101
|
Improving safety of cancer immunotherapy via delivery technology. Biomaterials 2020; 265:120407. [PMID: 32992118 DOI: 10.1016/j.biomaterials.2020.120407] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 09/09/2020] [Accepted: 09/18/2020] [Indexed: 12/13/2022]
Abstract
Breakthroughs in molecular mechanisms underlying immune-suppressive tumor microenvironment and paradigm shifts in the cancer-immunity response cycle have profoundly changed the landscape of cancer immunotherapy. However, one of the challenges is to mitigate the serious side effects caused by systemic autoimmunity and autoinflammatory responses following immunotherapy. Thus, restraining the activation of the immune system in healthy tissues is highly desirable to address this problem. Bioengineering and delivery technologies provide a solution to the issue. In this Review, we first introduce immune-related adverse effects of main immunotherapies and the underlying mechanisms, summarize strategies of designingde bioengineering and delivery systems to reduce their immunotoxicities, and highlight the importance of the development of immunotoxicity-related animal models.
Collapse
|
102
|
Abdou P, Wang Z, Chen Q, Chan A, Zhou DR, Gunadhi V, Gu Z. Advances in engineering local drug delivery systems for cancer immunotherapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2020; 12:e1632. [PMID: 32255276 PMCID: PMC7725287 DOI: 10.1002/wnan.1632] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/03/2020] [Accepted: 03/04/2020] [Indexed: 12/13/2022]
Abstract
Cancer immunotherapy aims to leverage the immune system to suppress the growth of tumors and to inhibit metastasis. The recent promising clinical outcomes associated with cancer immunotherapy have prompted research and development efforts towards enhancing the efficacy of immune checkpoint blockade, cancer vaccines, cytokine therapy, and adoptive T cell therapy. Advancements in biomaterials, nanomedicine, and micro-/nano-technology have facilitated the development of enhanced local delivery systems for cancer immunotherapy, which can enhance treatment efficacy while minimizing toxicity. Furthermore, locally administered cancer therapies that combine immunotherapy with chemotherapy, radiotherapy, or phototherapy have the potential to achieve synergistic antitumor effects. Herein, the latest studies on local delivery systems for cancer immunotherapy are surveyed, with an emphasis on the therapeutic benefits associated with the design of biomaterials and nanomedicines. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Peter Abdou
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Zejun Wang
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Qian Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren’ai Road, Suzhou, 215123, Jiangsu, PR China
| | - Amanda Chan
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Daojia R. Zhou
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| | - Vivienne Gunadhi
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| | - Zhen Gu
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, California NanoSystems Institute, and Center for Minimally Invasive Therapeutics, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
103
|
Shen X, Zhou Z, Qi D, Li Y, Zeng Z, He X, Xia X, Zhu C, Feng X. Highly uniform self-assembled microspheres from single macromolecule self-recognition for enhanced cancer immunotherapy. Chem Commun (Camb) 2020; 56:9190-9193. [PMID: 32661548 DOI: 10.1039/d0cc04255b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Self-assembled highly uniform microspheres from star-shaped biocompatible polymers have been prepared as a local delivery system for co-loading different immunoagents together and ensuring their release in different stages, so as to realize effective cooperative immunotherapy and minimize systemic side effects.
Collapse
Affiliation(s)
- Xin Shen
- Department of Pharmaceutical Sciences, Chongqing University, Chongqing, 401331, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
104
|
Jiang Y, Krishnan N, Heo J, Fang RH, Zhang L. Nanoparticle-hydrogel superstructures for biomedical applications. J Control Release 2020; 324:505-521. [PMID: 32464152 PMCID: PMC7429280 DOI: 10.1016/j.jconrel.2020.05.041] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 05/23/2020] [Accepted: 05/25/2020] [Indexed: 12/13/2022]
Abstract
The incorporation of nanoparticles into hydrogels yields novel superstructures that have become increasingly popular in biomedical research. Each component of these nanoparticle-hydrogel superstructures can be easily modified, resulting in platforms that are highly tunable and inherently multifunctional. The advantages of the nanoparticle and hydrogel constituents can be synergistically combined, enabling these superstructures to excel in scenarios where employing each component separately may have suboptimal outcomes. In this review, the synthesis and fabrication of different nanoparticle-hydrogel superstructures are discussed, followed by an overview of their use in a range of applications, including drug delivery, detoxification, immune modulation, and tissue engineering. Overall, these platforms hold significant clinical potential, and it is envisioned that future development along these lines will lead to unique solutions for addressing areas of pressing medical need.
Collapse
Affiliation(s)
- Yao Jiang
- Department of NanoEngineering, Chemical Engineering Program, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Nishta Krishnan
- Department of NanoEngineering, Chemical Engineering Program, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Jiyoung Heo
- Department of NanoEngineering, Chemical Engineering Program, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Ronnie H Fang
- Department of NanoEngineering, Chemical Engineering Program, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA.
| | - Liangfang Zhang
- Department of NanoEngineering, Chemical Engineering Program, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
105
|
Wu M, Zheng D, Zhang D, Yu P, Peng L, Chen F, Lin Z, Cai Z, Li J, Wei Z, Lin X, Liu J, Liu X. Converting Immune Cold into Hot by Biosynthetic Functional Vesicles to Boost Systematic Antitumor Immunity. iScience 2020; 23:101341. [PMID: 32683314 PMCID: PMC7371908 DOI: 10.1016/j.isci.2020.101341] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/01/2020] [Accepted: 07/02/2020] [Indexed: 01/01/2023] Open
Abstract
Immune cold tumor characterized by low immunogenicity, insufficient and exhausted tumor-infiltrating lymphocytes, and immunosuppressive microenvironment is the main bottleneck responsible for low patient response rate of immune checkpoint blockade. Here, we developed biosynthetic functional vesicles (BFVs) to convert immune cold into hot through overcoming hypoxia, inducing immunogenic cell death, and immune checkpoint inhibition. The BFVs present PD1 and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) on the surface, whereas load catalase into their inner core. The TRAIL can specifically induce immunogenic death of cancer cells to initiate immune response, which is further synergistically strengthened by blocking PD1/PDL1 checkpoint signal through ectogenic PD1 proteins on BFVs. The catalase can produce O2 to overcome tumor hypoxia, in turn to increase infiltration of effector T cells while deplete immunosuppressive cells in tumor. The BFVs elicit robust and systematic antitumor immunity, as demonstrated by significant regression of tumor growth, prevention of abscopal tumors, and excellent inhibition of lung metastasis.
Collapse
Affiliation(s)
- Ming Wu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China; CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, P. R. China; Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China
| | - Dongye Zheng
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China; CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, P. R. China
| | - Da Zhang
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China; Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China
| | - Peiwen Yu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China; Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China
| | - Lianli Peng
- College of Engineering and Computer Science, The Australian National University, Canberra 2601, Australia
| | - Feng Chen
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China; CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, P. R. China
| | - Ziguo Lin
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China
| | - Zhixiong Cai
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China; Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China
| | - Jiong Li
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China
| | - Zuwu Wei
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China; Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China
| | - Xinyi Lin
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China; Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China
| | - Jingfeng Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China; CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, P. R. China; Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China; Department of Translational Medicine, Xiamen Institute of Rare Earth Materials, Chinese Academy of Sciences, Xiamen 361024, P. R. China.
| | - Xiaolong Liu
- The United Innovation of Mengchao Hepatobiliary Technology Key Laboratory of Fujian Province, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou 350025, P. R. China; CAS Key Laboratory of Design and Assembly of Functional Nanostructures, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, P. R. China; Mengchao Med-X Center, Fuzhou University, Fuzhou 350116, P. R. China; Department of Translational Medicine, Xiamen Institute of Rare Earth Materials, Chinese Academy of Sciences, Xiamen 361024, P. R. China.
| |
Collapse
|
106
|
Pang X, Liang S, Wang T, Yu S, Yang R, Hou T, Liu Y, He C, Zhang N. Engineering Thermo-pH Dual Responsive Hydrogel for Enhanced Tumor Accumulation, Penetration, and Chemo-Protein Combination Therapy. Int J Nanomedicine 2020; 15:4739-4752. [PMID: 32753862 PMCID: PMC7342477 DOI: 10.2147/ijn.s253990] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 06/01/2020] [Indexed: 01/20/2023] Open
Abstract
Purpose Combined chemotherapeutic drug and protein drug has been a widely employed strategy for tumor treatment. To realize both tumor accumulation and deep tumor penetration for drugs with different pharmacokinetics, we propose a structure-transformable, thermo-pH dual responsive co-delivery system to co-load granzyme B/docetaxel (GrB/DTX). Methods Thermo-sensitive hydrogels based on diblock copolymers (mPEG-b-PELG) were synthesized through ring opening polymerization. GrB/DTX mini micelles (GDM) was developed by co-loading these two drugs in pH-sensitive mini micelles, and the GDM-incorporated thermo-sensitive hydrogel (GDMH) was constructed. The thermo-induced gelation behavior of diblock copolymers and the physiochemical properties of GDMH were characterized. GDMH degradation and deep tumor penetration of released mini micelles were confirmed. The pH-sensitive disassembly and lysosomal escape abilities of released mini micelles were evaluated. In vitro cytotoxicity was studied using MTT assays and the in vivo antitumor efficacy study was evaluated in B16-bearing C57BL/6 mice. Results GDMH was gelatinized at body temperature and can be degraded by proteinase to release mini micelles. The mini micelles incorporated in GDMH can achieve deep tumor penetration and escape from lysosomes to release GrB and DTX. MTT results showed that maximum synergistic antitumor efficacy of GrB and DTX was observed at mass ratio of 1:100. Our in vivo antitumor efficacy study showed that GDMH inhibited tumor growth in the subcutaneous tumor model and in the post-surgical recurrence model. Conclusion The smart-designed transformable GDMH can facilitate tumor accumulation, deep tumor penetration, and rapid drug release to achieve synergistic chemo-protein therapy.
Collapse
Affiliation(s)
- Xiuping Pang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong Province 250012, People's Republic of China
| | - Shuang Liang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong Province 250012, People's Republic of China
| | - Tianqi Wang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong Province 250012, People's Republic of China
| | - Shuangjiang Yu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, People's Republic of China
| | - Rui Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong Province 250012, People's Republic of China
| | - Teng Hou
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong Province 250012, People's Republic of China
| | - Yongjun Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong Province 250012, People's Republic of China
| | - Chaoliang He
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, People's Republic of China
| | - Na Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong Province 250012, People's Republic of China
| |
Collapse
|
107
|
Chen H, Cheng H, Wu W, Li D, Mao J, Chu C, Liu G. The blooming intersection of transcatheter hepatic artery chemoembolization and nanomedicine. CHINESE CHEM LETT 2020; 31:1375-1381. [DOI: 10.1016/j.cclet.2020.03.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
108
|
Meng Q, Cong H, Hu H, Xu FJ. Rational design and latest advances of codelivery systems for cancer therapy. Mater Today Bio 2020; 7:100056. [PMID: 32510051 PMCID: PMC7264083 DOI: 10.1016/j.mtbio.2020.100056] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/25/2020] [Accepted: 04/29/2020] [Indexed: 01/06/2023] Open
Abstract
Current treatments have limited effectiveness in treating tumors. The combination of multiple drugs or treatment strategies is widely studied to improve therapeutic effect and reduce adverse effects of cancer therapy. The codelivery system is the key to realize combined therapies. It is necessary to design and construct different codelivery systems in accordance with the variable structures and properties of cargoes and vectors. This review presented the typical design considerations about codelivery vectors for cancer therapy and described the current state of codelivery systems from two aspects: different types of vectors and collaborative treatment strategies. The commonly used loading methods of cargoes into the vectors, including physical and chemical processes, are discussed in detail. Finally, we outline the challenges and perspectives about the improvement of codelivery systems.
Collapse
Affiliation(s)
- Q.Y. Meng
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, Qingdao University, Qingdao, 266071, China
| | - H.L. Cong
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, Qingdao University, Qingdao, 266071, China
| | - H. Hu
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, Qingdao University, Qingdao, 266071, China
| | - F.-J. Xu
- Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
109
|
Cossío FP, Esteller M, Berdasco M. Towards a more precise therapy in cancer: Exploring epigenetic complexity. Curr Opin Chem Biol 2020; 57:41-49. [PMID: 32480315 DOI: 10.1016/j.cbpa.2020.04.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 04/13/2020] [Indexed: 12/21/2022]
Abstract
A plethora of preclinical evidences suggests that pharmacological targeting of epigenetic dysregulation is a potent strategy to combat human diseases. Nevertheless, the implementation of epidrugs in clinical practice is very scarce and mainly limited to haematological malignancies. In this review, we discuss cutting-edge strategies to foster the chemical design, the biological rationale and the clinical trial development of epidrugs. Specifically, we focus on the development of dual hybrids to exploit multitargeting of key epigenetic molecules deregulated in cancer; the study of epigenetic-synthetic lethality interactions as a mechanism to address loss-of-function mutations, and the combination of epidrugs with other therapies such as immunotherapy to avoid acquired chemoresistance and increase therapy sensitivity. By exploring these challenges, among others, the field of epigenetic chemical biology will increase its potential for clinical benefit, and more effective strategies targeting the aberrant epigenome in cancer are likely to be developed both in haematological and solid tumours.
Collapse
Affiliation(s)
- Fernando P Cossío
- Kimika Fakultatea, Kimika Organikoa I Saila, Universidad del País Vasco - Euskal Herriko Unibertsitaea, and Donostia International Physics Center (DIPC), San Sebastián-Donostia, Spain; Centro de Innovación en Química Avanzada (ORFEO-CINQA), Spain
| | - Manel Esteller
- Cancer Epigenetics Group, Cancer and Leukemia Epigenetics and Biology Program (PEBCL), Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain; Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Catalonia, Spain
| | - María Berdasco
- Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Catalonia, Spain; Epigenetic Therapies Group, Experimental and Clinical Hematology Program (PHEC), Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Catalonia, Spain.
| |
Collapse
|
110
|
Hsu RS, Fang JH, Shen WT, Sheu YC, Su CK, Chiang WH, Hu SH. Injectable DNA-architected nanoraspberry depot-mediated on-demand programmable refilling and release drug delivery. NANOSCALE 2020; 12:11153-11164. [PMID: 32400827 DOI: 10.1039/d0nr01185a] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Drug delivery depots boosting a local concentration of therapeutic agents have received great attention in clinical applications due to their low occurrence of side effects and high therapeutic efficacy. However, once the payload is exhausted, the local drug concentration will be lower than the therapeutic window. To address this issue, an injectable double-strand deoxyribonucleic acid (DNA)-architected nanoraspberry depot (DNR-depot) was developed that can refill doxorubicin (Dox, an anticancer drug) from the blood and remotely control drug release on demand. The large porous surface on a uniform nanoraspberry (NR) filled covalently with DNA serves as a Dox sponge-like refilling reservoir, and the NR serves as a magnetic electrical absorber. Via the strong affinity between Dox and DNA molecules, the refilling process of Dox can be achieved on DNR-depot both in vitro and in vivo. Upon high-frequency magnetic field (HFMF) treatment, the remotely triggered release of Dox is actuated by the dissociation of Dox and DNA molecules, facilitating an approximately 800% improvement in drug concentration at the tumor site compared to free Dox injection alone. Furthermore, the cycles of refilling and release can be carried out more than 3 times in vivo within 21 days. The combination of refilling and HFMF-programmable Dox release in tumors via DNR-depot can effectively inhibit tumor growth for 30 days.
Collapse
Affiliation(s)
- Ru-Siou Hsu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 300, Taiwan.
| | - Jen-Hung Fang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 300, Taiwan.
| | - Wei-Ting Shen
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 300, Taiwan.
| | - Yu-Chen Sheu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 300, Taiwan.
| | - Cheng-Kuan Su
- Department of Chemistry, National Chung Hsing University, Taichung 402, Taiwan
| | - Wen-Hsuan Chiang
- Department of Chemical Engineering, National Chung Hsing University, Taichung 402, Taiwan
| | - Shang-Hsiu Hu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 300, Taiwan.
| |
Collapse
|
111
|
Jin J, Zhao Q. Engineering nanoparticles to reprogram radiotherapy and immunotherapy: recent advances and future challenges. J Nanobiotechnology 2020; 18:75. [PMID: 32408880 PMCID: PMC7227304 DOI: 10.1186/s12951-020-00629-y] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 05/07/2020] [Indexed: 02/06/2023] Open
Abstract
Nanoparticles (NPs) have been increasingly studied for radiosensitization. The principle of NPs radio-enhancement is to use high-atomic number NPs (e.g. gold, hafnium, bismuth and gadolinium) or deliver radiosensitizing substances, such as cisplatin and selenium. Nowadays, cancer immunotherapy is emerged as a promising treatment and immune checkpoint regulation has a potential property to improve clinical outcomes in cancer immunotherapy. Furthermore, NPs have been served as an ideal platform for immunomodulator system delivery. Owing to enhanced permeability and retention (EPR) effect, modified-NPs increase the targeting and retention of antibodies in target cells. The purpose of this review is to highlight the latest progress of nanotechnology in radiotherapy (RT) and immunotherapy, as well as combining these three strategies in cancer treatment. Overall, nanomedicine as an effective strategy for RT can significantly enhance the outcome of immunotherapy response and might be beneficial for clinical transformation.
Collapse
Affiliation(s)
- Jing Jin
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
| | - Qijie Zhao
- Laboratory of Molecular Pharmacology, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China. .,Department of Pathophysiology, College of Basic Medical Science, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China. .,South Sichuan Institute of Translational Medicine, Luzhou, 646000, Sichuan, People's Republic of China.
| |
Collapse
|
112
|
Qu M, Jiang X, Zhou X, Wang C, Wu Q, Ren L, Zhu J, Zhu S, Tebon P, Sun W, Khademhosseini A. Stimuli-Responsive Delivery of Growth Factors for Tissue Engineering. Adv Healthc Mater 2020; 9:e1901714. [PMID: 32125786 PMCID: PMC7189772 DOI: 10.1002/adhm.201901714] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 02/03/2020] [Indexed: 02/05/2023]
Abstract
Growth factors (GFs) play a crucial role in directing stem cell behavior and transmitting information between different cell populations for tissue regeneration. However, their utility as therapeutics is limited by their short half-life within the physiological microenvironment and significant side effects caused by off-target effects or improper dosage. "Smart" materials that can not only sustain therapeutic delivery over a treatment period but also facilitate on-demand release upon activation are attracting significant interest in the field of GF delivery for tissue engineering. Three properties are essential in engineering these "smart" materials: 1) the cargo vehicle protects the encapsulated therapeutic; 2) release is targeted to the site of injury; 3) cargo release can be modulated by disease-specific stimuli. The aim of this review is to summarize the current research on stimuli-responsive materials as intelligent vehicles for controlled GF delivery; Five main subfields of tissue engineering are discussed: skin, bone and cartilage, muscle, blood vessel, and nerve. Challenges in achieving such "smart" materials and perspectives on future applications of stimuli-responsive GF delivery for tissue regeneration are also discussed.
Collapse
Affiliation(s)
- Moyuan Qu
- Department of Bioengineering, California NanoSystems Institute and Center for Minimally Invasive Therapeutics (C-MIT) University of California, Los Angeles, Los Angeles, CA 90095, USA
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xing Jiang
- Department of Bioengineering, California NanoSystems Institute and Center for Minimally Invasive Therapeutics (C-MIT) University of California, Los Angeles, Los Angeles, CA 90095, USA
- School of Nursing, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xingwu Zhou
- Department of Bioengineering, California NanoSystems Institute and Center for Minimally Invasive Therapeutics (C-MIT) University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Chemical and Biomolecular Engineering, University of California-Los Angeles, Los Angeles, CA 90095, USA
| | - Canran Wang
- Department of Bioengineering, California NanoSystems Institute and Center for Minimally Invasive Therapeutics (C-MIT) University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qingzhi Wu
- Department of Bioengineering, California NanoSystems Institute and Center for Minimally Invasive Therapeutics (C-MIT) University of California, Los Angeles, Los Angeles, CA 90095, USA
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, China
| | - Li Ren
- Department of Bioengineering, California NanoSystems Institute and Center for Minimally Invasive Therapeutics (C-MIT) University of California, Los Angeles, Los Angeles, CA 90095, USA
- School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Jixiang Zhu
- Department of Bioengineering, California NanoSystems Institute and Center for Minimally Invasive Therapeutics (C-MIT) University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Biomedical Engineering, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Songsong Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Peyton Tebon
- Department of Bioengineering, California NanoSystems Institute and Center for Minimally Invasive Therapeutics (C-MIT) University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Wujin Sun
- Department of Bioengineering, California NanoSystems Institute and Center for Minimally Invasive Therapeutics (C-MIT) University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ali Khademhosseini
- Department of Bioengineering, California NanoSystems Institute and Center for Minimally Invasive Therapeutics (C-MIT) University of California, Los Angeles, Los Angeles, CA 90095, USA
- Department of Chemical and Biomolecular Engineering, University of California-Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, Department of Radiology University of California-Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
113
|
Nazir R, Parida D, Guex AG, Rentsch D, Zarei A, Gooneie A, Salmeia KA, Yar KM, Alihosseini F, Sadeghpour A, Gaan S. Structurally Tunable pH-responsive Phosphine Oxide Based Gels by Facile Synthesis Strategy. ACS APPLIED MATERIALS & INTERFACES 2020; 12:7639-7649. [PMID: 31972075 DOI: 10.1021/acsami.9b22808] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Design and synthesis of nanostructured responsive gels have attracted increasing attention, particularly in the biomedical domain. Polymer chain configurations and nanodomain sizes within the network can be used to steer their functions as drug carriers. Here, a catalyst-free facile one-step synthesis strategy is reported for the design of pH-responsive gels and controlled structures in nanoscale. Transparent and impurity free gels were directly synthesized from trivinylphosphine oxide (TVPO) and cyclic secondary diamine monomers via Michael addition polymerization under mild conditions. NMR analysis confirmed the consumption of all TVPO and the absence of side products, thereby eliminating post purification steps. The small-angle X-ray scattering (SAXS) elucidates the nanoscale structural features in gels, that is, it demonstrates the presence of collapsed nanodomains within gel networks and it was possible to tune the size of these domains by varying the amine monomers and the nature of the solvent. The fabricated gels demonstrate structure tunability via solvent-polymer interactions and pH specific drug release behavior. Three different anionic dyes (acid blue 80, acid blue 90, and fluorescein) of varying size and chemistry were incorporated into the hydrogel as model drugs and their release behavior was studied. Compared to acidic pH, a higher and faster release of acid blue 80 and fluorescein was observed at pH 10, possibly because of their increased solubility in alkaline pH. In addition, their release in phosphate buffered saline (PBS) and simulated body fluid (SBF) matrix was positively influenced by the ionic interaction with positively charged metal ions. In the case of hydrogel containing acid blue 90 a very low drug release (<1%) was observed, which is due to the reaction of its accessible free amino group with the vinyl groups of the TVPO. In vitro evaluation of the prepared hydrogel using human dermal fibroblasts indicates no cytotoxic effects, warranting further research for biomedical applications. Our strategy of such gel synthesis lays the basis for the design of other gel-based functional materials.
Collapse
Affiliation(s)
- Rashid Nazir
- Laboratory of Advanced Fibers , Empa, Swiss Federal Laboratories for Materials Science and Technology , Lerchenfeldstrasse 5 , CH-9014 St. Gallen , Switzerland
| | - Dambarudhar Parida
- Laboratory of Advanced Fibers , Empa, Swiss Federal Laboratories for Materials Science and Technology , Lerchenfeldstrasse 5 , CH-9014 St. Gallen , Switzerland
| | - Anne Géraldine Guex
- Laboratory for Biointerfaces and Laboratory for Biomimetic Membranes and Textiles , Empa, Swiss Federal Laboratories for Materials Science and Technology , Lerchenfeldstrasse 5 , CH-9014 St. Gallen , Switzerland
| | - Daniel Rentsch
- Laboratory for Functional Polymers , Empa, Swiss Federal Laboratories for Materials Science and Technology , Überlandstrasse 129 , 8600 Dübendorf , Switzerland
| | - Afsaneh Zarei
- Department of Textile Engineering , Isfahan University of Technology , Isfahan , 84156-83111 , Iran
| | - Ali Gooneie
- Laboratory of Advanced Fibers , Empa, Swiss Federal Laboratories for Materials Science and Technology , Lerchenfeldstrasse 5 , CH-9014 St. Gallen , Switzerland
| | - Khalifah A Salmeia
- Laboratory of Advanced Fibers , Empa, Swiss Federal Laboratories for Materials Science and Technology , Lerchenfeldstrasse 5 , CH-9014 St. Gallen , Switzerland
| | - Kevin M Yar
- Laboratory of Advanced Fibers , Empa, Swiss Federal Laboratories for Materials Science and Technology , Lerchenfeldstrasse 5 , CH-9014 St. Gallen , Switzerland
| | - Farzaneh Alihosseini
- Department of Textile Engineering , Isfahan University of Technology , Isfahan , 84156-83111 , Iran
| | - Amin Sadeghpour
- Center for X-Ray Analytics , Empa, Swiss Federal Laboratories for Materials Science and Technology , Lerchenfeldstrasse 5 , CH-9014 St. Gallen , Switzerland
| | - Sabyasachi Gaan
- Laboratory of Advanced Fibers , Empa, Swiss Federal Laboratories for Materials Science and Technology , Lerchenfeldstrasse 5 , CH-9014 St. Gallen , Switzerland
| |
Collapse
|
114
|
Liu Y, Guo J, Huang L. Modulation of tumor microenvironment for immunotherapy: focus on nanomaterial-based strategies. Am J Cancer Res 2020; 10:3099-3117. [PMID: 32194857 PMCID: PMC7053194 DOI: 10.7150/thno.42998] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 01/19/2020] [Indexed: 02/07/2023] Open
Abstract
Recent advances in the field of immunotherapy have profoundly opened up the potential for improved cancer therapy and reduced side effects. However, the tumor microenvironment (TME) is highly immunosuppressive, therefore, clinical outcomes of currently available cancer immunotherapy are still poor. Recently, nanomaterial-based strategies have been developed to modulate the TME for robust immunotherapeutic responses. In this review, the immunoregulatory cell types (cells relating to the regulation of immune responses) inside the TME in terms of stimulatory and suppressive roles are described, and the technologies used to identify and quantify these cells are provided. In addition, recent examples of nanomaterial-based cancer immunotherapy are discussed, with particular emphasis on those designed to overcome barriers caused by the complexity and diversity of TME.
Collapse
|
115
|
Zuo L, Ding J, Li C, Lin F, Chen PR, Wang P, Lu G, Zhang J, Huang LL, Xie HY. Coordinating bioorthogonal reactions with two tumor-microenvironment-responsive nanovehicles for spatiotemporally controlled prodrug activation. Chem Sci 2020; 11:2155-2160. [PMID: 34123305 PMCID: PMC8150104 DOI: 10.1039/c9sc05036a] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 01/09/2020] [Indexed: 02/04/2023] Open
Abstract
Precise activation of prodrugs in tumor tissues is critical to ensuring specific antitumor efficacy, meanwhile reducing the serious adverse effects. Here, a spatiotemporally controlled prodrug activation strategy was provided by integrating the inverse electron demand Diels-Alder (IEDDA) reaction with two tumor-microenvironment-responsive nanovehicles. The prodrug (Dox-TCO) and [4-(6-methyl-1,2,4,5-tetrazin-3-yl)phenyl]methanamine (Tz) were separately camouflaged into low pH and matrix metalloproteinase 2 (MMP-2) sensitive micellar nanoparticles. After systemic administration, only in the tumor tissues could both the nanovehicles dissociate via responding to two special tumor microenvironments, with Dox-TCO and Tz released and then immediately triggering the prodrug activation through the IEDDA reaction. The hierarchically regulated and locally confined Dox liberation led to dramatically decreased side-effects that were much lower than those of the clinical Doxorubicin Hydrochloride Liposomal Injection (Doxil), while the antitumor therapeutic effect was potent.
Collapse
Affiliation(s)
- Liping Zuo
- School of Life Science, Beijing Institute of Technology No. 5 South Zhong Guan Cun Street Beijing 100081 China
| | - Jingjing Ding
- School of Life Science, Beijing Institute of Technology No. 5 South Zhong Guan Cun Street Beijing 100081 China
| | - Changkun Li
- Shimadzu (China) Co., Ltd Beijing Branch Beijing 100020 PR China
| | - Feng Lin
- Peking-Tsinghua Center for Life Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University Beijing 100871 China
| | - Peng R Chen
- Peking-Tsinghua Center for Life Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University Beijing 100871 China
| | - Peilin Wang
- School of Life Science, Beijing Institute of Technology No. 5 South Zhong Guan Cun Street Beijing 100081 China
| | - Guihong Lu
- School of Life Science, Beijing Institute of Technology No. 5 South Zhong Guan Cun Street Beijing 100081 China
| | - Jinfeng Zhang
- School of Life Science, Beijing Institute of Technology No. 5 South Zhong Guan Cun Street Beijing 100081 China
| | - Li-Li Huang
- School of Life Science, Beijing Institute of Technology No. 5 South Zhong Guan Cun Street Beijing 100081 China
| | - Hai-Yan Xie
- School of Life Science, Beijing Institute of Technology No. 5 South Zhong Guan Cun Street Beijing 100081 China
| |
Collapse
|
116
|
Han S, Huang K, Gu Z, Wu J. Tumor immune microenvironment modulation-based drug delivery strategies for cancer immunotherapy. NANOSCALE 2020; 12:413-436. [PMID: 31829394 DOI: 10.1039/c9nr08086d] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The past years have witnessed promising clinical feedback for anti-cancer immunotherapies, which have become one of the hot research topics; however, they are limited by poor delivery kinetics, narrow patient response profiles, and systemic side effects. To the best of our knowledge, the development of cancer is highly associated with the immune system, especially the tumor immune microenvironment (TIME). Based on the comprehensive understanding of the complexity and diversity of TIME, drug delivery strategies focused on the modulation of TIME can be of great significance for directing and improving cancer immunotherapy. This review highlights the TIME modulation in cancer immunotherapy and summarizes the versatile TIME modulation-based cancer immunotherapeutic strategies, medicative principles and accessory biotechniques for further clinical transformation. Remarkably, the recent advances of cancer immunotherapeutic drug delivery systems and future prospects of TIME modulation-based drug delivery systems for much more controlled and precise cancer immunotherapy will be emphatically discussed.
Collapse
Affiliation(s)
- Shuyan Han
- Key Laboratory of Sensing Technology and Biomedical Instrument of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Guangzhou 510006, PR China.
| | | | | | | |
Collapse
|
117
|
Chao Y, Chen Q, Liu Z. Smart Injectable Hydrogels for Cancer Immunotherapy. ADVANCED FUNCTIONAL MATERIALS 2020; 30. [DOI: 10.1002/adfm.201902785] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Indexed: 01/06/2025]
Abstract
AbstractHydrogels, a class of materials with a 3D network structure, are widely used in various fields especially in biomedicine. Injectable hydrogels could facilitate the encapsulation and controlled release of small molecular drugs, macromolecular therapeutics, and even cells. With the rapid development of cancer immunotherapy, such injectable hydrogels have attracted wide attention for local immunomodulation to boost systemic anticancer immune responses, realizing more effective immunotherapy at lower doses. The latest progresses in the development of various smart injectable hydrogels for cancer immunotherapy are summarized here. Although applied locally, such injectable hydrogels can activate systemic antitumor immune responses, safely and effectively inhibiting the tumor metastasis and recurrence. Moreover, it is discussed how injectable hydrogel‐based cancer immunotherapy would contribute to the development of next generation of cancer treatment together with their potential for clinical translation.
Collapse
Affiliation(s)
- Yu Chao
- Institute of Functional Nano and Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon‐Based Functional Materials and Devices Soochow University Suzhou Jiangsu 215123 China
| | - Qian Chen
- Institute of Functional Nano and Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon‐Based Functional Materials and Devices Soochow University Suzhou Jiangsu 215123 China
| | - Zhuang Liu
- Institute of Functional Nano and Soft Materials (FUNSOM) Jiangsu Key Laboratory for Carbon‐Based Functional Materials and Devices Soochow University Suzhou Jiangsu 215123 China
| |
Collapse
|
118
|
Colao A, de Nigris F, Modica R, Napoli C. Clinical Epigenetics of Neuroendocrine Tumors: The Road Ahead. Front Endocrinol (Lausanne) 2020; 11:604341. [PMID: 33384663 PMCID: PMC7770585 DOI: 10.3389/fendo.2020.604341] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/12/2020] [Indexed: 12/16/2022] Open
Abstract
Neuroendocrine tumors, or NETs, are cancer originating in neuroendocrine cells. They are mostly found in the gastrointestinal tract or lungs. Functional NETs are characterized by signs and symptoms caused by the oversecretion of hormones and other substances, but most NETs are non-functioning and diagnosis in advanced stages is common. Thus, novel diagnostic and therapeutic strategies are warranted. Epigenetics may contribute to refining the diagnosis, as well as to identify targeted therapy interfering with epigenetic-sensitive pathways. The goal of this review was to discuss the recent advancement in the epigenetic characterization of NETs highlighting their role in clinical findings.
Collapse
Affiliation(s)
- Annamaria Colao
- Department of Clinical Medicine and Surgery, Unesco Chair Health Education and Sustainable Development, Federico II University of Naples, Naples, Italy
| | - Filomena de Nigris
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy
| | - Roberta Modica
- Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples, Italy
- *Correspondence: Roberta Modica,
| | - Claudio Napoli
- Department of Advanced Medical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, Naples, Italy
| |
Collapse
|
119
|
Gu M, Toh TB, Hooi L, Lim JJ, Zhang X, Chow EKH. Nanodiamond-Mediated Delivery of a G9a Inhibitor for Hepatocellular Carcinoma Therapy. ACS APPLIED MATERIALS & INTERFACES 2019; 11:45427-45441. [PMID: 31718136 DOI: 10.1021/acsami.9b16323] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver cancer with high mortality but limited therapeutic options. Epigenetic regulations including DNA methylation and histone modification control gene expressions and play a crucial role during tumorigenesis. G9a, also known as EHMT2 (euchromatic histone-lysine N-methyltransferase 2), is a histone methyltransferase predominantly responsible for dimethylation of histone H3 lysine 9 (H3K9). G9a has been shown to play a key role in promoting tumor progression. Recent studies have identified that G9a is a critical mediator of HCC pathogenesis. UNC0646 is a G9a inhibitor that has shown potent in vitro efficacy. However, due to its water insolubility, the in vivo efficacy of UNC0646 is not satisfactory. In this study, nanodiamonds (NDs) were utilized as a drug delivery platform to improve in vivo delivery of this small-molecule inhibitor. Our results showed that ND-UNC0646 complexes could be rapidly synthesized by physical adsorption, meanwhile possessing favorable drug delivery properties and was able to improve the dispersibility of UNC0646 in water, therefore making it amenable for intravenous administration. The release profile of UNC0646 from ND-UNC0646 was demonstrated to be pH-responsive. Moreover, ND-UNC0646 maintained the biological functionality of UNC0646, with higher efficacy in reducing H3K9 methylation as well as enhanced invasion suppressive effects. Most importantly, increased in vivo efficacy was demonstrated using an orthotopic HCC mouse model, which paves the way of translating this small-molecule inhibitor toward HCC treatment. Our work demonstrates the potential of NDs in the clinical application for HCC treatment.
Collapse
Affiliation(s)
- Mengjie Gu
- Department of Pharmacology, Yong Loo Lin School of Medicine , National University of Singapore , 117600 , Singapore
- Cancer Science Institute of Singapore , National University of Singapore , 117599 , Singapore
| | - Tan Boon Toh
- The N.1 Institute for Health , National University of Singapore , 117456 , Singapore
| | - Lissa Hooi
- Cancer Science Institute of Singapore , National University of Singapore , 117599 , Singapore
| | - Jhin Jieh Lim
- Cancer Science Institute of Singapore , National University of Singapore , 117599 , Singapore
| | - Xiyun Zhang
- Cancer Science Institute of Singapore , National University of Singapore , 117599 , Singapore
- Department of Medicine, Yong Loo Lin School of Medicine , National University of Singapore , 119228 , Singapore
| | - Edward Kai-Hua Chow
- Department of Pharmacology, Yong Loo Lin School of Medicine , National University of Singapore , 117600 , Singapore
- Cancer Science Institute of Singapore , National University of Singapore , 117599 , Singapore
- The N.1 Institute for Health , National University of Singapore , 117456 , Singapore
| |
Collapse
|
120
|
Ni K, Lan G, Chan C, Duan X, Guo N, Veroneau SS, Weichselbaum RR, Lin W. Ultrathin metal-organic layer-mediated radiotherapy-radiodynamic therapy enhances immunotherapy of metastatic cancers. MATTER 2019; 1:1331-1353. [PMID: 32832885 DOI: 10.1016/j.matt.2019.06.007] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Checkpoint blockade immunotherapy (CBI) is effective in promoting a systemic immune response against some metastatic tumors. The reliance on the pre-existing immune environment of the tumor, however, limits the efficacy of CBI on a broad spectrum of cancers. Herein, we report the design of a novel nanoscale metal-organic layer (nMOL), Hf-MOL, for effective treatment of local tumors by enabling radiotherapy-radiodynamic therapy (RT-RDT) with low-dose X-rays and, when in combination with an immune checkpoint inhibitor, regression of metastatic tumors by re-activating anti-tumor immunity and inhibiting myeloid-derived suppressor cells. Owing to the reduced dimensionality, nMOLs allow facile diffusion of reactive oxygen species and exhibit superior RT-RDT effects. The synergy of Hf-MOL-enabled RT-RDT immune activation and anti-programmed death ligand 1 (anti-PD-L1) CBI led to robust abscopal effects on a series of bilateral models of colon, head and neck, and breast cancers and significant anti-metastatic effects on an orthotopic model of breast cancer.
Collapse
Affiliation(s)
- Kaiyuan Ni
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- These authors contributed equally to this work
| | - Guangxu Lan
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- These authors contributed equally to this work
| | - Christina Chan
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
| | - Xiaopin Duan
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
| | - Nining Guo
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Department of Radiation and Cellular Oncology and The Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL 60637, USA
| | - Samuel S Veroneau
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology and The Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL 60637, USA
| | - Wenbin Lin
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
- Department of Radiation and Cellular Oncology and The Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
121
|
Zhang M, Liu K, Wang M. Development of cancer immunotherapy based on PD-1/PD-L1 pathway blockade. RSC Adv 2019; 9:33903-33911. [PMID: 35528929 PMCID: PMC9073714 DOI: 10.1039/c9ra04590b] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 10/16/2019] [Indexed: 12/29/2022] Open
Abstract
Programmed death receptor 1 (PD-1)/programmed death ligand 1 (PD-L1) blockade therapy has achieved considerable success in various tumours. However, only a fraction of patients benefit from its clinical application, and some patients might be suffer from tumour resistance against PD-1/PD-L1 blockade therapy after the original response. In this review, we summarized the main reasons that caused the low response rate of PD-/PD-L1 blockade therapy: firstly, the off-target of PD-1/PD-L1 blocking agents, which is also the main factor of the side effect of autoimmune disorders; secondly, the insufficient infiltration of T cells in a tumour microenvironment; thirdly, the low immunogenicity of tumor cells; fourth, other immunosuppressive components impairing the therapeutic efficacy of the immunotherapy based on the PD-/PD-L1 blockade, and introducing some updated the delivery system of PD-1/PD-L1 blocking agents and the combination therapy based on PD-1/PD-L1 inhibitors and other therapeutics that can complement and promote each other to achieve improved immune response.
Collapse
Affiliation(s)
- Min Zhang
- College of Food Science and Technology, Shanghai Ocean University 999 Hucheng Ring Road Shanghai 201306 China
| | - Kehai Liu
- College of Food Science and Technology, Shanghai Ocean University 999 Hucheng Ring Road Shanghai 201306 China
| | - Mingfu Wang
- College of Food Science and Technology, Shanghai Ocean University 999 Hucheng Ring Road Shanghai 201306 China
- University Hong Kong, School of Biological Sciences Pokfulam Road Hong Kong 999077 China
| |
Collapse
|
122
|
Saeed M, Gao J, Shi Y, Lammers T, Yu H. Engineering Nanoparticles to Reprogram the Tumor Immune Microenvironment for Improved Cancer Immunotherapy. Am J Cancer Res 2019; 9:7981-8000. [PMID: 31754376 PMCID: PMC6857062 DOI: 10.7150/thno.37568] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 08/12/2019] [Indexed: 02/06/2023] Open
Abstract
Immunotherapy is rapidly maturing towards extensive clinical use. However, it does not work well in large patient populations because of an immunosuppressed microenvironment and limited reinvigoration of antitumor immunity. The tumor microenvironment is a complex milieu in which the principles of physiology and anatomy are defied and which is considered an immune-privileged site promoting T cell exhaustion. Tremendous research interest exists in developing nanoparticle-based approaches to modulate antitumor immune responses. The increasing use of immunotherapies in the clinic requires robust programming of immune cells to boost antitumor immunity. This review summarizes recent advances in the engineering of nanoparticles for improved anticancer immunotherapy. It discusses emerging nanoparticle-based approaches for the modulation of tumor cells and immune cells, such as dendritic cells, T cells and tumor-associated macrophages, with the intention to overcome challenges currently faced in the clinic. Furthermore, this review describes potentially curative combination therapeutic approaches to provoke effective tumor antigen-specific immune responses. We foresee a future in which improvement in patient's surveillance will become a mainstream practice.
Collapse
|
123
|
Wang M, Yu Z, Feng H, Wang J, Wang L, Zhang Y, Yin L, Du Y, Jiang H, Wang X, Zhou J. In situ biosynthesized gold nanoclusters inhibiting cancer development via the PI3K-AKT signaling pathway. J Mater Chem B 2019; 7:5336-5344. [PMID: 31393501 DOI: 10.1039/c9tb01070j] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Nanomaterials have made great breakthroughs in drug delivery. However, in previous studies, nanomaterials have been mostly used as vehicles to transport drugs into tumors. Herein, we first found that the in situ biosynthesized gold nanoparticles (Au NCs) can inhibit cancer development via the inhibition of some signaling pathways. Classical cell phenotypic assay tests and orthotropic liver tumor model both showed that the in situ biosynthesized Au NCs could inhibit tumor development. With the help of the RNA-seq analysis, we found that the in situ biosynthesized Au NCs could significantly inhibit the PI3K-AKT signaling pathway, thus effectively impeding tumor development. This facile and effective tumor targeting theranostics in vivo can effectively cure cancers in future clinical applications.
Collapse
Affiliation(s)
- Maonan Wang
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab), School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Zeqian Yu
- Department of Hepatic-Biliary-Pancreatic Center, Zhongda Hospital, Southeast University, Nanjing, China. and Department of Hepatobiliary Surgery Research Institute, Southeast University, Nanjing, China
| | - Huan Feng
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab), School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Jianling Wang
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab), School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Lishan Wang
- Department of Hepatic-Biliary-Pancreatic Center, Zhongda Hospital, Southeast University, Nanjing, China. and Department of Hepatobiliary Surgery Research Institute, Southeast University, Nanjing, China
| | - Yu Zhang
- School of Medicine, Southeast University, Nanjing, China
| | - Lihong Yin
- School of Public Health, Southeast University, Nanjing, China
| | - Ying Du
- School of Public Health, Southeast University, Nanjing, China
| | - Hui Jiang
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab), School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Xuemei Wang
- State Key Laboratory of Bioelectronics (Chien-Shiung Wu Lab), School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Jiahua Zhou
- Department of Hepatic-Biliary-Pancreatic Center, Zhongda Hospital, Southeast University, Nanjing, China. and Department of Hepatobiliary Surgery Research Institute, Southeast University, Nanjing, China
| |
Collapse
|
124
|
Li H, Zhang P, Luo J, Hu D, Huang Y, Zhang ZR, Fu Y, Gong T. Chondroitin Sulfate-Linked Prodrug Nanoparticles Target the Golgi Apparatus for Cancer Metastasis Treatment. ACS NANO 2019; 13:9386-9396. [PMID: 31375027 DOI: 10.1021/acsnano.9b04166] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Metastasis is a multistep biological process regulated by multiple signaling pathways. The integrity of the Golgi apparatus plays an important role in these signaling pathways. Inspired by the mechanism and our previous finding about accumulation of chondroitin sulfate in Golgi apparatus in hepatic stellate cells, we developed a Golgi apparatus-targeting prodrug nanoparticle system by synthesizing retinoic acid (RA)-conjugated chondroitin sulfate (CS) (CS-RA). The prodrug nanoparticles appeared to accumulate in the Golgi apparatus in cancer cells and realized RA release under an acidic environment. We confirmed that CS-RA exhibited successful inhibition of multiple metastasis-associated proteins expression in vitro and in vivo by disruption of the Golgi apparatus structure. Following loading with paclitaxel (PTX), the CS-RA based nanoformulation (PTX-CS-RA) inhibited migration, invasion, and angiogenesis in vitro and suppressed tumor growth and metastasis in 4T1-Luc bearing mice. This multistep targeted nanoparticle system potentially enhanced the effect of antimetastasis combined with chemotherapy.
Collapse
Affiliation(s)
- Haohuan Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , Chengdu 610064 , China
| | - Pei Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , Chengdu 610064 , China
| | - Jingwen Luo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , Chengdu 610064 , China
| | - Danrong Hu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , Chengdu 610064 , China
| | - Yuan Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , Chengdu 610064 , China
| | - Zhi-Rong Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , Chengdu 610064 , China
| | - Yao Fu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , Chengdu 610064 , China
| | - Tao Gong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy , Sichuan University , Chengdu 610064 , China
| |
Collapse
|
125
|
Gang F, Yan H, Ma C, Jiang L, Gu Y, Liu Z, Zhao L, Wang X, Zhang J, Sun X. Robust magnetic double-network hydrogels with self-healing, MR imaging, cytocompatibility and 3D printability. Chem Commun (Camb) 2019; 55:9801-9804. [PMID: 31360942 DOI: 10.1039/c9cc04241e] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Herein, we have fabricated a novel robust self-healing magnetic double-network hydrogel by multiple interactions between bondable magnetic Fe3O4 and chitosan-polyolefin matrix, and the hydrogel also exhibits an excellent magnetogenic effect and MR imageability. The practical potential of the magnetic double-network hydrogel is further revealed by its 3D-printing performance.
Collapse
Affiliation(s)
- Fangli Gang
- State Key Laboratory of Crop Stress Biology for Arid Areas and College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|