51
|
Utilizing chemotherapy-induced tumor RNA nanoparticles to improve cancer chemoimmunotherapy. Acta Biomater 2023; 158:698-707. [PMID: 36563773 DOI: 10.1016/j.actbio.2022.12.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/11/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022]
Abstract
Chemotherapy has become a popular combination strategy to improve the response rate of immunotherapy since certain chemotherapeutic drugs kill tumor cells by an immunogenic cell death (ICD) pathway, which activates antitumor immune responses. Unfortunately, the synergistic effect of chemoimmunotherapy can be impaired due to the toxicities of chemotherapeutic agent-induced lymphatic depletion and immunosuppression. In this study, we present an approach to improve immunotherapy by using tumor RNA nanoparticles (RNA-NPs) where RNA is directly extracted from chemotherapy-treated cancer cells and then condensed by protamine via electrostatic interactions to form complexes. Such RNA-NPs can be effectively taken up by dendritic cells (DCs) in the draining lymph nodes after subcutaneous injection. Compared with noninduced tumor RNA nanoparticles (N-RNA-NPs), chemotherapy-induced tumor RNA nanoparticles (C-RNA-NPs) can significantly promote DC maturation and stimulate a stronger immune response against established CT-26 colon carcinoma. Besides, C-RNA-NPs can improve the efficacy of immune checkpoint blockade (ICB) therapy by facilitating the infiltration of intratumoral T cells and increasing the ratio of CD8+ T cells to regulatory T cells (Tregs). More importantly, the synergistic effect of chemoimmunotherapy is also enhanced by treatment with C-RNA-NPs. STATEMENT OF SIGNIFICANCE: Although immune checkpoint blockade therapy has been demonstrated to be effective in some advanced cancers, the low response rate has significantly limited its clinical application. To address this issue, a new strategy for improving cancer immunotherapy using chemotherapy-induced tumor RNA nanoparticles (C-RNA-NPs) is developed in this work. The proposed C-RNA-NPs could be captured by dendritic cells, which were then stimulated to the maturation status to initiate an anticancer immune response. Furthermore, the response rate to immunotherapy was significantly increased by promoting intratumoral T-cell infiltration and elevating the intratumoral ratio of CD8+ T cells to regulatory T cells after treatment with C-RNA-NPs. Therefore, C-RNA-NPs have the potential to improve cancer immunotherapy.
Collapse
|
52
|
Xia C, Bai W, Deng T, Li T, Zhang L, Lu Z, Zhang Z, Li M, He Q. Sponge-like nano-system suppresses tumor recurrence and metastasis by restraining myeloid-derived suppressor cells-mediated immunosuppression and formation of pre-metastatic niche. Acta Biomater 2023; 158:708-724. [PMID: 36638937 DOI: 10.1016/j.actbio.2023.01.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 12/18/2022] [Accepted: 01/03/2023] [Indexed: 01/12/2023]
Abstract
Tumor recurrence and metastasis still greatly limit the therapeutic efficiency on the majority of postoperative clinical cases. With the aim to realize more powerful treatment outcomes on postoperative malignant tumors, a sponge-like neutrophil membrane-coated nano-system (NM/PPcDG/D) was fabricated to inhibit tumor recurrence and metastasis by inhibiting the recruitment and functions of myeloid-derived suppressor cell (MDSCs), which reinforced anti-tumor immunity and also suppressed pulmonary metastasis by inhibiting the formation of pre-metastatic niche (PMN). Firstly, PPcDG/D nanoparticles (NPs) were formulated by the self-assembling and crosslinking of synthesized redox-responsive polymer (PPDG) with doxorubicin (DOX) loading in the nanocore (PPcDG/D), followed by coating with activated neutrophils membrane to fabricate biomimetic NM/PPcDG/D. The sponge-like NM/PPcDG/D not only showed obvious natural tropism to postoperative inflammatory site, but also inhibited the recruitment and functions of MDSCs, thus relieved MDSCs-mediated immunosuppression. Additionally, NM/PPcDG/D also suppressed the formation of PMN to inhibit pulmonary metastasis by reducing the recruitment of MDSCs, decreasing the permeability of pulmonary vessels and inhibiting the implantation of circulating tumor cell (CTCs). Eventually, this fabricated NM/PPcDG/D NPs significantly inhibited tumor recurrence and metastasis on postoperative triple negative breast cancer (TNBC) model, presenting a promising therapeutic strategy on postoperative malignant tumors. STATEMENT OF SIGNIFICANCE: Myeloid-derived suppressor cells (MDSCs) play important roles in accelerating tumor recurrence and metastasis by promoting the establishment of immunosuppression in postoperative inflammatory regions and facilitating the formation of pulmonary pre-metastasis niche (PMN). In order to achieve enhanced suppression of recurrence and metastasis, a sponge-like NM/PPcDG/D nano-system was designed and fabricated. This nano-system is also the first attempt to integrate the regulation effects of a nano-sponge and anti-inflammatory agent to achieve enhanced multi-mode manipulation of MDSCs. Ultimately, NM/PPcDG/D powerfully restrained the recurrence and spontaneous metastasis on TNBC model. This article also revealed the particular roles of MDSCs involved in the regulation networks of postoperative recurrence and metastasis, immunosuppression and inflammation.
Collapse
Affiliation(s)
- Chunyu Xia
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu 610041, China
| | - Wenjing Bai
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu 610041, China
| | - Tao Deng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu 610041, China
| | - Ting Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu 610041, China
| | - Ling Zhang
- College of Polymer Science and Engineering, Sichuan University, China
| | - Zhengze Lu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu 610041, China
| | - Zhirong Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu 610041, China
| | - Man Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu 610041, China.
| | - Qin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Med-X Center for Materials, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
53
|
Huang D, Xu D, Chen W, Wu R, Wen Y, Liu A, Lin L, Lin X, Wang X. Fe-MnO 2 nanosheets loading dihydroartemisinin for ferroptosis and immunotherapy. Biomed Pharmacother 2023; 161:114431. [PMID: 36827713 DOI: 10.1016/j.biopha.2023.114431] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/16/2023] [Accepted: 02/20/2023] [Indexed: 02/24/2023] Open
Abstract
Ferroptosis has emerged as a therapeutic tactic to trigger cancer cell death driven by abnormal accumulation of reactive oxygen species (ROS). However, a single ferroptosis treatment modality is often limited. In this work, a combination therapy of ferroptosis and immunotherapy for cancer was proposed. Specifically, a versatile nanodrug was designed for the multiple treatment of hepatocellular carcinoma (HCC) by loading dihydroartemisinin (DHA) on Fe3+-doped MnO2 nanosheets (Fe-MnO2/DHA). Firstly, Fe-MnO2/DHA was degraded by glutathione (GSH) in the tumor microenvironment (TME) to release Fe2+, Mn2+ and DHA, leading to aberrant ROS accumulation due to Fenton/Fenton-like reaction. Secondly, breakage of endoperoxide bridge from DHA was caused by Fe2+ to further induce oxidative stress. Thirdly, the depleted GSH promoted the inactivation of glutathione peroxidase 4 (GPX4), resulting in lipid peroxide (LPO) accumulation. The resulting LPO and ROS could induce ferroptosis and apoptosis of liver cancer cells. Furthermore, Fe-MnO2/DHA mediated three-pronged stimulation of oxidative stress, resulting in high levels of targeted immunogenic cell death (ICD). It could enhance the infiltration of CD4+ T and CD8+ T cells, and promote macrophage polarization. DHA also acted as an immunomodulator to inhibit regulatory T cells (Tregs) for systemic antitumor. Overall, Fe-MnO2/DHA presents a multi-modal therapy for HCC driven by ferroptosis, apoptosis and immune activation, significantly advancing synergistic cancer treatment.
Collapse
Affiliation(s)
- Dandan Huang
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Dafen Xu
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Wenxin Chen
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Ruimei Wu
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Yujuan Wen
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China
| | - Ailin Liu
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China; Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Fujian Medical University, Fuzhou 350122, China
| | - Liqing Lin
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China; Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Fujian Medical University, Fuzhou 350122, China.
| | - Xinhua Lin
- Department of Pharmaceutical Analysis, School of Pharmacy, Fujian Medical University, Fuzhou 350122, China; Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Fujian Medical University, Fuzhou 350122, China.
| | - Xuewen Wang
- Department of Preventive Medicine, School of Public Health, Fujian Medical University, Fuzhou 350122, China.
| |
Collapse
|
54
|
Wu H, Wei G, Luo L, Li L, Gao Y, Tan X, Wang S, Chang H, Liu Y, Wei Y, Song J, Zhang Z, Huo J. Ginsenoside Rg3 nanoparticles with permeation enhancing based chitosan derivatives were encapsulated with doxorubicin by thermosensitive hydrogel and anti-cancer evaluation of peritumoral hydrogel injection combined with PD-L1 antibody. Biomater Res 2022; 26:77. [PMID: 36494759 PMCID: PMC9733157 DOI: 10.1186/s40824-022-00329-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Combination of chemotherapy and immune checkpoint inhibitor therapy has greatly improved the anticancer effect on multiple malignancies. However, the efficiency on triple-negative breast cancer (TNBC) is limited, since most patients bear "cold" tumors with low tumor immunogenicity. Doxorubicin (DOX), one of the most effective chemotherapy agents, can induce immunogenic cell death (ICD) and thus initiating immune response. METHODS In this study, to maximize the ICD effect induced by DOX, chitosan and cell-penetrating peptide (R6F3)-modified nanoparticles (PNPs) loaded with ginsenoside Rg3 (Rg3) were fabricated using the self-assembly technique, followed by co-encapsulation with DOX based on thermo-sensitive hydrogel. Orthotopic tumor model and contralateral tumor model were established to observe the antitumor efficacy of the thermo-sensitive hydrogel combined with anti-PD-L1 immunotherapy, besides, the biocompatibility was also evaluated by histopathological. RESULTS Rg3-PNPs strengthened the immunogenic cell death (ICD) effect induced by DOX. Moreover, the hydrogel co-loading Rg3-PNPs and DOX provoked stronger immune response in originally nonimmunogenic 4T1 tumors than DOX monotherapy. Following combination with PD-L1 blocking, substantial antitumor effect was achieved due to the recruitment of memory T cells and the decline of adaptive PD-L1 enrichment. CONCLUSION The hydrogel encapsulating DOX and highly permeable Rg3-PNPs provided an efficient strategy for remodeling immunosuppressive tumor microenvironment and converting immune "cold" 4T1 into "hot" tumors.
Collapse
Affiliation(s)
- Hao Wu
- grid.410745.30000 0004 1765 1045Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 210023 Nanjing, China ,Jiangsu Province Academy of Traditional Chinese Medicine, 210028 Nanjing, China ,grid.411671.40000 0004 1757 5070School of Material Science and Chemical Engineering, Chuzhou University, 239000 Chuzhou, China
| | - Guoli Wei
- grid.410745.30000 0004 1765 1045Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 210023 Nanjing, China ,Jiangsu Province Academy of Traditional Chinese Medicine, 210028 Nanjing, China ,Department of Oncology, Nanjing Lishui District Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Lixia Luo
- grid.410745.30000 0004 1765 1045Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 210023 Nanjing, China ,Jiangsu Province Academy of Traditional Chinese Medicine, 210028 Nanjing, China
| | - Lingchang Li
- grid.410745.30000 0004 1765 1045Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 210023 Nanjing, China ,Jiangsu Province Academy of Traditional Chinese Medicine, 210028 Nanjing, China
| | - Yibo Gao
- grid.410745.30000 0004 1765 1045Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 210023 Nanjing, China ,Jiangsu Province Academy of Traditional Chinese Medicine, 210028 Nanjing, China
| | - Xiaobin Tan
- grid.410745.30000 0004 1765 1045Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 210023 Nanjing, China ,Jiangsu Province Academy of Traditional Chinese Medicine, 210028 Nanjing, China
| | - Sen Wang
- grid.410745.30000 0004 1765 1045Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 210023 Nanjing, China ,Jiangsu Province Academy of Traditional Chinese Medicine, 210028 Nanjing, China
| | - Haoxiao Chang
- grid.24696.3f0000 0004 0369 153XDepartment of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yuxi Liu
- grid.411671.40000 0004 1757 5070School of Material Science and Chemical Engineering, Chuzhou University, 239000 Chuzhou, China
| | - Yingjie Wei
- grid.410745.30000 0004 1765 1045Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 210023 Nanjing, China ,Jiangsu Province Academy of Traditional Chinese Medicine, 210028 Nanjing, China
| | - Jie Song
- grid.410745.30000 0004 1765 1045Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 210023 Nanjing, China ,Jiangsu Province Academy of Traditional Chinese Medicine, 210028 Nanjing, China
| | - Zhenhai Zhang
- grid.410745.30000 0004 1765 1045Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 210023 Nanjing, China ,Jiangsu Province Academy of Traditional Chinese Medicine, 210028 Nanjing, China
| | - Jiege Huo
- grid.410745.30000 0004 1765 1045Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 210023 Nanjing, China ,Jiangsu Province Academy of Traditional Chinese Medicine, 210028 Nanjing, China
| |
Collapse
|
55
|
Su L, Hao Y, Li R, Pan W, Ma X, Weng J, Min Y. Red blood cell-based vaccines for ameliorating cancer chemoimmunotherapy. Acta Biomater 2022; 154:401-411. [PMID: 36241013 DOI: 10.1016/j.actbio.2022.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 09/23/2022] [Accepted: 10/03/2022] [Indexed: 12/14/2022]
Abstract
Immune checkpoint blockade (ICB) therapy has shown promising antitumor effects, but its immune response rate remains unsatisfactory. In recent years, chemotherapy has been proven to have synergistic effects with ICB therapy because some chemotherapeutic agents can enhance the immunogenicity of tumor cells by inducing immunogenic cell death (ICD). However, it cannot be ignored that chemotherapy often shows limited therapeutic efficacy due to high cytotoxicity, drug resistance, and some other side effects. Herein, we report a strategy to improve cancer immunotherapy by utilizing red blood cell-based vaccines (RBC-vaccines) where chemotherapy-induced tumor antigens (cAgs) are anchored onto red blood cells (RBCs) via the EDC/NHS-mediated amine coupling reaction. In this work, RBC-vaccines administered subcutaneously are primarily devoured by dendritic cells (DCs) and significantly improve the efficacy of αPD-1 (anti-programmed cell death 1) treatment by increasing the infiltration of intratumoral CD8+ and CD4+ T cells and elevating the intratumoral ratio of CD8+ T cells to regulatory T cells in the CT-26 colon cancer model. Finally, based on the rejection of tumor rechallenge in cured mice, the combination therapy of RBC-vaccines and αPD-1 can induce the expansion of memory T cells and thereby establish a long-term antitumor immune response. Taken together, the proposed RBC-vaccines have great potential to improve chemoimmunotherapy. STATEMENT OF SIGNIFICANCE: Immunotherapy, especially immune checkpoint blockade therapy, has made great contributions to the treatment of some advanced cancers. Unfortunately, the great majority of patients with cancer do not benefit from immunotherapy. To enhance the response rate of immunotherapy, we developed red blood cell-based vaccines (RBC-vaccines) against cancers where antigens were harvested from chemotherapy-treated cancer cells and then attached to erythrocytes via covalent surface modification. Such RBC-vaccines could provide a wide variety of tumor antigens and damage-associated molecular patterns without the use of any extra ingredients to trigger a stronger antitumor immune response. More importantly, the combination of RBC-vaccines with PD-1 blockade could significantly improve the efficacy of cancer immunotherapy and induce durable antitumor immunity.
Collapse
Affiliation(s)
- Lanhong Su
- Department of Chemistry, University of Science and Technology of China, Hefei, China
| | - Yuhao Hao
- Department of Chemistry, University of Science and Technology of China, Hefei, China
| | - Rui Li
- Department of Chemistry, University of Science and Technology of China, Hefei, China
| | - Wen Pan
- Department of Chemistry, University of Science and Technology of China, Hefei, China
| | - Xiaopeng Ma
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Jianping Weng
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yuanzeng Min
- Department of Chemistry, University of Science and Technology of China, Hefei, China; The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, China; CAS Key Lab of Soft Matter Chemistry, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
56
|
Yu H, Wu M, Chen S, Song M, Yue Y. Biomimetic nanoparticles for tumor immunotherapy. Front Bioeng Biotechnol 2022; 10:989881. [PMID: 36440446 PMCID: PMC9682960 DOI: 10.3389/fbioe.2022.989881] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 10/26/2022] [Indexed: 12/11/2023] Open
Abstract
Currently, tumor treatment research still focuses on the cancer cells themselves, but the fact that the immune system plays an important role in inhibiting tumor development cannot be ignored. The activation of the immune system depends on the difference between self and non-self. Unfortunately, cancer is characterized by genetic changes in the host cells that lead to uncontrolled cell proliferation and evade immune surveillance. Cancer immunotherapy aims to coordinate a patient's immune system to target, fight, and destroy cancer cells without destroying the normal cells. Nevertheless, antitumor immunity driven by the autoimmune system alone may be inadequate for treatment. The development of drug delivery systems (DDS) based on nanoparticles can not only promote immunotherapy but also improve the immunosuppressive tumor microenvironment (ITM), which provides promising strategies for cancer treatment. However, conventional nano drug delivery systems (NDDS) are subject to several limitations in clinical transformation, such as immunogenicity and the potential toxicity risks of the carrier materials, premature drug leakage at off-target sites during circulation and drug load content. In order to address these limitations, this paper reviews the trends and progress of biomimetic NDDS and discusses the applications of each biomimetic system in tumor immunotherapy. Furthermore, we review the various combination immunotherapies based on biomimetic NDDS and key considerations for clinical transformation.
Collapse
Affiliation(s)
- Hanqing Yu
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Meng Wu
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Siyu Chen
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Mingming Song
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yulin Yue
- Department of Clinical Laboratory, Children’s Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
57
|
Wei Q, Zhang L, Zhao N, Cheng Z, Xin H, Ding J. Immunosuppressive adenosine-targeted biomaterials for emerging cancer immunotherapy. Front Immunol 2022; 13:1012927. [PMID: 36389700 PMCID: PMC9641176 DOI: 10.3389/fimmu.2022.1012927] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 09/29/2022] [Indexed: 12/28/2024] Open
Abstract
Immunotherapy has paved the way for the future of cancer therapy, but there are still significant challenges to be overcome, such as the occurrence of immune escape or suppression. Adenosine is essential in modulating the immune responses of immune cells and maintaining immune tolerance. Emerging adenosine pathway inhibitors are considered a breakthrough in cancer immunotherapy, with emphasis first being placed on the top-down blockade of adenosine signaling axis, followed by combination therapy. However, these therapeutic strategies rely on adenosine inhibitors, mainly small molecules or antibody proteins, which are limited by a single route of administration and off-target toxicity. Therefore, synergistic nanomedicine with accurate delivery targeting deeper tumors is focused on in preclinical studies. This review discusses how adenosine reshapes immunosuppressive microenvironments through its effects on immune cells, including lymphocytes and myeloid cells. Additionally, it will be the first discussion of a comprehensive strategy of biomaterials in modulating the adenosine signaling pathway, including inhibition of adenosine production, inhibition of adenosine binding to immune cells, and depletion of adenosine in the microenvironments. Furthermore, biomaterials integrating multiple therapeutic modalities with adenosine blocking are also discussed as a promising strategy for promoting cancer immunotherapy.
Collapse
Affiliation(s)
- Qi Wei
- Department of Thoracic Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Lening Zhang
- Department of Thoracic Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Nan Zhao
- Department of Thoracic Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhihua Cheng
- Department of Vascular Surgery, General Surgery Center, the First Hospital of Jilin University, Changchun, China
| | - Hua Xin
- Department of Thoracic Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| |
Collapse
|
58
|
Chen Y, Luo X, Liu Y, Zou Y, Yang S, Liu C, Zhao Y. Targeted Nanobubbles of PD-L1 mAb Combined with Doxorubicin as a Synergistic Tumor Repressor in Hepatocarcinoma. Int J Nanomedicine 2022; 17:3989-4008. [PMID: 36105615 PMCID: PMC9464779 DOI: 10.2147/ijn.s376172] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/21/2022] [Indexed: 12/14/2022] Open
Abstract
Purpose Ultrasound nanobubbles (NBs) can kill tumor cells, mediated by their effects of cavitation and acoustic perforation through ultrasound, while as novel drug carriers, biomaterial-modified NBs release drugs at a target region. In this work, the ultrasound NBs bridged by biotin-streptavidin were prepared simultaneously to be loaded with both programmed death ligand 1 monoclonal antibody (PD-L1 mAb) and doxorubicin (DOX), which are immune checkpoint inhibitors (ICIs) and chemotherapeutic agents, to synergize immunotherapy and chemotherapy combined with sonodynamic therapy (SDT). Methods The PD-L1 mAb/DOX NBs, using bridging affinity biotin (BRAB) technology as a bridge, were prepared by thin-film hydration and mechanical oscillation for the targeted delivery of biotinylated PD-L1 mAb and DOX. Characterization and pharmacokinetic studies of PD-L1 mAb/DOX NBs were performed in vitro and in vivo. The antitumor effect of ultrasound-mediated PD-L1 mAb/DOX-NBs was studied in the subcutaneously transplanted tumor of the H22 hepatoma model, and the mechanism of synergistic tumor repression was investigated. Results The data of in vitro targeting experiments, contrast-enhanced ultrasound imaging (CEUS), in vivo imaging of the small animals imaging system (IVIS), and frozen sections showed that PD-L1 mAb/DOX-NBs have well-targeted aggregation in the tumor. By observing tumor inhibition rate, tissue cell apoptosis, and apoptosis-related gene and protein expression, the PD-L1 mAb/DOX-NBs group showed the best immunotherapy effects, and its tumor volume and mass inhibition rates were about 69.64% and 75.97%, respectively (P < 0.01). Therefore, blocking the PD-1/PD-L1 pathway could improve immune cells’ tumor-killing ability. Antitumor immune cytokines were further enhanced when combined with DOX-induced tumor cell apoptosis and immunogenic cell death (ICD). Conclusion In summary, ultrasound-mediated PD-L1 mAb/DOX-NBs showed significant synergistic antitumor effects, providing a potential combined immunotherapy strategy for HCC.
Collapse
Affiliation(s)
- Yezi Chen
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, People's Republic of China.,Medical College of China Three Gorges University, Yichang, People's Republic of China
| | - Xiaoqin Luo
- Medical College of China Three Gorges University, Yichang, People's Republic of China.,Department of Medical Imaging Center, Renmin Hospital Affiliated to Hubei University of Medicine, Shiyan, People's Republic of China
| | - Yun Liu
- Department of Ultrasonography, Yichang Central People's Hospital, Yichang, People's Republic of China
| | - Yunlei Zou
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, People's Republic of China.,Medical College of China Three Gorges University, Yichang, People's Republic of China
| | - Shiqi Yang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, People's Republic of China.,Medical College of China Three Gorges University, Yichang, People's Republic of China
| | - Chaoqi Liu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, People's Republic of China.,Medical College of China Three Gorges University, Yichang, People's Republic of China
| | - Yun Zhao
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, China Three Gorges University, Yichang, People's Republic of China.,Medical College of China Three Gorges University, Yichang, People's Republic of China
| |
Collapse
|
59
|
Liu Y, Xie J, Zhao X, Zhang Y, Zhong Z, Deng C. A polymeric IDO inhibitor based on poly(ethylene glycol)- b-poly(L-tyrosine- co-1-methyl-D-tryptophan) enables facile trident cancer immunotherapy. Biomater Sci 2022; 10:5731-5743. [PMID: 36039890 DOI: 10.1039/d2bm01181f] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
Indoleamine 2,3-dioxygenase (IDO), with an immunoregulatory effect related to tryptophan metabolism, has emerged as an attractive target for cancer immunotherapy. Here, a polymeric IDO inhibitor based on the poly(ethylene glycol)-b-poly(L-tyrosine-co-1-methyl-D-tryptophan) copolymer (PEG-b-P(Tyr-co-1-MT)) was developed for facile trident cancer immunotherapy. PEG-b-P(Tyr-co-1-MT) could self-assemble into nanoparticles (NPs), which were subject to enzyme degradation and capable of retarding the metabolism of L-tryptophan (TRP) to L-kynurenine (KYN) in B16F10 cancer cells. Notably, cRGD-functionalized NPs showed efficient encapsulation and an enzyme-responsive release of doxorubicin (DOX) and the BET bromodomain inhibitor JQ1. DOX in drug-loaded nanoparticles (cRGD-NPDJ) could activate immunization by inducing the discernible immunogenic cell death (ICD) of cancer cells and promoting the secretion of interferon-γ (IFN-γ), which besides activating the antitumor cellular immunity often upregulates the expression of PD-L1 and IDO to accelerate tumor progression. The encapsulated JQ1 and polymeric 1-MT in cRGD-NPDJ could reverse the expression by disrupting the binding of BET proteins with chromatin and elevating the TRP/KYN ratio. In B16F10 tumor-bearing C57BL/6 mice, cRGD-NPDJ displayed significantly increased CD8+ T cells, matured dendritic cells (mDCs), and cytokines (IFN-γ, TNF-α), as well as reduced regulatory T cells and downregulated PD-L1 expression at tumor sites, generating immune cascade reactions and a distinct improvement of the tumor microenvironment (TME), leading to significant tumor suppression and survival prolongation. The polymeric IDO inhibitor provides a facile strategy for the co-delivery of chemotherapeutics and inhibitors for efficient and safe combination cancer immunotherapy.
Collapse
Affiliation(s)
- Yuanyuan Liu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.
| | - Jiguo Xie
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.
| | - Xiaofei Zhao
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.
| | - Yueyue Zhang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.
| | - Chao Deng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.
| |
Collapse
|
60
|
Yu L, Jin Y, Song M, Zhao Y, Zhang H. When Natural Compounds Meet Nanotechnology: Nature-Inspired Nanomedicines for Cancer Immunotherapy. Pharmaceutics 2022; 14:pharmaceutics14081589. [PMID: 36015215 PMCID: PMC9412684 DOI: 10.3390/pharmaceutics14081589] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/25/2022] [Accepted: 07/28/2022] [Indexed: 02/01/2023] Open
Abstract
Recent significant strides of natural compounds in immunomodulation have highlighted their great potential against cancer. Despite many attempts being made for cancer immunotherapy, the biomedical application of natural compounds encounters a bottleneck because of their unclear mechanisms, low solubility and bioavailability, and limited efficacy. Herein, we summarize the immune regulatory mechanisms of different natural compounds at each step of the cancer-immunity cycle and highlight their anti-tumor potential and current limitations. We then propose and present various drug delivery strategies based on nanotechnology, including traditional nanoparticles (NPs)-based delivery strategies (lipid-based NPs, micelles, and polysaccharide/peptide/protein-based NPs) and novel delivery strategies (cell-derived NPs and carrier-free NPs), thus providing solutions to break through existing bottlenecks. Furthermore, representative applications of nature-inspired nanomedicines are also emphasized in detail with the advantages and disadvantages discussed. Finally, the challenges and prospects of natural compounds for cancer immunotherapy are provided, hopefully, to facilitate their far-reaching development toward clinical translation.
Collapse
Affiliation(s)
- Linna Yu
- People’s Hospital of Qianxinan Buyi and Miao Minority Autonomous Prefecture, Xingyi 562400, China;
| | - Yi Jin
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicines, Department of Pharmaceutics, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, China; (Y.J.); (M.S.)
| | - Mingjie Song
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicines, Department of Pharmaceutics, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, China; (Y.J.); (M.S.)
| | - Yu Zhao
- People’s Hospital of Qianxinan Buyi and Miao Minority Autonomous Prefecture, Xingyi 562400, China;
- Correspondence: (Y.Z.); (H.Z.)
| | - Huaqing Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance (Ministry of Education), State Key Laboratory of Natural Medicines, Department of Pharmaceutics, NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, Nanjing 210009, China; (Y.J.); (M.S.)
- Correspondence: (Y.Z.); (H.Z.)
| |
Collapse
|
61
|
Xu K, Khan M, Yu J, Snyder NW, Wu S, Vazquez-Padron RI, Wang H, Yang X. Editorial: Insights in cardiovascular therapeutics: 2021 - cell death, cardiovascular injuries, and novel targets of cardiovascular therapeutics. Front Cardiovasc Med 2022; 9:981544. [PMID: 35958425 PMCID: PMC9361401 DOI: 10.3389/fcvm.2022.981544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 07/11/2022] [Indexed: 11/15/2022] Open
Affiliation(s)
- Keman Xu
- Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Mohsin Khan
- Departments of Cardiovascular Sciences and Biomendical Education and Data Sciences, Centers for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Jun Yu
- Departments of Cardiovascular Sciences and Biomendical Education and Data Sciences, Centers for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Nathaniel W. Snyder
- Departments of Cardiovascular Sciences and Biomendical Education and Data Sciences, Centers for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Sheng Wu
- Departments of Cardiovascular Sciences and Biomendical Education and Data Sciences, Centers for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Roberto I. Vazquez-Padron
- DeWitt Daughtry Family Department of Surgery, Leonard M. Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Hong Wang
- Departments of Cardiovascular Sciences and Biomendical Education and Data Sciences, Centers for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Xiaofeng Yang
- Departments of Cardiovascular Sciences and Biomedical Education and Data Sciences, Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
- Departments of Cardiovascular Sciences and Biomendical Education and Data Sciences, Centers for Metabolic Disease Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
62
|
Selection of cryoprotectants for freezing and freeze-drying of gold nanoparticles towards further uses in various applications. Colloids Surf B Biointerfaces 2022; 217:112702. [PMID: 35863234 DOI: 10.1016/j.colsurfb.2022.112702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 11/22/2022]
Abstract
Recently, cryopreservation of AuNPs without aggregation has been attempted to improve their long-term stability. This study investigated criteria to select cryoprotectants for AuNPs using a variety of materials, including sugar (sucrose), surfactant (Tween 20), polymers (polyvinyl alcohol (PVA) and polyvinylpyrrolidone (PVP)), and biopolymer (pectin). For cryoprotective performance, UV-vis spectroscopy reveals the potential of all cryoprotectants for preventing citrate-capped AuNPs (cit-AuNPs) from irreversible aggregation under freezing. While sucrose, PVP, and pectin were more suitable than Tween 20 and PVA as cryoprotectants for lyophilization of AuNPs with the maintained redispersability. For storage and further use, Luria-Bertani agar plate, dynamic light scattering (DLS), and transmission electron microscopy (TEM) results indicate impacts of the cryoprotectant coexisted with AuNPs after resuspension and imply that washing of the restored AuNPs is encouraged. Otherwise, running the restored AuNPs through applications, such as functionalization, protein conjugation, and surface-enhanced Raman scattering (SERS), without washing the cryoprotectant could lead to inaccurate results. This study also serves as a guideline for a comprehensive practice flow of AuNP handling, encompassing the synthesis step, cryopreservation, and use after resuspension.
Collapse
|
63
|
Hwang J, An EK, Kim SJ, Zhang W, Jin JO. Escherichia coli Mimetic Gold Nanorod-Mediated Photo- and Immunotherapy for Treating Cancer and Its Metastasis. ACS NANO 2022; 16:8472-8483. [PMID: 35466668 DOI: 10.1021/acsnano.2c03379] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Most cancer-related deaths are due to metastasis or recurrence. Therefore, the ultimate goal of cancer therapy will be to treat metastatic and recurrent cancers. Combination therapy for cancer will be one of trial for effective treating metastasis and recurrence. In this study, Escherichia coli-mimetic nanomaterials are synthesized using Escherichia coli membrane proteins, adhesion proteins, and gold nanorods, which are named E. coli mimetic AuNRs (ECA), for combination therapy against cancer and its recurrence. ECA treatment with 808 nm laser irradiation eliminates CT-26 or 4T1 tumors via a photothermal effect. ECA with laser irradiation induces activation of immune cells in the tumor-draining lymph nodes. The mice cured from CT-26 or 4T1 tumor by ECA are rechallenged with those cancer in the lung metastatic form, and the results showed that ECA treatment for the first CT-26 or 4T1 tumor challenge prevents cancer infiltration to the lung in the second challenge. This preventive effect of ECA against tumor growth in the second challenge is aided by cancer antigen-specific T cell immunity. Overall, these findings show that ECA is a nanomaterial with dual functions as a photothermal therapy for treating primary cancers and as immunotherapy for preventing recurrence and metastasis.
Collapse
Affiliation(s)
- Juyoung Hwang
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 201508, China
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea
| | - Eun-Koung An
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea
| | - So-Jung Kim
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea
| | - Wei Zhang
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 201508, China
| | - Jun-O Jin
- Shanghai Public Health Clinical Center & Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, 201508, China
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea
| |
Collapse
|
64
|
Tarannum M, Vivero-Escoto JL. Nanoparticle-based therapeutic strategies targeting major clinical challenges in pancreatic cancer treatment. Adv Drug Deliv Rev 2022; 187:114357. [PMID: 35605679 DOI: 10.1016/j.addr.2022.114357] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 04/11/2022] [Accepted: 05/17/2022] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers due to its aggressiveness and the challenges for early diagnosis and treatment. Recently, nanotechnology has demonstrated relevant strategies to overcome some of the major clinical issues in the treatment of PDAC. This review is focused on the pathological hallmarks of PDAC and the impact of nanotechnology to find solutions. It describes the use of nanoparticle-based systems designed for the delivery of chemotherapeutic agents and combinatorial alternatives that address the chemoresistance associated with PDAC, the development of combination therapies targeting the molecular heterogeneity in PDAC, the investigation of novel therapies dealing with the improvement of immunotherapy and handling the desmoplastic stroma in PDAC by remodeling the tumor microenvironment. A special section is dedicated to the design of nanoparticles for unique non-traditional modalities that could be promising in the future for the improvement in the dismal prognosis of PDAC.
Collapse
|
65
|
Hu D, Zhang W, Xiang J, Li D, Chen Y, Yuan P, Shao S, Zhou Z, Shen Y, Tang J. A ROS-responsive synergistic delivery system for combined immunotherapy and chemotherapy. Mater Today Bio 2022; 14:100284. [PMID: 35647515 PMCID: PMC9130108 DOI: 10.1016/j.mtbio.2022.100284] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Revised: 05/05/2022] [Accepted: 05/08/2022] [Indexed: 01/12/2023]
Abstract
Immune checkpoint blockade (ICB) therapies that target programmed cell death-1 (PD-1)/programmed cell death-ligand 1 (PD-L1) pathway are currently used for the treatment of various cancer types. However, low response rates of ICB remain the major issue and limit their applications in clinic. Here, we developed a ROS-responsive synergistic delivery system (pep-PAPM@PTX) by integrating physically-encapsulated paclitaxel (PTX) and surface-modified anti-PD-L1 peptide (pep) for combined chemotherapy and ICB therapy. Pep-PAPM@PTX could bind the cell surface PD-L1 and drive its recycling to lysosomal degradation, thus reverting PTX-induced PD-L1 upregulation and downregulating PD-L1 expression. As a result, pep-PAPM@PTX significantly promoted T cell infiltration and increased tumor immunoactivating factors, synergizing PTX chemotherapy to achieve enhanced anticancer potency in a triple-negative breast cancer (TNBC) model.
Collapse
|
66
|
Zhao Y, Liu X, Liu X, Yu J, Bai X, Wu X, Guo X, Liu Z, Liu X. Combination of phototherapy with immune checkpoint blockade: Theory and practice in cancer. Front Immunol 2022; 13:955920. [PMID: 36119019 PMCID: PMC9478587 DOI: 10.3389/fimmu.2022.955920] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 08/19/2022] [Indexed: 12/21/2022] Open
Abstract
Immune checkpoint blockade (ICB) therapy has evolved as a revolutionized therapeutic modality to eradicate tumor cells by releasing the brake of the antitumor immune response. However, only a subset of patients could benefit from ICB treatment currently. Phototherapy usually includes photothermal therapy (PTT) and photodynamic therapy (PDT). PTT exerts a local therapeutic effect by using photothermal agents to generate heat upon laser irradiation. PDT utilizes irradiated photosensitizers with a laser to produce reactive oxygen species to kill the target cells. Both PTT and PDT can induce immunogenic cell death in tumors to activate antigen-presenting cells and promote T cell infiltration. Therefore, combining ICB treatment with PTT/PDT can enhance the antitumor immune response and prevent tumor metastases and recurrence. In this review, we summarized the mechanism of phototherapy in cancer immunotherapy and discussed the recent advances in the development of phototherapy combined with ICB therapy to treat malignant tumors. Moreover, we also outlined the significant progress of phototherapy combined with targeted therapy or chemotherapy to improve ICB in preclinical and clinical studies. Finally, we analyzed the current challenges of this novel combination treatment regimen. We believe that the next-generation technology breakthrough in cancer treatment may come from this combinational win-win strategy of photoimmunotherapy.
Collapse
Affiliation(s)
- Yujie Zhao
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xu Liu
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,Department of Head, Neck and Mammary Gland Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xinyu Liu
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Yu
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Bai
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xi Wu
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xinyu Guo
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zhihui Liu
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaowei Liu
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
67
|
Xia Y, Wang T, Wu L, Zhong H, Zhou Y, Ke P, Bao X, Dai Q, Yang Q, Lu Y, Xu D, Liang M, Han M. Blocking CD47 with restructured peptide nanoparticles for motivating phagocytosis to inhibit tumor progression. Biomater Sci 2022; 10:4324-4334. [PMID: 35775458 DOI: 10.1039/d2bm00189f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Phagocytosis checkpoints, especially targeting CD47, have shown encouraging therapeutic effects. However, there are currently many shortcomings and challenges with immune checkpoint blockades (ICBs). Inspired by the phenomenon of molecular self-assembly,...
Collapse
Affiliation(s)
- Yiyi Xia
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Tiantian Wang
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.
| | - Linjie Wu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Haiqing Zhong
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Yi Zhou
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Peng Ke
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Xiaoyan Bao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Qi Dai
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Qiyao Yang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yiying Lu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Donghang Xu
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.
| | - Min Liang
- Department of Anesthesiology, Anesthesiology Research Institute, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, China.
| | - Min Han
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|