with a ratio of 3 in the case of urokinase-type plasminogen activator receptor (uPAR)-mediated targeting and human serum albumin (HSA)-mediated EPR effect. Therefore, this review highlights the dynamics of active and passive targeting of NMs on their accumulation at tumor sites, and is valuable for future design of NMs in cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Rui Li
- Key Laboratory of Animal Immunology of the Ministry of Agriculture, Henan Provincial Key Laboratory of Animal Immunology, Henan Academy of Agricultural Sciences, Zhengzhou, Henan, 450002, China
| | - Ke Zheng
- Fuzhou University, Fuzhou, Fujian, 350116, China
| | - Cai Yuan
- Fuzhou University, Fuzhou, Fujian, 350116, China
| | - Zhuo Chen
- Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian, 350002, China
| | - Mingdong Huang
- Fuzhou University, Fuzhou, Fujian, 350116, China.,Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, Fujian, 350002, China
| |
Collapse
|