51
|
Haegebaert RM, Kempers M, Ceelen W, Lentacker I, Remaut K. Nanoparticle mediated targeting of toll-like receptors to treat colorectal cancer. Eur J Pharm Biopharm 2022; 172:16-30. [PMID: 35074555 DOI: 10.1016/j.ejpb.2022.01.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 12/16/2021] [Accepted: 01/17/2022] [Indexed: 02/07/2023]
|
52
|
Cheng X, Yu P, Zhou X, Zhu J, Han Y, Zhang C, Kong L. Enhanced tumor homing of pathogen-mimicking liposomes driven by R848 stimulation: A new platform for synergistic oncology therapy. Acta Pharm Sin B 2022; 12:924-938. [PMID: 35256955 PMCID: PMC8897206 DOI: 10.1016/j.apsb.2021.08.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/20/2021] [Accepted: 07/02/2021] [Indexed: 11/01/2022] Open
Abstract
Although multifarious tumor-targeting modifications of nanoparticulate systems have been attempted in joint efforts by our predecessors, it remains challenging for nanomedicine to traverse physiological barriers involving blood vessels, tissues, and cell barriers to thereafter demonstrate excellent antitumor effects. To further overcome these inherent obstacles, we designed and prepared mycoplasma membrane (MM)-fused liposomes (LPs) with the goal of employing circulating neutrophils with the advantage of inflammatory cytokine-guided autonomous tumor localization to transport nanoparticles. We also utilized in vivo neutrophil activation induced by the liposomal form of the immune activator resiquimod (LPs-R848). Fused LPs preparations retained mycoplasma pathogen characteristics and achieved rapid recognition and endocytosis by activated neutrophils stimulated by LPs-R848. The enhanced neutrophil infiltration in homing of the inflammatory tumor microenvironment allowed more nanoparticles to be delivered into solid tumors. Facilitated by the formation of neutrophil extracellular traps (NETs), podophyllotoxin (POD)-loaded MM-fused LPs (MM-LPs-POD) were concomitantly released from neutrophils and subsequently engulfed by tumor cells during inflammation. MM-LPs-POD displayed superior suppression efficacy of tumor growth and lung metastasis in a 4T1 breast tumor model. Overall, such a strategy of pathogen-mimicking nanoparticles hijacking neutrophils in situ combined with enhanced neutrophil infiltration indeed elevates the potential of chemotherapeutics for tumor targeting therapy.
Collapse
|
53
|
Luo L, Qi Y, Zhong H, Jiang S, Zhang H, Cai H, Wu Y, Gu Z, Gong Q, Luo K. GSH-sensitive polymeric prodrug: Synthesis and loading with photosensitizers as nanoscale chemo-photodynamic anti-cancer nanomedicine. Acta Pharm Sin B 2022; 12:424-436. [PMID: 35127396 PMCID: PMC8799999 DOI: 10.1016/j.apsb.2021.05.003] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/23/2021] [Accepted: 03/30/2021] [Indexed: 12/17/2022] Open
Abstract
Precisely delivering combinational therapeutic agents has become a crucial challenge for anti-tumor treatment. In this study, a novel redox-responsive polymeric prodrug (molecular weight, MW: 93.5 kDa) was produced by reversible addition-fragmentation chain transfer (RAFT) polymerization. The amphiphilic block polymer-doxorubicin (DOX) prodrug was employed to deliver a hydrophobic photosensitizer (PS), chlorin e6 (Ce6), and the as-prepared nanoscale system [NPs(Ce6)] was investigated as a chemo-photodynamic anti-cancer agent. The glutathione (GSH)-cleavable disulfide bond was inserted into the backbone of the polymer for biodegradation inside tumor cells, and DOX conjugated onto the polymer with a disulfide bond was successfully released intracellularly. NPs(Ce6) released DOX and Ce6 with their original molecular structures and degraded into segments with low MWs of 41.2 kDa in the presence of GSH. NPs(Ce6) showed a chemo-photodynamic therapeutic effect to kill 4T1 murine breast cancer cells, which was confirmed from a collapsed cell morphology, a lifted level in the intracellular reactive oxygen species, a reduced viability and induced apoptosis. Moreover, ex vivo fluorescence images indicated that NPs(Ce6) retained in the tumor, and exhibited a remarkable in vivo anticancer efficacy. The combinational therapy showed a significantly increased tumor growth inhibition (TGI, 58.53%). Therefore, the redox-responsive, amphiphilic block polymeric prodrug could have a great potential as a chemo-photodynamic anti-cancer agent.
Collapse
Affiliation(s)
- Lei Luo
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Yiming Qi
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Hong Zhong
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Shinan Jiang
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400716, China
| | - Hu Zhang
- Amgen Bioprocessing Centre, Keck Graduate Institute, Claremont, CA 91711, USA
| | - Hao Cai
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yahui Wu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhongwei Gu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, China
- Corresponding author. Tel./fax: +86 28 85422538.
| |
Collapse
|
54
|
Hua J, Wu P, Gan L, Zhang Z, He J, Zhong L, Zhao Y, Huang Y. Current Strategies for Tumor Photodynamic Therapy Combined With Immunotherapy. Front Oncol 2021; 11:738323. [PMID: 34868932 PMCID: PMC8635494 DOI: 10.3389/fonc.2021.738323] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 10/27/2021] [Indexed: 12/30/2022] Open
Abstract
Photodynamic therapy (PDT) is a low invasive antitumor therapy with fewer side effects. On the other hand, immunotherapy also has significant clinical applications in the treatment of cancer. Both therapies, on their own, have some limitations and are incapable of meeting the demands of the current cancer treatment. The efficacy of PDT and immunotherapy against tumor metastasis and tumor recurrence may be improved by combination strategies. In this review, we discussed the possibility that PDT could be used to activate immune responses by inducing immunogenic cell death or generating cancer vaccines. Furthermore, we explored the latest advances in PDT antitumor therapy in combination with some immunotherapy such as immune adjuvants, inhibitors of immune suppression, and immune checkpoint blockade.
Collapse
Affiliation(s)
- Jianfeng Hua
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, China
| | - Pan Wu
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, China
| | - Lu Gan
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, China
| | - Zhikun Zhang
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, China
| | - Jian He
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, China
| | - Liping Zhong
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, China
| | - Yongxiang Zhao
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, China
| | - Yong Huang
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, China
- The First People’s Hospital of Changde City, Changde, China
| |
Collapse
|
55
|
Jiang Z, Li T, Cheng H, Zhang F, Yang X, Wang S, Zhou J, Ding Y. Nanomedicine potentiates mild photothermal therapy for tumor ablation. Asian J Pharm Sci 2021; 16:738-761. [PMID: 35027951 PMCID: PMC8739255 DOI: 10.1016/j.ajps.2021.10.001] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 10/03/2021] [Accepted: 10/05/2021] [Indexed: 12/17/2022] Open
Abstract
The booming photothermal therapy (PTT) has achieved great progress in non-invasive oncotherapy, and paves a novel way for clinical oncotherapy. Of note, mild temperature PTT (mPTT) of 42–45 °C could avoid treatment bottleneck of the traditional PTT, including nonspecific injury to normal tissues, vasculature and host antitumor immunity. However, cancer cells can resist mPTT via heat shock response and autophagy, thus leading to insufficient mPTT monotherapy to ablate tumor. To overcome the deficient antitumor efficacy caused by thermo-resistance of cancer cells and mono mPTT, synergistic therapies towards cancer cells have been conducted with mPTT. This review summarizes the recent advances in nanomedicine-potentiated mPTT for cancer treatment, including strategies for enhanced single-mode mPTT and mPTT plus synergistic therapies. Moreover, challenges and prospects for clinical translation of nanomedicine-potentiated mPTT are discussed.
Collapse
|
56
|
Yang Y, Guo T, Xu J, Xiong Y, Cui X, Ke Y, Wang C. Micelle nanovehicles for co-delivery of Lepidium meyenii Walp. (maca) polysaccharide and chloroquine to tumor-associated macrophages for synergistic cancer immunotherapy. Int J Biol Macromol 2021; 189:577-589. [PMID: 34450149 DOI: 10.1016/j.ijbiomac.2021.08.155] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 01/18/2023]
Abstract
Here, we fabricated amphiphilic polysaccharide micelles for synergistic cancer immunotherapy targeting tumor-associated macrophages (TAMs). Lepidium meyenii Walp. (maca) polysaccharide (MP), a naturally derived macromolecule with a strong TAM-remodeling effect, was grafted on a hydrophobic poly(lactic-co-glycolic acid) (PLGA) segment, with a disulfide bond for redox-sensitive linkage. The amphiphilic polysaccharide derivatives could self-assemble into core (PLGA)-shell (MP)-structured micelles and encapsulate chloroquine (CQ) into the hydrophobic core. By using a 4T1-M2 macrophage co-culture model and a 4T1 tumor xenograft mouse model, we showed that the prepared micelles could co-deliver MP and CQ to the tumor sites and selectively accumulate at TAMs because of the specific properties of MP. Furthermore, the nanoparticles exerted synergistic tumor immunotherapeutic and antimetastatic effects, which might be attributable to the enhanced cell internalization of the micelles and the multiple regulatory mechanisms of MP and CQ. Thus, immunomodulatory MP may be a promising biomaterial for cancer immunotherapy.
Collapse
Affiliation(s)
- Ye Yang
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; Key Laboratory of Sustainable Utilization of Panax Notoginseng Resources of Yunnan, Province, Kunming 650500, China
| | - Tingting Guo
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; Key Laboratory of Sustainable Utilization of Panax Notoginseng Resources of Yunnan, Province, Kunming 650500, China
| | - Junwei Xu
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China
| | - Yin Xiong
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; Key Laboratory of Sustainable Utilization of Panax Notoginseng Resources of Yunnan, Province, Kunming 650500, China
| | - Xiuming Cui
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; Key Laboratory of Sustainable Utilization of Panax Notoginseng Resources of Yunnan, Province, Kunming 650500, China
| | - Yang Ke
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Kunming Medical University, Kunming 650500, China.
| | - Chengxiao Wang
- School of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China; Key Laboratory of Sustainable Utilization of Panax Notoginseng Resources of Yunnan, Province, Kunming 650500, China.
| |
Collapse
|
57
|
Pauli G, Chao PH, Qin Z, Böttger R, Lee SE, Li SD. Liposomal Resiquimod for Enhanced Immunotherapy of Peritoneal Metastases of Colorectal Cancer. Pharmaceutics 2021; 13:1696. [PMID: 34683992 PMCID: PMC8540734 DOI: 10.3390/pharmaceutics13101696] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/25/2021] [Accepted: 10/12/2021] [Indexed: 12/22/2022] Open
Abstract
Colorectal cancer with peritoneal metastases is currently treated by cytoreductive surgery and locoregional chemotherapeutics. This standard treatment is associated with high morbidity, mortality, and recurrence rate. To augment the existing therapy, we developed a liposome-based delivery system containing 1,2-stearoyl-3-trimethylammonium-propane chloride (DSTAP), a cationic lipid, to localize a toll-like receptor agonist, resiquimod (R848), in the peritoneal cavity (PerC) for enhancing the immune response against cancer that had spread to the PerC. The liposomes delivered by intraperitoneal injection increased peritoneal retention of R848 by 14-fold while retarding its systemic absorption, leading to a 5-fold decreased peak plasma concentration compared to free R848 in mice. Within the PerC, the DSTAP-liposomes were found in ~40% of the dendritic cells by flow cytometry. DSTAP-R848 significantly upregulated interferon α (IFN-α) in the peritoneal fluid by 2-fold compared to free R848, without increasing the systemic level. Combined with oxaliplatin, a cytotoxic agent inducing immunogenic cell death, DSTAP-R848 effectively inhibited the progression of CT26 murine colorectal tumor in the PerC, while the combination with free R848 only showed a mild effect. Moreover, the combination of oxaliplatin and DSTAP-R848 significantly increased infiltration of CD8+ T cells in the PerC compared to oxaliplatin combined with free R848, indicating enhanced immune response against the tumor. The results suggest that DSTAP-R848 exhibits potential in augmenting existing therapies for treating colorectal cancer with peritoneal metastases via immune activation.
Collapse
Affiliation(s)
- Griffin Pauli
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; (G.P.); (P.-H.C.); (Z.Q.); (R.B.); (S.E.L.)
| | - Po-Han Chao
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; (G.P.); (P.-H.C.); (Z.Q.); (R.B.); (S.E.L.)
| | - Zhu Qin
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; (G.P.); (P.-H.C.); (Z.Q.); (R.B.); (S.E.L.)
- Institute of Veterinary Immunology & Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
| | - Roland Böttger
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; (G.P.); (P.-H.C.); (Z.Q.); (R.B.); (S.E.L.)
| | - Suen Ern Lee
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; (G.P.); (P.-H.C.); (Z.Q.); (R.B.); (S.E.L.)
| | - Shyh-Dar Li
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; (G.P.); (P.-H.C.); (Z.Q.); (R.B.); (S.E.L.)
| |
Collapse
|
58
|
Zeng Y, Xiang Y, Sheng R, Tomás H, Rodrigues J, Gu Z, Zhang H, Gong Q, Luo K. Polysaccharide-based nanomedicines for cancer immunotherapy: A review. Bioact Mater 2021; 6:3358-3382. [PMID: 33817416 PMCID: PMC8005658 DOI: 10.1016/j.bioactmat.2021.03.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/19/2021] [Accepted: 03/02/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer immunotherapy is an effective antitumor approach through activating immune systems to eradicate tumors by immunotherapeutics. However, direct administration of "naked" immunotherapeutic agents (such as nucleic acids, cytokines, adjuvants or antigens without delivery vehicles) often results in: (1) an unsatisfactory efficacy due to suboptimal pharmacokinetics; (2) strong toxic and side effects due to low targeting (or off-target) efficiency. To overcome these shortcomings, a series of polysaccharide-based nanoparticles have been developed to carry immunotherapeutics to enhance antitumor immune responses with reduced toxicity and side effects. Polysaccharides are a family of natural polymers that hold unique physicochemical and biological properties, as they could interact with immune system to stimulate an enhanced immune response. Their structures offer versatility in synthesizing multifunctional nanocomposites, which could be chemically modified to achieve high stability and bioavailability for delivering therapeutics into tumor tissues. This review aims to highlight recent advances in polysaccharide-based nanomedicines for cancer immunotherapy and propose new perspectives on the use of polysaccharide-based immunotherapeutics.
Collapse
Affiliation(s)
- Yujun Zeng
- Huaxi MR Research Center (HMRRC), Department of Radiology, Department of Neurosurgery, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yufan Xiang
- Huaxi MR Research Center (HMRRC), Department of Radiology, Department of Neurosurgery, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ruilong Sheng
- CQM-Centro de Quimica da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9000-390, Funchal, Madeira, Portugal
| | - Helena Tomás
- CQM-Centro de Quimica da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9000-390, Funchal, Madeira, Portugal
| | - João Rodrigues
- CQM-Centro de Quimica da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9000-390, Funchal, Madeira, Portugal
| | - Zhongwei Gu
- Huaxi MR Research Center (HMRRC), Department of Radiology, Department of Neurosurgery, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Hu Zhang
- Amgen Bioprocessing Centre, Keck Graduate Institute, Claremont, CA, 91711, USA
| | - Qiyong Gong
- Huaxi MR Research Center (HMRRC), Department of Radiology, Department of Neurosurgery, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Kui Luo
- Huaxi MR Research Center (HMRRC), Department of Radiology, Department of Neurosurgery, Functional and Molecular Imaging Key Laboratory of Sichuan Province, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| |
Collapse
|
59
|
Li M, Zhao Y, Zhang W, Zhang S, Zhang S. Multiple-therapy strategies via polysaccharides-based nano-systems in fighting cancer. Carbohydr Polym 2021; 269:118323. [PMID: 34294335 DOI: 10.1016/j.carbpol.2021.118323] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/31/2021] [Accepted: 06/07/2021] [Indexed: 12/30/2022]
Abstract
Polysaccharide-based biomaterials (e.g., chitosan, dextran, hyaluronic acid, chondroitin sulfate and heparin) have received great attention in healthcare, particularly in drug delivery for tumor therapy. They are naturally abundant and available, outstandingly biodegradable and biocompatible, and they generally have negligible toxicity and low immunogenicity. In addition, they are easily chemically or physically modified. Therefore, PSs-based nanoparticles (NPs) have been extensively investigated for the enhancement of tumor treatment. In this review, we introduce the synthetic pathways of amphiphilic PS derivatives, which allow the constructs to self-assemble into NPs with various structures. We especially offer an overview of the emerging applications of self-assembled PSs-based NPs in tumor chemotherapy, photothermal therapy (PTT), photodynamic therapy (PDT), gene therapy and immunotherapy. We believe that this review can provide criteria for a rational and molecular level-based design of PS-based NPs, and comprehensive insight into the potential of PS-based NPs used in multiple cancer therapies.
Collapse
Affiliation(s)
- Min Li
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, PR China; State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116024, PR China
| | - Yinan Zhao
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, PR China
| | - Wenjun Zhang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116024, PR China
| | - Shufen Zhang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian 116024, PR China.
| | - Shubiao Zhang
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, PR China.
| |
Collapse
|
60
|
Jeong S, Choi Y, Kim K. Engineering Therapeutic Strategies in Cancer Immunotherapy via Exogenous Delivery of Toll-like Receptor Agonists. Pharmaceutics 2021; 13:1374. [PMID: 34575449 PMCID: PMC8466827 DOI: 10.3390/pharmaceutics13091374] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/15/2021] [Accepted: 08/17/2021] [Indexed: 12/11/2022] Open
Abstract
As a currently spotlighted method for cancer treatment, cancer immunotherapy has made a lot of progress in recent years. Among tremendous cancer immunotherapy boosters available nowadays, Toll-like receptor (TLR) agonists were specifically selected, because of their effective activation of innate and adaptive immune cells, such as dendritic cells (DCs), T cells, and macrophages. TLR agonists can activate signaling pathways of DCs to express CD80 and CD86 molecules, and secrete various cytokines and chemokines. The maturation of DCs stimulates naïve T cells to differentiate into functional cells, and induces B cell activation. Although TLR agonists have anti-tumor ability by activating the immune system of the host, their drawbacks, which include poor efficiency and remarkably short retention time in the body, must be overcome. In this review, we classify and summarize the recently reported delivery strategies using (1) exogenous TLR agonists to maintain the biological and physiological signaling activities of cargo agonists, (2) usage of multiple TLR agonists for synergistic immune responses, and (3) co-delivery using the combination with other immunomodulators or stimulants. In contrast to naked TLR agonists, these exogenous TLR delivery strategies successfully facilitated immune responses and subsequently mediated anti-tumor efficacy.
Collapse
Affiliation(s)
| | | | - Kyobum Kim
- Department of Chemical & Biochemical Engineering, Dongguk University, 30, Pildong-ro 1-gil, Jung-gu, Seoul 22012, Korea; (S.J.); (Y.C.)
| |
Collapse
|
61
|
Song Q, Zhang G, Wang B, Cao G, Li D, Wang Y, Zhang Y, Geng J, Li H, Li Y. Reinforcing the Combinational Immuno-Oncotherapy of Switching "Cold" Tumor to "Hot" by Responsive Penetrating Nanogels. ACS APPLIED MATERIALS & INTERFACES 2021; 13:36824-36838. [PMID: 34314148 DOI: 10.1021/acsami.1c08201] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Although immuno-oncotherapy in clinic has gained great success, the immunosuppressive tumor microenvironment (TME) existing in the "cold" tumor with insufficient and exhausted lymphocytes may result in a lower-than-expected therapeutic efficiency. Therefore, a properly designed synergistic strategy that can effectively turn the "cold" tumor to "hot" should be considered to improve the therapeutic effects of immuno-oncotherapy. Herein, TME-responsive penetrating nanogels (NGs) were developed, which can improve the delivery and penetration of the co-loaded resiquimod (R848) and green tea catechin (EGCG) in tumors by a nano-sized controlled releasing system of the soluble cyclodextrin-drug inclusion complex. Consequently, the NGs effectively promoted the maturation of dendritic cells, stimulated the cytotoxic T lymphocytes (CTLs), and decreased the PD-L1 expression in tumors. The combination of NGs with the OX40 agonist (αOX40) further synergistically enhanced the activation and infiltration of CTLs into the deep tumor and inhibited the suppression effects from the regulatory T cells (Tregs). As a result, an increased ratio of active CTLs to Tregs in tumors (20.66-fold) was achieved with a 91.56% tumor suppression effect, indicating a successful switch of "cold" tumors to "hot" for an immunologically beneficial TME with significantly improved anti-tumor immune therapeutics. This strategy could be tailored to other immuno-oncotherapeutic approaches to solve the urgent efficiency concerns of the checkpoint-based treatment in clinic.
Collapse
Affiliation(s)
- Qingle Song
- Laboratory of Immunology and Nanomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Guofang Zhang
- Laboratory of Immunology and Nanomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Bo Wang
- Laboratory of Immunology and Nanomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Guoli Cao
- Laboratory of Immunology and Nanomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Dongjie Li
- Laboratory of Immunology and Nanomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- College of Life Science, Hebei Normal University, Shijiazhuang 050016, China
| | - Yu Wang
- Laboratory of Immunology and Nanomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yuqian Zhang
- Laboratory of Immunology and Nanomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jin Geng
- Center for Polymers in Medicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Hongchang Li
- Guangdong Key Laboratory of Nanomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Yang Li
- Laboratory of Immunology and Nanomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
62
|
Ding M, Shao K, Wu L, Jiang Y, Cheng B, Wang L, Shi J, Kong X. A NO/ROS/RNS cascaded-releasing nano-platform for gas/PDT/PTT/immunotherapy of tumors. Biomater Sci 2021; 9:5824-5840. [PMID: 34269777 DOI: 10.1039/d1bm00726b] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Nitric oxide (NO) gas treatment offers a promising strategy for tumor therapy; however, its practical application is still limited due to its poor efficacy and biotoxicity which were caused by gas leakage during blood delivery. Herein, a nano-platform (CMH-OBN) composed of chlorin e6-melanin-hyaluronic acid nanoparticles (Ce6-MNP-HA, CMH) and oxidized bletilla striata polysaccharide microcapsules (Oxi-BSP) carrying NO donors was prepared for responsive and cascaded release of NO, reactive oxygen species (ROS) and its secondary metabolite reactive nitrogen species (RNS) in tumor sites. Melanin not only endowed CMH with good photothermal properties, but also helped Ce6 to produce a large number of ROS under near-infrared (NIR) irradiation. OBN microcapsules, which were sensitive to ROS, can release NO donors under the stimulation of ROS released by CMH nanoparticles under NIR irradiation and can further release NO in the tumor microenvironment (TME) with high expression of glutathione (GSH). NO could further up-regulate soluble guanylate cyclase-cyclic guanosine monophosphate (sGC-cGMP) signal pathways to relieve hypoxia, thus further enhancing the photodynamic therapy (PDT). Moreover, the cascaded release of ROS and NO could produce RNS with higher lethality, which could sequentially initiate the cellular apoptotic procedure and promote immunotherapy by activating T cells at the tumor sites. More interestingly, the CMH-OBN nano-platform could supply magnetic resonance imaging (MRI) and infrared photothermal imaging guidance for tumor therapy. In conclusion, the development of a CMH-OBN nano-platform provides a satisfactory demonstration by combining NO therapy with photothermal therapy (PTT), PDT and immunotherapy for the treatment of cancer.
Collapse
Affiliation(s)
- Mengchao Ding
- School of Chemistry and Pharmaceutical Sciences, Qingdao Agricultural University, Qingdao, 266109, China.
| | | | | | | | | | | | | | | |
Collapse
|
63
|
Kumar AVP, Dubey SK, Tiwari S, Puri A, Hejmady S, Gorain B, Kesharwani P. Recent advances in nanoparticles mediated photothermal therapy induced tumor regression. Int J Pharm 2021; 606:120848. [PMID: 34216762 DOI: 10.1016/j.ijpharm.2021.120848] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/20/2021] [Accepted: 06/28/2021] [Indexed: 12/13/2022]
Abstract
Photothermal therapy (PTT) is a minimally invasive procedure for treating cancer. The two significant prerequisites of PTT are the photothermal therapeutic agent (PTA) and near-infrared radiation (NIR). The PTA absorbs NIR, causing hyperthermia in the malignant cells. This increased temperature at the tumor microenvironment finally results in tumor cell damage. Nanoparticles play a crucial role in PTT, aiding in the passive and active targeting of the PTA to the tumor microenvironment. Through enhanced permeation and retention effect and surface-engineering, specific targeting could be achieved. This novel delivery tool provides the advantages of changing the shape, size, and surface attributes of the carriers containing PTAs, which might facilitate tumor regression significantly. Further, inclusion of surface engineering of nanoparticles is facilitated through ligating ligands specific to overexpressed receptors on the cancer cell surface. Thus, transforming nanoparticles grants the ability to combine different treatment strategies with PTT to enhance cancer treatment. This review emphasizes properties of PTAs, conjugated biomolecules of PTAs, and the combinatorial techniques for a better therapeutic effect of PTT using the nanoparticle platform.
Collapse
Affiliation(s)
- Achalla Vaishnav Pavan Kumar
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Pilani Campus, Rajasthan 333031, India
| | - Sunil K Dubey
- R&D Healthcare Division, Emami Ltd, 13, BT Road, Belgharia, Kolkata 700056, India.
| | - Sanjay Tiwari
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Raebareli, Lucknow 226002, India
| | - Anu Puri
- RNA Structure and Design Section, RNA Biology Laboratory (RBL), Center for Cancer Research, National Cancer Institute-Frederick, Frederick, MD 21702, USA
| | - Siddhanth Hejmady
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani, Pilani Campus, Rajasthan 333031, India
| | - Bapi Gorain
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Selangor 47500, Malaysia
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
64
|
Varshney D, Qiu SY, Graf TP, McHugh KJ. Employing Drug Delivery Strategies to Overcome Challenges Using TLR7/8 Agonists for Cancer Immunotherapy. AAPS JOURNAL 2021; 23:90. [PMID: 34181117 DOI: 10.1208/s12248-021-00620-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/04/2021] [Indexed: 12/11/2022]
Abstract
Toll-like receptors (TLRs) are a potential target for cancer immunotherapy due to their role in the activation of the innate immune system. More specifically, TLR7 and TLR8, two structurally similar pattern recognition receptors that trigger interferon and cytokine responses, have proven to be therapeutically relevant targets for cancer in numerous preclinical and clinical studies. When triggered by an agonist, such as imiquimod or resiquimod, the TLR7/8 activation pathway induces cellular and humoral immune responses that can kill cancer cells with high specificity. Unfortunately, TLR7/8 agonists also present a number of issues that must be overcome prior to broad clinical implementation, such as poor drug solubility and systemic toxic effects. To overcome the key limitations of TLR7/8 agonists as a cancer therapy, biomaterial-based drug delivery systems have been developed. These delivery devices are highly diverse in their design and include systems that can be directly administered to the tumor, passively accumulated in relevant cancerous and lymph tissues, triggered by environmental stimuli, or actively targeted to specific physiological areas and cellular populations. In addition to improved delivery systems, recent studies have also demonstrated the potential benefits of TLR7/8 agonist co-delivery with other types of therapies, particularly checkpoint inhibitors, cancer vaccines, and chemotherapeutics, which can yield impressive anti-cancer effects. In this review, we discuss recent advances in the development of TLR7/8 agonist delivery systems and provide perspective on promising future directions.
Collapse
Affiliation(s)
- Dhruv Varshney
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, Texas, 77005, USA.,David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts, 02139, USA
| | - Sherry Yue Qiu
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, Texas, 77005, USA
| | - Tyler P Graf
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, Texas, 77005, USA
| | - Kevin J McHugh
- Department of Bioengineering, Rice University, 6100 Main Street, Houston, Texas, 77005, USA.
| |
Collapse
|
65
|
Gorbet MJ, Singh A, Mao C, Fiering S, Ranjan A. Using nanoparticles for in situ vaccination against cancer: mechanisms and immunotherapy benefits. Int J Hyperthermia 2021; 37:18-33. [PMID: 33426995 DOI: 10.1080/02656736.2020.1802519] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Immunotherapy to treat cancer is now an established clinical approach. Immunotherapy can be applied systemically, as done with checkpoint blockade antibodies, but it can also be injected directly into identified tumors, in a strategy of in situ vaccination (ISV). ISV is designed to stimulate a strong local antitumor immune response involving both innate and adaptive immune cells, and through this generate a systemic antitumor immune response against metastatic tumors. A variety of ISVs have been utilized to generate an immunostimulatory tumor microenvironment (TME). These include attenuated microorganisms, recombinant proteins, small molecules, physical disruptors of TME (alternating magnetic and focused ultrasound heating, photothermal therapy, and radiotherapy), and more recently nanoparticles (NPs). NPs are attractive and unique since they can load multiple drugs or other reagents to influence immune and cancer cell functions in the TME, affording a unique opportunity to stimulate antitumor immunity. Here, we describe the NP-ISV therapeutic mechanisms, review chemically synthesized NPs (i.e., liposomes, polymeric, chitosan-based, inorganic NPs, etc.), biologically derived NPs (virus and bacteria-based NPs), and energy-activated NP-ISVs in the context of their use as local ISV. Data suggests that NP-ISVs can enhance outcomes of immunotherapeutic regimens including those utilizing tumor hyperthermia and checkpoint blockade therapies.
Collapse
Affiliation(s)
| | - Akansha Singh
- College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, USA
| | - Chenkai Mao
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Steven Fiering
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.,Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center at Dartmouth and Dartmouth Hitchcock, Lebanon, NH, USA
| | - Ashish Ranjan
- College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, USA
| |
Collapse
|
66
|
Immunological effects of nano-enabled hyperthermia for solid tumors: opportunity and challenge. Front Chem Sci Eng 2021. [DOI: 10.1007/s11705-021-2059-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
67
|
Huang X, Lu Y, Guo M, Du S, Han N. Recent strategies for nano-based PTT combined with immunotherapy: from a biomaterial point of view. Theranostics 2021; 11:7546-7569. [PMID: 34158866 PMCID: PMC8210617 DOI: 10.7150/thno.56482] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 05/23/2021] [Indexed: 01/04/2023] Open
Abstract
Cancer has been a great threat to humans for decades. Due to the limitations of monotherapy, combinational therapies such as photothermal therapy (PTT) and immunotherapy have gained increasing attention with expectation to overcome the shortfalls of each other and obtain satisfactory therapeutic outcomes. PTT can inhibit primary tumors by thermal ablation but usually fails to achieve complete eradication and cannot prevent metastasis and recurrence. Meanwhile, the efficacy of immunotherapy is usually attenuated by the weak immunogenicity of tumor and the immunosuppressive tumor microenvironment (ITM). Therefore, many recent studies have attempted to synergize PTT with immunotherapy in order to enhance the therapeutic efficacy. In this review, we aim to summarize the cutting-edge strategies in combining nano-based PTT with immunotherapy for cancer treatment. Herein, the combination strategies were mainly classified into four categories, including 1) nano-based PTT combined with antigens to induce host immune responses; 2) nano-based PTT in combination with immune adjuvants acting as in situ vaccines; 3) nano-based PTT synergized with immune checkpoint blockade or other regulators to relieve the ITM; 4) nano-based PTT combined with CAR-T therapy or cytokine therapy for tumor treatment. The characteristics of various photothermal agents and nanoplatforms as well as the immunological mechanisms for the synergism were also introduced in detail. Finally, we discussed the existing challenges and future prospects in combined PTT and immunotherapy.
Collapse
Affiliation(s)
| | | | | | - Shouying Du
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Ning Han
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| |
Collapse
|
68
|
Xie X, Wang J, Zhang L, Zeng S, Su X, Chen Q. Bioresorbable Depot for Sustained Release of Immunostimulatory Resiquimod in Suppressing Both Primary Triple-Negative Breast Tumors and Metastatic Occurrence. Bioconjug Chem 2021; 32:1008-1016. [PMID: 33882675 DOI: 10.1021/acs.bioconjchem.1c00171] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In light of immune facilities trafficking toward the pathological sites along upward gradient of immunostimulatory cytokines, a localized resiquimod (Toll-like receptor 7/8 agonist) release depot was manufactured for pursuit of precision immunostimulation toward intractable triple-negative breast carcinoma. In principle, resiquimod/poly(lactic-co-glycolic acid) microspheres were fabricated and embedded into injectable and biodegradable poly(ethylene glycol) (PEG)-based hydrogel. The subsequent investigations approved persistent retention of immunostimulatory resiquimod in tumors upon peritumoral administration, which consequently led to localized and consistent secretion of immunostimulatory cytokines. Initially, not only innate tumor phagocytosis but also adaptive antitumor immunities were successfully cultivated for in situ suppression of the growth of primary solid tumors, more importantly, capable of inhibiting distant pulmonary metastasis, as evidenced by observation of enormous lymphocytes selectively gathering in the pulmonary artery. Hence, our presented study provided an important clinical indication of using immunostimulatory drugs to activate potent innate and adaptive antitumor immunities for precision antitumor therapy. Further immunomodulatory strategies, such as checkpoint blockage and tumor immunogenicity, could also be complementary for development of advanced antitumor immunotherapeutics in treatment of a number of intractable tumors.
Collapse
Affiliation(s)
- Xizhe Xie
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, China
- School of Bioengineering, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, China
| | - Jingyun Wang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, China
- School of Bioengineering, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, China
| | - Liuwei Zhang
- School of Bioengineering, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, China
| | - Shuang Zeng
- School of Bioengineering, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, China
| | - Xiaohui Su
- Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No. 44 Xiaoheyan Road, Dadong District, Shenyang 110042, China
| | - Qixian Chen
- School of Bioengineering, Dalian University of Technology, No. 2 Linggong Road, Dalian 116024, China
| |
Collapse
|
69
|
Yi X, Duan QY, Wu FG. Low-Temperature Photothermal Therapy: Strategies and Applications. RESEARCH (WASHINGTON, D.C.) 2021; 2021:9816594. [PMID: 34041494 PMCID: PMC8125200 DOI: 10.34133/2021/9816594] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 03/01/2021] [Indexed: 12/13/2022]
Abstract
Although photothermal therapy (PTT) with the assistance of nanotechnology has been considered as an indispensable strategy in the biomedical field, it still encounters some severe problems that need to be solved. Excessive heat can induce treated cells to develop thermal resistance, and thus, the efficacy of PTT may be dramatically decreased. In the meantime, the uncontrollable diffusion of heat can pose a threat to the surrounding healthy tissues. Recently, low-temperature PTT (also known as mild PTT or mild-temperature PTT) has demonstrated its remarkable capacity of conquering these obstacles and has shown excellent performance in bacterial elimination, wound healing, and cancer treatments. Herein, we summarize the recently proposed strategies for achieving low-temperature PTT based on nanomaterials and introduce the synthesis, characteristics, and applications of these nanoplatforms. Additionally, the combination of PTT and other therapeutic modalities for defeating cancers and the synergistic cancer therapeutic effect of the combined treatments are discussed. Finally, the current limitations and future directions are proposed for inspiring more researchers to make contributions to promoting low-temperature PTT toward more successful preclinical and clinical disease treatments.
Collapse
Affiliation(s)
- Xiulin Yi
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, China
| | - Qiu-Yi Duan
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, China
| | - Fu-Gen Wu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing 210096, China
| |
Collapse
|
70
|
Wang H, Guo Y, Gan S, Liu H, Chen Q, Yuan A, Hu Y, Wu J. Photosynthetic Microorganisms-Based Biophotothermal Therapy with Enhanced Immune Response. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2007734. [PMID: 33738929 DOI: 10.1002/smll.202007734] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/21/2021] [Indexed: 06/12/2023]
Abstract
The production of oxygen by photosynthetic microorganisms (PSMs) has recently attracted interest concerning the in vivo treatment of multiple diseases for their photosynthetic oxygen production in vivo, since PSMs have good biological safety. Here, the first evidence that PSMs can be used as a photothermal source to perform biophotothermal therapy (bio-PTT) is provided. In vitro and in vivo experiments proved that PSMs can generate heat for the direct elimination of tumors and release a series of pathogen-associated molecular patterns and adjuvants for immune stimulation under light irradiation. Bio-PTT enabled a local tumor inhibition rate exceeding 90% and an abscopal tumor inhibition rate exceeding 75%. This strategy also produced a stronger antitumor immune memory effect to prevent tumor recurrence. The bio-PTT strategy provides a novel direction for photothermal therapy as it simultaneously produces local and abscopal antitumor effects.
Collapse
Affiliation(s)
- Haoran Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Sciences, Nanjing University, Nanjing, 210093, China
| | - Yunfei Guo
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Sciences, Nanjing University, Nanjing, 210093, China
| | - Shaoju Gan
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Sciences, Nanjing University, Nanjing, 210093, China
| | - Honghui Liu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Sciences, Nanjing University, Nanjing, 210093, China
| | - Qian Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Sciences, Nanjing University, Nanjing, 210093, China
| | - Ahu Yuan
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Sciences, Nanjing University, Nanjing, 210093, China
- Institute of Drug R&D, Nanjing University, Nanjing, 210093, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, 210093, China
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Sciences, Nanjing University, Nanjing, 210093, China
- Institute of Drug R&D, Nanjing University, Nanjing, 210093, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, 210093, China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School and School of Life Sciences, Nanjing University, Nanjing, 210093, China
- Institute of Drug R&D, Nanjing University, Nanjing, 210093, China
- Jiangsu Key Laboratory for Nano Technology, Nanjing University, Nanjing, 210093, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing, 210093, China
| |
Collapse
|
71
|
Fu L, Zhou X, He C. Polymeric Nanosystems for Immunogenic Cell Death-Based Cancer Immunotherapy. Macromol Biosci 2021; 21:e2100075. [PMID: 33885225 DOI: 10.1002/mabi.202100075] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/26/2021] [Indexed: 12/11/2022]
Abstract
Immunotherapy has pointed out a scientific and promising direction for cancer treatment through the rouse of immunosurveillance and the decrease of possible side effects in recent years. In immunotherapy, immunogenic cancer cell death (ICD) plays a critical role in regulating anti-cancer immune system in vivo via the release of damage-associated molecular patterns. ICD can not only induce in situ cancer cells apoptosis, but also arouse the immune response against metastatic tumors, which is of great clinical significance to eradicate tumors. In cancer immunotherapy, polymer nanoparticles have drawn increasing attention as an important component of ICD-based immunotherapy attributing to their controllable size, excellent biocompatibility, promising ability of protecting cargo from surrounding environment, which delivers the antigens or immune inducers to antigen-presenting cells, and further triggers sinnvoll T cell response. In this review, the recent advances in the development of polymeric material-based nanosystems for ICD-mediated cancer immunotherapy are summarized. The mechanism of ICD and some current restrictions inhibiting the efficiency of immunotherapy and future prospects are also discussed.
Collapse
Affiliation(s)
- Liwen Fu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, China
| | - Xiaojun Zhou
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, China
| | - Chuanglong He
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, 201620, China
| |
Collapse
|
72
|
Lin W, Li C, Xu N, Watanabe M, Xue R, Xu A, Araki M, Sun R, Liu C, Nasu Y, Huang P. Dual-Functional PLGA Nanoparticles Co-Loaded with Indocyanine Green and Resiquimod for Prostate Cancer Treatment. Int J Nanomedicine 2021; 16:2775-2787. [PMID: 33880023 PMCID: PMC8052122 DOI: 10.2147/ijn.s301552] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/24/2021] [Indexed: 12/28/2022] Open
Abstract
PURPOSE With the advance of screening techniques, there is a growing number of low-risk or intermediate-risk prostate cancer (PCa) cases, remaining a serious threat to men's health. To obtain better efficacy, a growing interest has been attracted to develop such emerging treatments as immunotherapy and focal therapy. However, few studies offer guidance on whether and how to combine these modalities against PCa. This study was designed to develop dual-functional nanoparticles (NPs) which combined photothermal therapy (PTT) with immunotherapy and determine the anti-tumor efficacy for PCa treatment. METHODS By a double emulsion technique, the drug nanocarrier, poly(lactic-co-glycolic acid) or PLGA, was applied for co-loading of a fluorescent dye, indocyanine green (ICG) and a toll-like receptor 7/8 (TLR7/8) agonist resiquimod (R848) to synthesize PLGA-ICG-R848 NPs. Next, we determined their characteristic features and evaluated whether they inhibited the cell viability in multiple PCa cell lines. After treatment with PLGA-ICG-R848, the maturation markers of bone marrow-derived dendritic cells (BMDCs) were detected by flow cytometry. By establishing a subcutaneous xenograft model of mouse PCa, we explored both the anti-tumor effect and immune response following the NPs-based laser ablation. RESULTS With a mean diameter of 157.7 nm, PLGA-ICG-R848 exhibited no cytotoxic effect in PCa cells, but they significantly decreased RM9 cell viability to (3.9±1.0)% after laser irradiation. Moreover, PLGA-ICG-R848 promoted BMDCs maturation with the significantly elevated proportions of CD11c+CD86+ and CD11c+CD80+ cells. Following PLGA-ICG-R848-based laser ablation in vivo, the decreased bioluminescent signals indicated a significant inhibition of PCa growth, while the ratio of splenic natural killer (NK) cells in PLGA-ICG-R848 was (3.96±1.88)% compared with (0.99±0.10)% in PBS group, revealing the enhanced immune response against PCa. CONCLUSION The dual-functional PLGA-ICG-R848 NPs under laser irradiation exhibit the anti-tumor efficacy for PCa treatment by combining PTT with immunotherapy.
Collapse
Affiliation(s)
- Wenfeng Lin
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Chaoming Li
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Naijin Xu
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Masami Watanabe
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama, Japan
| | - Ruizhi Xue
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Abai Xu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Motoo Araki
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Ruifen Sun
- Center for Scientific Research, Yunnan University of Chinese Traditional Medicine, Kunming, People’s Republic of China
| | - Chunxiao Liu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
| | - Yasutomo Nasu
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Peng Huang
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, People’s Republic of China
- Okayama Medical Innovation Center, Okayama University, Okayama, Japan
| |
Collapse
|
73
|
Zhang Y, Zhang G, Wang G, Wu L, Monteiro-Riviere NA, Li Y. The synergistic strategies for the immuno-oncotherapy with photothermal nanoagents. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1717. [PMID: 33825343 DOI: 10.1002/wnan.1717] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/12/2021] [Accepted: 03/09/2021] [Indexed: 12/13/2022]
Abstract
Immuno-oncotherapy has shown great promise for the cure of late-stage and metastatic cancer. Great efforts have tried to improve the overall response rate (ORR) and to reduce the immune-related adverse events (irAEs). Antigen presentation, T cell activation and killing are interlocking and distinct steps to initiate effective anti-tumor immune responses. Aiming to overcome the tumor immune evasion whose mechanisms include limited release of neoantigen, suppressed infiltration of antigen-presenting cells (APCs) and T cells, and the expression of immune checkpoints (ICPs), combinational therapeutic strategies have shown great potential by activating the anti-tumor immune responses together with deactivating immunosuppressive conditions simultaneously. In this direction, photothermal therapy (PTT) has attracted attention due to the efficient ablation of tumor cells, of which the released immunogenic tumor debris can activate host immune responses. The combination of immunoadjuvants and/or ICP inhibitors can boost the anti-tumor immune responses, realizing PTT-synergized immuno-oncotherapy. In this regard, numerous multifunctional nanomaterials have been designed with integration of photothermal and immuno-oncotherapeutic agents into one package via well-designed surface modification and functionalization. This review summarizes the recent studies on the synergistic strategies for the immuno-oncotherapy based on photothermal nanoagents and the mechanisms that trigger the systemic anti-tumor immune responses and PTT-synergized immuno-oncotherapy. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Yuqian Zhang
- Laboratory of Immunology and Nanomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Guofang Zhang
- Laboratory of Immunology and Nanomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Guocheng Wang
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Lidong Wu
- Key Laboratory of Control of Quality and Safety for Aquatic Products, Chinese Academy of Fishery Sciences, Beijing, China
| | - Nancy A Monteiro-Riviere
- Nanotechnology Innovation Center of Kansas State, Kansas State University, Manhattan, Kansas, USA
| | - Yang Li
- Laboratory of Immunology and Nanomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
74
|
Duan Y, Yang H, Gao J, Wei D, Zhang Y, Wang J, Zhang X, Zhang J, Ge K, Wu X, Chang J. Immune Modulator and Low-Temperature PTT-Induced Synergistic Immunotherapy for Cancer Treatment. ACS APPLIED BIO MATERIALS 2021; 4:1524-1535. [PMID: 35014502 DOI: 10.1021/acsabm.0c01397] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Immunotherapy has shown great potential in cancer therapeutics but has limitations of the insufficient activation of dendritic cells (DCs) and immune-suppressive microenvironment. To overcome these obstacles, a cascade synergistic immunotherapy nanosystem (denoted as CpG@PDA-FA) was designed to elevate anticancer immune response. The combination nanosystem including a photothermal agent polydopamine (PDA) and immunomodulator CpG oligodeoxynucleotides (CpG ODNs). On the one hand, polydopamine (PDA) acts as a photothermal agent to induce low-temperature PTT. It leads to immunogenic cell death (ICD), a programmed cell death pathway, which can activate DCs and enhance the antitumor immune response of T cells. On the other hand, CpG ODNs further promote maturation and migration of DCs as well as ameliorates the immunosuppression microenvironment of the tumor (TME). This paper focuses on a cancer synergistic treatment of ICD-induced immunotherapy by low-temperature PTT and ameliorates TME by immunomodulator CpG ODNs. We proved that CpG@PDA-FA NPs realized a remarkable synergistic treatment effect compared with respective single PTT or CpG therapy in the maturation of DCs and activation of T cells. In addition, CpG@PDA-FA NPs also reduced myeloid-derived suppressor cells and regulatory T cells to relieve immunosuppression. Hence, CpG@PDA-FA NPs provide a bidirectional immunotherapy strategy for tumor inhibition and highlight the cascade effects of low-temperature PTT and immunotherapy.
Collapse
Affiliation(s)
- Yue Duan
- School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, P. R. China
| | - Han Yang
- School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, P. R. China
| | - Junxiao Gao
- School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, P. R. China
| | - Daohe Wei
- School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, P. R. China
| | - Yue Zhang
- School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, P. R. China
| | - Jian Wang
- School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, P. R. China
| | - Xinhui Zhang
- School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, P. R. China
| | - Jinchao Zhang
- Key Laboratory of Analytical Science and Technology of Hebei Province, College of Chemistry and Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding 071002, P. R. China
| | - Kun Ge
- Key Laboratory of Analytical Science and Technology of Hebei Province, College of Chemistry and Environmental Science, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of the Ministry of Education, Hebei University, Baoding 071002, P. R. China
| | - Xiaoli Wu
- School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, P. R. China
| | - Jin Chang
- School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin 300072, P. R. China
| |
Collapse
|
75
|
Roma-Rodrigues C, Raposo LR, Valente R, Fernandes AR, Baptista PV. Combined cancer therapeutics-Tackling the complexity of the tumor microenvironment. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1704. [PMID: 33565269 DOI: 10.1002/wnan.1704] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/05/2021] [Accepted: 01/12/2021] [Indexed: 12/12/2022]
Abstract
Cancer treatment has yet to find a "silver bullet" capable of selectively and effectively kill tumor cells without damaging healthy cells. Nanomedicine is a promising field that can combine several moieties in one system to produce a multifaceted nanoplatform. The tumor microenvironment (TME) is considered responsible for the ineffectiveness of cancer therapeutics and the difficulty in the translation from the bench to bed side of novel nanomedicines. A promising approach is the use of combinatorial therapies targeting the TME with the use of stimuli-responsive nanomaterials which would increase tumor targeting. Contemporary combined strategies for TME-targeting nanoformulations are based on the application of external stimuli therapies, such as photothermy, hyperthermia or ultrasounds, in combination with stimuli-responsive nanoparticles containing a core, usually composed by metal oxides or graphene, and a biocompatible stimuli-responsive coating layer that could also contain tumor targeting moieties and a chemotherapeutic agent to enhance the therapeutic efficacy. The obstacles that nanotherapeutics must overcome in the TME to accomplish an effective therapeutic cargo delivery and the proposed strategies for improved nanotherapeutics will be reviewed. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Catarina Roma-Rodrigues
- UCIBIO, Department of Life Sciences, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Luís R Raposo
- UCIBIO, Department of Life Sciences, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Rúben Valente
- UCIBIO, Department of Life Sciences, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Alexandra R Fernandes
- UCIBIO, Department of Life Sciences, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| | - Pedro V Baptista
- UCIBIO, Department of Life Sciences, Faculdade de Ciências e Tecnologia, Universidade NOVA de Lisboa, Caparica, Portugal
| |
Collapse
|
76
|
Uthaman S, Pillarisetti S, Hwang HS, Mathew AP, Huh KM, Rhee JH, Park IK. Tumor Microenvironment-Regulating Immunosenescence-Independent Nanostimulant Synergizing with Near-Infrared Light Irradiation for Antitumor Immunity. ACS APPLIED MATERIALS & INTERFACES 2021; 13:4844-4852. [PMID: 33486952 DOI: 10.1021/acsami.0c20063] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The combination of photothermal therapy (PTT) and toll-like receptor (TLR)-mediated immunotherapy can elicit antitumor immunity and modulate the immunosuppressive tumor microenvironment (TME). Unlike other TLRs, TLR-5 is a promising target for immune activation, as its expression is well-maintained even during immunosenescence. Here, we developed a unique tumor microenvironment-regulating immunosenescence-independent nanostimulant consisting of TLR-5 adjuvant Vibrio vulnificus flagellin B (FlaB) conjugated onto the surface to an IR 780-loaded hyaluronic acid-stearylamine (HIF) micelles. These HIF micelles induced immune-mediated cell death via PTT when irradiated with a near-infrared laser. In comparison with PTT alone, the combination of in situ-generated tumor-associated antigens produced during PTT and the immune adjuvant FlaB demonstrated enhanced vaccine-like properties and modulated the TME by suppressing immune-suppressive regulatory cells (Tregs) and increasing the fraction of CD103+ migratory dendritic cells, which are responsible for trafficking tumor antigens to draining lymph nodes (DLNs). This combinatorial strategy (i.e., applying a TLR-5 adjuvant targeted to immunosenescence-independent TLR-5 and the in situ photothermal generation of tumor-associated antigens) is a robust system for next-generation immunotherapy and could even be applied in elderly patients, thus broadening the clinical scope of immunotherapy strategies.
Collapse
Affiliation(s)
- Saji Uthaman
- Department of Polymer Science and Engineering, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Shameer Pillarisetti
- Department of Biomedical Sciences, Chonnam National University Medical School, 160 Baekseo-ro, Gwangju 58128, Republic of Korea
| | - Hye Suk Hwang
- Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Jeonnam 58128, Republic of Korea
| | - Ansuja Pulickal Mathew
- Department of Biomedical Sciences, Chonnam National University Medical School, 160 Baekseo-ro, Gwangju 58128, Republic of Korea
| | - Kang Moo Huh
- Department of Polymer Science and Engineering, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon 34134, Republic of Korea
| | - Joon Haeng Rhee
- Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Jeonnam 58128, Republic of Korea
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam 58128, Republic of Korea
| | - In-Kyu Park
- Department of Biomedical Sciences, Chonnam National University Medical School, 160 Baekseo-ro, Gwangju 58128, Republic of Korea
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam 58128, Republic of Korea
- Immunotherapy Innovation Center, Chonnam National University, Hwasun-gun, Jeonnam 58128, Republic of Korea
| |
Collapse
|
77
|
Li J, Luo Y, Pu K. Electromagnetic Nanomedicines for Combinational Cancer Immunotherapy. Angew Chem Int Ed Engl 2021; 60:12682-12705. [DOI: 10.1002/anie.202008386] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Indexed: 12/12/2022]
Affiliation(s)
- Jingchao Li
- School of Chemical and Biomedical Engineering Nanyang Technological University 70 Nanyang Drive Singapore 637457 Singapore
| | - Yu Luo
- School of Chemical Science and Engineering Tongji University 1239 Siping Road Shanghai 200092 China
| | - Kanyi Pu
- School of Chemical and Biomedical Engineering Nanyang Technological University 70 Nanyang Drive Singapore 637457 Singapore
| |
Collapse
|
78
|
Li J, Luo Y, Pu K. Electromagnetic Nanomedicines for Combinational Cancer Immunotherapy. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202008386] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Jingchao Li
- School of Chemical and Biomedical Engineering Nanyang Technological University 70 Nanyang Drive Singapore 637457 Singapore
| | - Yu Luo
- School of Chemical Science and Engineering Tongji University 1239 Siping Road Shanghai 200092 China
| | - Kanyi Pu
- School of Chemical and Biomedical Engineering Nanyang Technological University 70 Nanyang Drive Singapore 637457 Singapore
| |
Collapse
|
79
|
Cheng H, Tsao H, Chiang C, Chen S. Advances in Magnetic Nanoparticle-Mediated Cancer Immune-Theranostics. Adv Healthc Mater 2021; 10:e2001451. [PMID: 33135398 DOI: 10.1002/adhm.202001451] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/12/2020] [Indexed: 12/13/2022]
Abstract
Cancer immunotherapy is a cutting-edge strategy that eliminates cancer cells by amplifying the host's immune system. However, the low response rate and risks of inducing systemic toxicity have raised uncertainty in the treatment. Magnetic nanoparticles (MNPs) as a versatile theranostic tool can be used to target delivery of multiple immunotherapeutics and monitor cell/tissue responses. These capabilities enable the real-time characterization of the factors that contribute to immunoactivity so that future treatments can be optimized. The magnetic properties of MNPs further allow the implementation of magnetic navigation and magnetic hyperthermia for boosting the efficacy of immunotherapy. The multimodal approach opens an avenue to induce robust immune responses, minimize safety issues, and monitor immune activities simultaneously. Thus, the object of this review is to provide an overview of the burgeoning fields and to highlight novel technologies for next-generation immunotherapy. The review further correlates the properties of MNPs with the latest treatment strategies to explore the crosstalk between magnetic nanomaterials and the immune system. This comprehensive review of MNP-derived immunotherapy covers the obstacles and opportunities for future development and clinical translation.
Collapse
Affiliation(s)
- Hung‐Wei Cheng
- Department of Materials Science and Engineering National Chiao Tung University Hsinchu 30010 Taiwan
| | - Hsin‐Yi Tsao
- Department of Materials Science and Engineering National Chiao Tung University Hsinchu 30010 Taiwan
| | - Chih‐Sheng Chiang
- Cell Therapy Center China Medical University Hospital Taichung 40421 Taiwan
| | - San‐Yuan Chen
- Department of Materials Science and Engineering National Chiao Tung University Hsinchu 30010 Taiwan
- Frontier Research Centre on Fundamental and Applied Sciences of Matters National Tsing Hua University Hsinchu 30013 Taiwan
- School of Dentistry College of Dental Medicine Kaohsiung Medical University Kaohsiung 807378 Taiwan
- Graduate Institute of Biomedical Science China Medical University Taichung 40421 Taiwan
| |
Collapse
|
80
|
Stephen ZR, Zhang M. Recent Progress in the Synergistic Combination of Nanoparticle-Mediated Hyperthermia and Immunotherapy for Treatment of Cancer. Adv Healthc Mater 2021; 10:e2001415. [PMID: 33236511 PMCID: PMC8034553 DOI: 10.1002/adhm.202001415] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/11/2020] [Indexed: 02/06/2023]
Abstract
Immunotherapy has demonstrated great clinical success in certain cancers, driven primarily by immune checkpoint blockade and adoptive cell therapies. Immunotherapy can elicit strong, durable responses in some patients, but others do not respond, and to date immunotherapy has demonstrated success in only a limited number of cancers. To address this limitation, combinatorial approaches with chemo- and radiotherapy have been applied in the clinic. Extensive preclinical evidence suggests that hyperthermia therapy (HT) has considerable potential to augment immunotherapy with minimal toxicity. This progress report will provide a brief overview of immunotherapy and HT approaches and highlight recent progress in the application of nanoparticle (NP)-based HT in combination with immunotherapy. NPs allow for tumor-specific targeting of deep tissue tumors while potentially providing more even heating. NP-based HT increases tumor immunogenicity and tumor permeability, which improves immune cell infiltration and creates an environment more responsive to immunotherapy, particularly in solid tumors.
Collapse
Affiliation(s)
- Zachary R Stephen
- Department of Materials Science and Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Miqin Zhang
- Department of Materials Science and Engineering, Department of Neurological Surgery, University of Washington, Seattle, WA, 98195, USA
| |
Collapse
|
81
|
Chang M, Hou Z, Wang M, Li C, Lin J. Recent Advances in Hyperthermia Therapy-Based Synergistic Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2004788. [PMID: 33289219 DOI: 10.1002/adma.202004788] [Citation(s) in RCA: 233] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/17/2020] [Indexed: 06/12/2023]
Abstract
The past decades have witnessed hyperthermia therapy (HTT) as an emerging strategy against malignant tumors. Nanomaterial-based photothermal therapy (PTT) and magnetic hyperthermia (MHT), as highly effective and noninvasive treatment models, offer advantages over other strategies in the treatment of different types of tumors. However, both PTT and MHT cannot completely cure cancer due to recurrence and distal metastasis. In recent years, cancer immunotherapy has attracted widespread attention owing to its capability to activate the body's own natural defense to identify, attack, and eradicate cancer cells. Significant efforts have been devoted to studying the activated immune responses caused by hyperthermia-ablated tumors. In this article, the synergistic mechanism of HTT in immunotherapy, including immunogenic cell death and reversal of the immunosuppressive tumor microenvironment is discussed. The reports of the combination of HTT or HTT-based multimodal therapy with immunotherapy, including immunoadjuvant exploitation, immune checkpoint blockade therapy, and adoptive cellular immunotherapy are summarized. As highlighted, these strategies could achieve synergistically enhanced therapeutic outcomes against both primary tumors and metastatic lesions, prevent cancer recurrence, and prolong the survival period. Finally, current challenges and prospective developments in HTT-synergized immunotherapy are also reviewed.
Collapse
Affiliation(s)
- Mengyu Chang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Sciences and Technology of China, Hefei, 230026, P. R. China
| | - Zhiyao Hou
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, Guangdong, 511436, P. R. China
- Department of Abdominal Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, P. R. China
| | - Man Wang
- Institute of Molecular Sciences and Engineering, Shandong University, Qingdao, 266237, P. R. China
| | - Chunxia Li
- Institute of Molecular Sciences and Engineering, Shandong University, Qingdao, 266237, P. R. China
| | - Jun Lin
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Sciences and Technology of China, Hefei, 230026, P. R. China
| |
Collapse
|
82
|
Huang X, Shi Q, Du S, Lu Y, Han N. Poly-tannic acid coated paclitaxel nanocrystals for combinational photothermal-chemotherapy. Colloids Surf B Biointerfaces 2021; 197:111377. [DOI: 10.1016/j.colsurfb.2020.111377] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 08/25/2020] [Accepted: 09/16/2020] [Indexed: 02/02/2023]
|
83
|
Engineering immunogenic cell death with nanosized drug delivery systems improving cancer immunotherapy. Curr Opin Biotechnol 2020; 66:36-43. [DOI: 10.1016/j.copbio.2020.06.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/11/2020] [Accepted: 06/13/2020] [Indexed: 02/06/2023]
|
84
|
Li J, Burgess DJ. Nanomedicine-based drug delivery towards tumor biological and immunological microenvironment. Acta Pharm Sin B 2020; 10:2110-2124. [PMID: 33304781 PMCID: PMC7714990 DOI: 10.1016/j.apsb.2020.05.008] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/15/2020] [Accepted: 05/21/2020] [Indexed: 12/17/2022] Open
Abstract
The complex tumor microenvironment is a most important factor in cancer development. The biological microenvironment is composed of a variety of barriers including the extracellular matrix and associated cells such as endothelia cells, pericytes, and cancer-associated fibroblasts. Different strategies can be utilized to enhance nanoparticle-based drug delivery and distribution into tumor tissues addressing the extracellular matrix or cellular components. In addition to the biological microenvironment, the immunological conditions around the tumor tissue can be very complicated and cancer cells have various ways of evading immune surveillance. Nanoparticle drug delivery systems can enhance cancer immunotherapy by tuning the immunological response and memory of various immune cells such as T cells, B cells, macrophages, and dendritic cells. In this review, the main components in the tumor biological and immunological environment are discussed. The focus is on recent advances in nanoparticle-based drug delivery systems towards targets within the tumor microenvironment to improve cancer chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Jin Li
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269-3092, USA
| | - Diane J. Burgess
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT 06269-3092, USA
| |
Collapse
|
85
|
Shang T, Yu X, Han S, Yang B. Nanomedicine-based tumor photothermal therapy synergized immunotherapy. Biomater Sci 2020; 8:5241-5259. [PMID: 32996922 DOI: 10.1039/d0bm01158d] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The emerging anti-tumor immunotherapy has made significant progress in clinical application. However, single immunotherapy is not effective for all anti-tumor treatments, owing to the low objective response rate and the risk of immune-related side effects. Meanwhile, photothermal therapy (PTT) has attracted significant attention because of its non-invasiveness, spatiotemporal controllability and small side effects. Combining PTT with immunotherapy overcomes the issue that single photothermal therapy cannot eradicate tumors with metastasis and recurrence. However, it improves the therapeutic effect of immunotherapy, as the photothermal therapy usually promotes release of tumor-related antigens, triggers immune response by the immunogenic cell death (ICD), thereby, endowing unique synergistic mechanisms for cancer therapy. This review summarizes recent research advances in utilizing nanomedicines for PTT in combination with immunotherapy to improve the outcome of cancer treatment. The strategies include immunogenic cell death, immune agonists and cancer vaccines, immune checkpoint blockades and tumor specific monoclonal antibodies, and small-molecule immune inhibitors. The combination of synergized PTT-immunotherapy with other therapeutic strategies is also discussed.
Collapse
Affiliation(s)
- Tongyi Shang
- The Sixth Affiliated Hospital, Department of Biomedical Engineering, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, P. R. China.
| | | | | | | |
Collapse
|
86
|
Korupalli C, Kalluru P, Nuthalapati K, Kuthala N, Thangudu S, Vankayala R. Recent Advances of Polyaniline-Based Biomaterials for Phototherapeutic Treatments of Tumors and Bacterial Infections. Bioengineering (Basel) 2020; 7:E94. [PMID: 32823566 PMCID: PMC7552745 DOI: 10.3390/bioengineering7030094] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/05/2020] [Accepted: 08/11/2020] [Indexed: 01/01/2023] Open
Abstract
Conventional treatments fail to completely eradicate tumor or bacterial infections due to their inherent shortcomings. In recent years, photothermal therapy (PTT) has emerged as an attractive treatment modality that relies on the absorption of photothermal agents (PTAs) at a specific wavelength, thereby transforming the excitation light energy into heat. The advantages of PTT are its high efficacy, specificity, and minimal damage to normal tissues. To this end, various inorganic nanomaterials such as gold nanostructures, carbon nanostructures, and transition metal dichalcogenides have been extensively explored for PTT applications. Subsequently, the focus has shifted to the development of polymeric PTAs, owing to their unique properties such as biodegradability, biocompatibility, non-immunogenicity, and low toxicity when compared to inorganic PTAs. Among various organic PTAs, polyaniline (PANI) is one of the best-known and earliest-reported organic PTAs. Hence, in this review, we cover the recent advances and progress of PANI-based biomaterials for PTT application in tumors and bacterial infections. The future prospects in this exciting area are also addressed.
Collapse
Affiliation(s)
- Chiranjeevi Korupalli
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan;
| | - Poliraju Kalluru
- Department of Chemistry, University of Calgary, Calgary, AB T2N1N4, Canada;
| | - Karthik Nuthalapati
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan; (K.N.); (N.K.); (S.T.)
| | - Naresh Kuthala
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan; (K.N.); (N.K.); (S.T.)
| | - Suresh Thangudu
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan; (K.N.); (N.K.); (S.T.)
| | - Raviraj Vankayala
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan 342037, India
| |
Collapse
|
87
|
Piao HY, Guo S, Wang Y, Zhang J. Exosome-transmitted lncRNA PCGEM1 promotes invasive and metastasis in gastric cancer by maintaining the stability of SNAI1. Clin Transl Oncol 2020; 23:246-256. [PMID: 32519176 DOI: 10.1007/s12094-020-02412-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 05/25/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE Clinically, hypoxia is associated with increased distant metastasis and poor survival in gastric cancer (GC). In this study, we set out from the cellular interaction to further explain the molecular mechanism of invasion in GC cells under hypoxic conditions. METHODS Gastric cancer cells were cultured under 1% O2 (hypoxia-cultured gastric cancer cells, HGC) and 20% O2 condition (normoxic-cultured gastric cancer cells, NGC). NGC was co-cultured with HGC-medium. Scrape and Transwell were used to evaluate invasion and migration. Exosomes from GC were extracted by ultracentrifugation. Electron microscopy images, western-blot used to analyze the size distributions and the number of exosomes. RESULTS HGC-medium induced NGC dissociated. Long non-coding RNA (lncRNA) prostate cancer gene expression marker 1 (PCGEM1) was specifically expressed in HGC exosomes. HGC-derived PCGEM1-riched exosomes could promote the invasion and migration of NGC. On the mechanism, PCGEM1 maintained stability and reduced the degradation of SNAI1, which could induce the epithelial-mesenchymal transition of GC. CONCLUSION LncRNA PCGEM1 was overexpressed in GC cells. And part of the PCGEM1 can be encapsulated into exosomes. These exosomes promoted invasion and migration of other GC cells. We considered PCGEM1 might act as a "scaffold" combined with SNAI1 and prompt the invasion and migration of GC.
Collapse
Affiliation(s)
- H-Y Piao
- Medical Oncology Department of Gastrointestinal Cancer, Liaoning Province Cancer Hospital and Institute (Cancer Hospital of China Medical University), No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning, China
| | - S Guo
- Gastric Cancer Department, Liaoning Province Cancer Hospital and Institute (Cancer Hospital of China Medical University), No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning, China
| | - Y Wang
- Gastric Cancer Department, Liaoning Province Cancer Hospital and Institute (Cancer Hospital of China Medical University), No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning, China
| | - J Zhang
- Gastric Cancer Department, Liaoning Province Cancer Hospital and Institute (Cancer Hospital of China Medical University), No. 44 Xiaoheyan Road, Dadong District, Shenyang, 110042, Liaoning, China.
| |
Collapse
|
88
|
Ni J, Zhang X, Yang G, Kang T, Lin X, Zha M, Li Y, Wang L, Li K. A Photoinduced Nonadiabatic Decay‐Guided Molecular Motor Triggers Effective Photothermal Conversion for Cancer Therapy. Angew Chem Int Ed Engl 2020; 59:11298-11302. [DOI: 10.1002/anie.202002516] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 04/05/2020] [Indexed: 01/14/2023]
Affiliation(s)
- Jen‐Shyang Ni
- Department of Biomedical Engineering SUSTech Academy for Advanced Interdisciplinary Studies Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
- HKUST-Shenzhen Research Institute Shenzhen 518057 China
| | - Xun Zhang
- Department of Biomedical Engineering SUSTech Academy for Advanced Interdisciplinary Studies Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Guang Yang
- Department of Biomedical Engineering SUSTech Academy for Advanced Interdisciplinary Studies Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Tianyi Kang
- Department of Biomedical Engineering SUSTech Academy for Advanced Interdisciplinary Studies Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Xiangwei Lin
- City University of Hong Kong Shenzhen Research Institute Shenzhen 518057 China
| | - Menglei Zha
- Department of Biomedical Engineering SUSTech Academy for Advanced Interdisciplinary Studies Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Yaxi Li
- Department of Biomedical Engineering SUSTech Academy for Advanced Interdisciplinary Studies Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Lidai Wang
- City University of Hong Kong Shenzhen Research Institute Shenzhen 518057 China
| | - Kai Li
- Department of Biomedical Engineering SUSTech Academy for Advanced Interdisciplinary Studies Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| |
Collapse
|
89
|
Ni J, Zhang X, Yang G, Kang T, Lin X, Zha M, Li Y, Wang L, Li K. A Photoinduced Nonadiabatic Decay‐Guided Molecular Motor Triggers Effective Photothermal Conversion for Cancer Therapy. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202002516] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Jen‐Shyang Ni
- Department of Biomedical Engineering SUSTech Academy for Advanced Interdisciplinary Studies Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
- HKUST-Shenzhen Research Institute Shenzhen 518057 China
| | - Xun Zhang
- Department of Biomedical Engineering SUSTech Academy for Advanced Interdisciplinary Studies Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Guang Yang
- Department of Biomedical Engineering SUSTech Academy for Advanced Interdisciplinary Studies Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Tianyi Kang
- Department of Biomedical Engineering SUSTech Academy for Advanced Interdisciplinary Studies Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Xiangwei Lin
- City University of Hong Kong Shenzhen Research Institute Shenzhen 518057 China
| | - Menglei Zha
- Department of Biomedical Engineering SUSTech Academy for Advanced Interdisciplinary Studies Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Yaxi Li
- Department of Biomedical Engineering SUSTech Academy for Advanced Interdisciplinary Studies Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| | - Lidai Wang
- City University of Hong Kong Shenzhen Research Institute Shenzhen 518057 China
| | - Kai Li
- Department of Biomedical Engineering SUSTech Academy for Advanced Interdisciplinary Studies Southern University of Science and Technology (SUSTech) Shenzhen 518055 China
| |
Collapse
|