1
|
Das R, Ge X, Fei F, Parvanian S, Weissleder R, Garris CS. Lipid Nanoparticle-mRNA Engineered Dendritic Cell Based Adoptive Cell Therapy Enhances Cancer Immune Response. SMALL METHODS 2025; 9:e2400633. [PMID: 39039995 PMCID: PMC11740962 DOI: 10.1002/smtd.202400633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/27/2024] [Indexed: 07/24/2024]
Abstract
Lipid nanoparticles encapsulating mRNA (LNP-mRNA) revolutionized medicine over the past several years. While clinically approved indications currently focus on infectious disease vaccination, LNP-mRNA based treatments also hold promise for cancer immunotherapy. However, the route of dosing may impact treatment efficacy, safety, and dose. To minimize adverse effects, it is hypothesized that LNP-mRNA can be used to activate and engineer dendritic cells (DC) ex vivo before re-administration of these cells. Here, it is shown that LNP-mRNA engineered DCs can indeed vaccinate recipient mice. Vaccinated mice showed strong anti-tumor T cell responses, rejected tumor challenge, and displayed no evidence of toxicity. Further, it is found that DC specific ablation of the immune activating kinase NFkB inducing kinase (NIK) abrogated vaccination efficacy, demonstrating that adoptively transferred DCs can be functionally modified in addition to their antigen presentation capacity. Collectively, these studies show that ex vivo LNP-mRNA engineering of DCs is a feasible and robust therapeutic strategy for cancer.
Collapse
Affiliation(s)
- Riddha Das
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
| | - Xinying Ge
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
- Master's Program in ImmunologyHarvard Medical School200 Longwood AveBostonMA02115USA
| | - Fan Fei
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
- Master's Program in ImmunologyHarvard Medical School200 Longwood AveBostonMA02115USA
| | - Sepideh Parvanian
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
| | - Ralph Weissleder
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
- Department of Systems BiologyHarvard Medical School200 Longwood AveBostonMA02115USA
- Department of RadiologyMassachusetts General Brigham32 Fruit StBostonMA02114USA
| | - Christopher S. Garris
- Center for Systems BiologyMassachusetts General Hospital185 Cambridge St, CPZN 5206BostonMA02114USA
- Department of PathologyMassachusetts General Hospital55 Fruit StBostonMA02114USA
| |
Collapse
|
2
|
Long Z, Wu Y, Zhong L, Lu J, Liu B. Bibliometric analysis of dendritic cell-based vaccines over the past 15 years. Hum Vaccin Immunother 2024; 20:2392961. [PMID: 39161160 PMCID: PMC11340764 DOI: 10.1080/21645515.2024.2392961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/17/2024] [Accepted: 08/12/2024] [Indexed: 08/21/2024] Open
Abstract
Dendritic cells, which are crucial for inducing T-cell responses, are pivotal in current immunotherapy strategies aiming to replenish depleted T cells within the tumor microenvironment to combat tumors. Consequently, dendritic cell vaccine-based cancer therapies have garnered significant attention. Through bibliometric analysis, we examined research trends in this field. We searched the Web of Science core database and identified 16,476 articles on dendritic cell-based vaccines published from January 1, 2009, to December 30, 2023. The United States leads in this research domain, with Emory University being a prominent collaborator. The Journal of Immunology is the primary publication outlet, and Banchereau, J emerges as the most influential author. Recent hot keywords include nanoparticle, delivery, cancer vaccine, and clinical trial, indicating that cancer immunotherapy research, especially dendritic cell-based vaccines, is poised to become a future trend and hotspot.
Collapse
Affiliation(s)
- Zhi Long
- Department of Neurosurgery, Xiangya Hospital, Central South University, Jiangxi, China
- Hypothalamic-Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yinghua Wu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Jiangxi, China
- Hypothalamic-Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Liangchen Zhong
- Department of Neurosurgery, Xiangya Hospital, Central South University, Jiangxi, China
| | - Jiping Lu
- Orthopedics Department, 921 Hospital, Joint Logistics Support Force People’s Liberation Army of China, Changsha, China
| | - Bo Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Jiangxi, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
3
|
Qi K, Jia D, Zhou S, Zhang K, Guan F, Yao M, Sui X. Cryopreservation of Immune Cells: Recent Progress and Challenges Ahead. Adv Biol (Weinh) 2024; 8:e2400201. [PMID: 39113431 DOI: 10.1002/adbi.202400201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/02/2024] [Indexed: 12/14/2024]
Abstract
Cryopreservation of immune cells is considered as a key enabling technology for adoptive cellular immunotherapy. However, current immune cell cryopreservation technologies face the challenges with poor biocompatibility of cryoprotection materials, low efficiency, and impaired post-thaw function, limiting their clinical translation. This review briefly introduces the adoptive cellular immunotherapy and the approved immune cell-based products, which involve T cells, natural killer cells and etc. The cryodamage mechanisms to these immune cells during cryopreservation process are described, including ice formation related mechanical and osmotic injuries, cryoprotectant induced toxic injuries, and other biochemical injuries. Meanwhile, the recent advances in the cryopreservation medium and freeze-thaw protocol for several representative immune cell type are summarized. Furthermore, the remaining challenges regarding on the cryoprotection materials, freeze-thaw protocol, and post-thaw functionality evaluation of current cryopreservation technologies are discussed. Finally, the future perspectives are proposed toward advancing highly efficient cryopreservation of immune cells.
Collapse
Affiliation(s)
- Kejun Qi
- School of Life Science, Zhengzhou University, Zhengzhou, 450001, P. R. China
| | - Danqi Jia
- School of Life Science, Zhengzhou University, Zhengzhou, 450001, P. R. China
| | - Shengxi Zhou
- School of Life Science, Zhengzhou University, Zhengzhou, 450001, P. R. China
| | - Kun Zhang
- School of Life Science, Zhengzhou University, Zhengzhou, 450001, P. R. China
| | - Fangxia Guan
- School of Life Science, Zhengzhou University, Zhengzhou, 450001, P. R. China
| | - Minghao Yao
- School of Life Science, Zhengzhou University, Zhengzhou, 450001, P. R. China
| | - Xiaojie Sui
- School of Life Science, Zhengzhou University, Zhengzhou, 450001, P. R. China
| |
Collapse
|
4
|
Ren D, Xiong S, Ren Y, Yang X, Zhao X, Jin J, Xu M, Liang T, Guo L, Weng L. Advances in therapeutic cancer vaccines: Harnessing immune adjuvants for enhanced efficacy and future perspectives. Comput Struct Biotechnol J 2024; 23:1833-1843. [PMID: 38707540 PMCID: PMC11066472 DOI: 10.1016/j.csbj.2024.04.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/19/2024] [Accepted: 04/20/2024] [Indexed: 05/07/2024] Open
Abstract
Preventive cancer vaccines are highly effective in preventing viral infection-induced cancer, but advances in therapeutic cancer vaccines with a focus on eliminating cancer cells through immunotherapy are limited. To develop therapeutic cancer vaccines, the integration of optimal adjuvants is a potential strategy to enhance or complement existing therapeutic approaches. However, conventional adjuvants do not satisfy the criteria of clinical trials for therapeutic cancer vaccines. To improve the effects of adjuvants in therapeutic cancer vaccines, effective vaccination strategies must be formulated and novel adjuvants must be identified. This review offers an overview of the current advancements in therapeutic cancer vaccines and highlights in situ vaccination approaches that can be synergistically combined with other immunotherapies by harnessing the adjuvant effects. Additionally, the refinement of adjuvant systems using cutting-edge technologies and the elucidation of molecular mechanisms underlying immunogenic cell death to facilitate the development of innovative adjuvants have been discussed.
Collapse
Affiliation(s)
- Dekang Ren
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Shizheng Xiong
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Yujie Ren
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Xueni Yang
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Xinmiao Zhao
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Jiaming Jin
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Miaomiao Xu
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Tingming Liang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Li Guo
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Lixing Weng
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| |
Collapse
|
5
|
Chen L, Rao W, Chen Y, Xie J. In vitro induction of anti‑lung cancer immune response by the A549 lung cancer stem cell lysate‑sensitized dendritic cell vaccine. Oncol Lett 2024; 28:550. [PMID: 39328277 PMCID: PMC11425031 DOI: 10.3892/ol.2024.14683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 08/21/2024] [Indexed: 09/28/2024] Open
Abstract
Lung adenocarcinoma is one of the most fatal types of cancer worldwide, with non-small cell lung cancer being the most common subtype. Therefore, there is need for improved treatment approaches. Tumor growth results from the proliferation of a very small number of tumor stem cells, giving rise to the theory of cancer stem cells (CSCs). Lung CSCs are associated with lung cancer development, and although chemotherapy drugs can inhibit the proliferation of lung cancer cells, they have difficulty acting on lung CSCs. Even if the tumor appears to have disappeared after chemotherapy, the presence of a small number of residual tumor stem cells can lead to cancer recurrence and metastasis. Hence, targeting and eliminating lung CSCs is of significant therapeutic importance. In this study, we cultured A549 cells in sphere-forming conditions using B27, EGF, and bFGF, isolated peripheral blood mononuclear cells (PBMCs), and induced and characterized dendritic cells (DCs). We also isolated and expanded T lymphocytes. DC vaccines were prepared using A549 stem cell lysate or A549 cell lysate for sensitization and compared with non-sensitized DC vaccines. The content of IFN-γ in the supernatant of cultures with vaccines and T cells was measured by ELISA. The cytotoxic effects of the vaccines on A549 cells and stem cells were assessed using the Cytotox96 assay, and the impact of the vaccines on A549 cell migration and apoptosis was evaluated using Transwell assays and flow cytometry. DC vaccines sensitized with human lung CSC lysates induced significant in vitro cytotoxic effects on A549 lung cancer cells and CSCs by T lymphocytes, while not producing immune cytotoxic effects on human airway epithelial cells. Moreover, the immune-killing effect induced by DC vaccines sensitized with lung CSC lysates was superior to that of DC vaccines sensitized with lung cancer cells.
Collapse
Affiliation(s)
- Letian Chen
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Wei Rao
- Department of Urology, Yingtan People's Hospital, Yingtan, Jiangxi 335000, P.R. China
| | - Yujuan Chen
- Department of Pulmonary and Critical Care Medicine, Gaoan People's Hospital, Yichun, Jiangxi 336000, P.R. China
| | - Junping Xie
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| |
Collapse
|
6
|
Somri-Gannam L, Meisel-Sharon S, Hantisteanu S, Bar-Noy T, Sigal E, Groisman G, Hallak M, Werner H, Bruchim I. IGF1R inhibition and PD-1 blockade improve anti-tumor immune response in epithelial ovarian cancer. Front Oncol 2024; 14:1410447. [PMID: 39450263 PMCID: PMC11499063 DOI: 10.3389/fonc.2024.1410447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 09/10/2024] [Indexed: 10/26/2024] Open
Abstract
Introduction The insulin-like growth factor (IGF) system plays a key role in regulating growth and invasiveness in epithelial ovarian cancer (EOC) and is considered a promising therapeutic target. EOC is an immunosuppressive disease, although there are limited data about the involvement of the IGF1R system in the anti-tumor immune response in the EOC microenvironment. Methods In the current study, we hypothesized that IGF 1 receptor (IGF1R) involvement in the maturation of dendritic cells (DC) with the co-inhibition of IGF1R and PD-1 would affect the EOC microenvironment. Results We found that DC pretreated with IGF1R inhibitor resulted in fewer EOC cells. Moreover, in vivo experiments conducted with an EOC mouse model, with anti-PD-1/IGF1R combined, resulted in lower tumor weight compared to individual treatments. Additionally, anti-PD-1/IGF1R treatment increased DC by 34% compared with AEW-541 and 40% with anti-PD-1. The combined treatment increased CD8+ T-cell levels compared to AEW-541 alone. RNA-seq data analysis indicated that anti-PD-1/IGF1R led to a more potent immune response, as reflected by altered gene expression levels related to anti-tumor immune response, compared with either treatment alone. Discussion These findings provide novel evidence that IGF1R axis inhibition combined with PD-1 blockade may be an effective therapeutic strategy for selected EOC patient populations.
Collapse
Affiliation(s)
- Lina Somri-Gannam
- Gynecology Oncology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Hadera, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Shilhav Meisel-Sharon
- Gynecology Oncology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Hadera, Israel
| | - Shay Hantisteanu
- Gynecology Oncology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Hadera, Israel
| | - Tomer Bar-Noy
- Gynecology and Gynecologic Oncology Department, Hillel Yaffe Medical Center, Hadera, Israel
| | - Emiliya Sigal
- Gynecology and Gynecologic Oncology Department, Hillel Yaffe Medical Center, Hadera, Israel
| | - Gabriel Groisman
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
- Institute of Pathology, Hillel Yaffe Medical Center, Hadera, Israel
| | - Mordechai Hallak
- Gynecology Oncology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Hadera, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
- Gynecology and Gynecologic Oncology Department, Hillel Yaffe Medical Center, Hadera, Israel
| | - Haim Werner
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ilan Bruchim
- Gynecology Oncology Laboratory, Department of Obstetrics and Gynecology, Hillel Yaffe Medical Center, Hadera, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
- Gynecology and Gynecologic Oncology Department, Hillel Yaffe Medical Center, Hadera, Israel
| |
Collapse
|
7
|
Sun C, Li S, Ding J. Biomaterials-Boosted Immunotherapy for Osteosarcoma. Adv Healthc Mater 2024; 13:e2400864. [PMID: 38771618 DOI: 10.1002/adhm.202400864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/05/2024] [Indexed: 05/22/2024]
Abstract
Osteosarcoma (OS) is a primary malignant bone tumor that emanates from mesenchymal cells, commonly found in the epiphyseal end of long bones. The highly recurrent and metastatic nature of OS poses significant challenges to the efficacy of treatment and negatively affects patient prognosis. Currently, available clinical treatment strategies primarily focus on maximizing tumor resection and reducing localized symptoms rather than the complete eradication of malignant tumor cells to achieve ideal outcomes. The biomaterials-boosted immunotherapy for OS is characterized by high effectiveness and a favorable safety profile. This therapeutic approach manipulates the tumor microenvironments at the cellular and molecular levels to impede tumor progression. This review delves into the mechanisms underlying the treatment of OS, emphasizing biomaterials-enhanced tumor immunity. Moreover, it summarizes the immune cell phenotype and tumor microenvironment regulation, along with the ability of immune checkpoint blockade to activate the autoimmune system. Gaining a profound comprehension of biomaterials-boosted OS immunotherapy is imperative to explore more efficacious immunotherapy protocols and treatment options in this setting.
Collapse
Affiliation(s)
- Chao Sun
- Department of Orthopedic Surgery, Orthopedic Center, The First Hospital of Jilin University, 1 Xinmin Street, Changchun, 130061, P. R. China
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
| | - Shuqiang Li
- Department of Orthopedic Surgery, Orthopedic Center, The First Hospital of Jilin University, 1 Xinmin Street, Changchun, 130061, P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun, 130022, P. R. China
| |
Collapse
|
8
|
Zhao G, Wang Y, Xing S, Jiang Y, Ding J, Cai Y, Ma P, Miao H, Fang Y, Jiang N, Cui D, Yu Y, Tang Q, Wang S, Li N. Exosome-based anticancer vaccines: From Bench to bedside. Cancer Lett 2024; 595:216989. [PMID: 38825162 DOI: 10.1016/j.canlet.2024.216989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/13/2024] [Accepted: 05/23/2024] [Indexed: 06/04/2024]
Abstract
Exosomes, a subset of extracellular vesicles, are released by all active cells and play a crucial role in intercellular communications. Exosomes could facilitate the transfer of various biologically active molecules, such as DNA, non-coding RNAs, and proteins, from donor to recipient cells, thereby participating in diverse biological and pathological processes. Besides, exosomes possess unique characteristics, including non-toxicity, low-immunogenicity, and stability within biological systems, rendering them highly advantageous for cancer drug development. Meanwhile, accumulating evidence suggests that exosomes originating from tumor cells and immune cells possess distinct composition profiles that play a direct role in anticancer immunotherapy. Of note, exosomes can transport their contents to specific cells, thereby exerting an impact on the phenotype and immune-regulatory functions of targeted cells. Therapeutic cancer vaccines, an emerging therapeutics of immunotherapy, could enhance antitumor immune responses by delivering a large number of tumor antigens, thereby augmenting the immune response against tumor cells. Therefore, the therapeutic rationale of cancer vaccines and exosome-based immunotherapy are almost similar to some extent, but some challenges have hindered their application in the clinical setting. Here, in this review, we first summarized the biogenesis, structure, compositions, and biological functions of exosomes. Then we described the roles of exosomes in cancer biology, particularly in tumor immunity. We also comprehensively reviewed current exosome-based anticancer vaccine development and we divided them into three types. Finally, we give some insights into clinical translation and clinical trial progress of exosome-based anticancer vaccines for future direction.
Collapse
Affiliation(s)
- Guo Zhao
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yuning Wang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shujun Xing
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yale Jiang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jiatong Ding
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yuanting Cai
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Peiwen Ma
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Huilei Miao
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yuan Fang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ning Jiang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Dandan Cui
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yue Yu
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Qiyu Tang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shuhang Wang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Ning Li
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
9
|
Zhang H, Heng X, Yang H, Rao Y, Yao L, Zhu Z, Chen G, Chen H. Metal-Free Atom Transfer Radical Polymerization to Prepare Recylable Micro-Adjuvants for Dendritic Cell Vaccine. Angew Chem Int Ed Engl 2024; 63:e202402853. [PMID: 38598262 DOI: 10.1002/anie.202402853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 04/11/2024]
Abstract
In the development of dendritic cell (DC) vaccines, the maturation of DCs is a critical stage. Adjuvants play a pivotal role in the maturation of DCs, with a major concern being to ensure both efficacy and safety. This study introduces an innovative approach that combines high efficacy with safety through the synthesis of micro-adjuvants grafted with copolymers of 2-(methacrylamido) glucopyranose (MAG) and methacryloxyethyl trimethyl ammonium chloride (DMC). The utilization of metal-free surface-initiated atom transfer radical polymerization enables the production of safe and recyclable adjuvants. These micrometer-sized adjuvants surpass the optimal size range for cellular endocytosis, enabling the retrieval and reuse of them during the ex vivo maturation process, mitigating potential toxicity concerns associated with the endocytosis of non-metabolized nanoparticles. Additionally, the adjuvants exhibit a "micro-ligand-mediated maturation enhancement" effect for DC maturation. This effect is influenced by the shape of the particle, as evidenced by the distinct promotion effects of rod-like and spherical micro-adjuvants with comparable sizes. Furthermore, the porous structure of the adjuvants enables them to function as cargo-carrying "micro-shuttles", releasing antigens upon binding to DCs to facilitate efficient antigen delivery.
Collapse
Affiliation(s)
- Hengyuan Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Xingyu Heng
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, Jiangsu, China
| | - He Yang
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Yu Rao
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Lihua Yao
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Zhichen Zhu
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Gaojian Chen
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, Jiangsu, China
| | - Hong Chen
- College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, Jiangsu, China
| |
Collapse
|
10
|
Ingels J, De Cock L, Stevens D, Mayer RL, Théry F, Sanchez GS, Vermijlen D, Weening K, De Smet S, Lootens N, Brusseel M, Verstraete T, Buyle J, Van Houtte E, Devreker P, Heyns K, De Munter S, Van Lint S, Goetgeluk G, Bonte S, Billiet L, Pille M, Jansen H, Pascal E, Deseins L, Vantomme L, Verdonckt M, Roelandt R, Eekhout T, Vandamme N, Leclercq G, Taghon T, Kerre T, Vanommeslaeghe F, Dhondt A, Ferdinande L, Van Dorpe J, Desender L, De Ryck F, Vermassen F, Surmont V, Impens F, Menten B, Vermaelen K, Vandekerckhove B. Neoantigen-targeted dendritic cell vaccination in lung cancer patients induces long-lived T cells exhibiting the full differentiation spectrum. Cell Rep Med 2024; 5:101516. [PMID: 38626769 PMCID: PMC11148567 DOI: 10.1016/j.xcrm.2024.101516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 02/09/2024] [Accepted: 03/25/2024] [Indexed: 05/24/2024]
Abstract
Non-small cell lung cancer (NSCLC) is known for high relapse rates despite resection in early stages. Here, we present the results of a phase I clinical trial in which a dendritic cell (DC) vaccine targeting patient-individual neoantigens is evaluated in patients with resected NSCLC. Vaccine manufacturing is feasible in six of 10 enrolled patients. Toxicity is limited to grade 1-2 adverse events. Systemic T cell responses are observed in five out of six vaccinated patients, with T cell responses remaining detectable up to 19 months post vaccination. Single-cell analysis indicates that the responsive T cell population is polyclonal and exhibits the near-entire spectrum of T cell differentiation states, including a naive-like state, but excluding exhausted cell states. Three of six vaccinated patients experience disease recurrence during the follow-up period of 2 years. Collectively, these data support the feasibility, safety, and immunogenicity of this treatment in resected NSCLC.
Collapse
Affiliation(s)
- Joline Ingels
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium
| | - Laurenz De Cock
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Dieter Stevens
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; Respiratory Medicine, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Rupert L Mayer
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, East-Flanders, Belgium; VIB-UGent Center for Medical Biotechnology, VIB, 9000 Ghent, East-Flanders, Belgium
| | - Fabien Théry
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, East-Flanders, Belgium; VIB-UGent Center for Medical Biotechnology, VIB, 9000 Ghent, East-Flanders, Belgium
| | - Guillem Sanchez Sanchez
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles, 1050 Brussels, Brussels, Belgium; Institute for Medical Immunology, Université Libre de Bruxelles, 1050 Brussels, Brussels, Belgium; Université Libre de Bruxelles Center for Research in Immunology, Université Libre de Bruxelles, 1050 Brussels, Brussels, Belgium; WELBIO Department, WEL Research Institute, 1300 Wavre, Walloon Brabant, Belgium
| | - David Vermijlen
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles, 1050 Brussels, Brussels, Belgium; Institute for Medical Immunology, Université Libre de Bruxelles, 1050 Brussels, Brussels, Belgium; Université Libre de Bruxelles Center for Research in Immunology, Université Libre de Bruxelles, 1050 Brussels, Brussels, Belgium; WELBIO Department, WEL Research Institute, 1300 Wavre, Walloon Brabant, Belgium
| | - Karin Weening
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Saskia De Smet
- GMP Unit Cell Therapy, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Nele Lootens
- GMP Unit Cell Therapy, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Marieke Brusseel
- GMP Unit Cell Therapy, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Tasja Verstraete
- Respiratory Medicine, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Jolien Buyle
- Respiratory Medicine, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Eva Van Houtte
- GMP Unit Cell Therapy, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Pam Devreker
- GMP Unit Cell Therapy, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Kelly Heyns
- GMP Unit Cell Therapy, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Stijn De Munter
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium
| | - Sandra Van Lint
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; Respiratory Medicine, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Glenn Goetgeluk
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Sarah Bonte
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; VIB-UGent Center for Medical Biotechnology, VIB, 9000 Ghent, East-Flanders, Belgium
| | - Lore Billiet
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium
| | - Melissa Pille
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Hanne Jansen
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Eva Pascal
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium
| | - Lucas Deseins
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium
| | - Lies Vantomme
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Maarten Verdonckt
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Ria Roelandt
- VIB Single Cell Core, VIB, 9000/3000 Ghent/Leuven, East-Flanders/Flemish Brabant, Belgium
| | - Thomas Eekhout
- VIB Single Cell Core, VIB, 9000/3000 Ghent/Leuven, East-Flanders/Flemish Brabant, Belgium
| | - Niels Vandamme
- VIB Single Cell Core, VIB, 9000/3000 Ghent/Leuven, East-Flanders/Flemish Brabant, Belgium
| | - Georges Leclercq
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Tom Taghon
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Tessa Kerre
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; VIB-UGent Center for Medical Biotechnology, VIB, 9000 Ghent, East-Flanders, Belgium; Hematology, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Floris Vanommeslaeghe
- Nephrology, Ghent University Hospital, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Annemieke Dhondt
- Nephrology, Ghent University Hospital, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Liesbeth Ferdinande
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; Pathology, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Jo Van Dorpe
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; Pathology, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Liesbeth Desender
- Thoracic and Vascular Surgery, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Frederic De Ryck
- Thoracic and Vascular Surgery, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Frank Vermassen
- Thoracic and Vascular Surgery, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Veerle Surmont
- Respiratory Medicine, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Francis Impens
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, East-Flanders, Belgium; VIB-UGent Center for Medical Biotechnology, VIB, 9000 Ghent, East-Flanders, Belgium
| | - Björn Menten
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Karim Vermaelen
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; Respiratory Medicine, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium.
| | - Bart Vandekerckhove
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; GMP Unit Cell Therapy, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium.
| |
Collapse
|
11
|
Katopodi T, Petanidis S, Grigoriadou E, Anestakis D, Charalampidis C, Chatziprodromidou I, Floros G, Eskitzis P, Zarogoulidis P, Koulouris C, Sevva C, Papadopoulos K, Roulia P, Mantalovas S, Dagher M, Karakousis AV, Varsamis N, Vlassopoulos K, Theodorou V, Mystakidou CM, Katsios NI, Farmakis K, Kosmidis C. Immune Specific and Tumor-Dependent mRNA Vaccines for Cancer Immunotherapy: Reprogramming Clinical Translation into Tumor Editing Therapy. Pharmaceutics 2024; 16:455. [PMID: 38675116 PMCID: PMC11053579 DOI: 10.3390/pharmaceutics16040455] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/28/2024] Open
Abstract
Extensive research into mRNA vaccines for cancer therapy in preclinical and clinical trials has prepared the ground for the quick development of immune-specific mRNA vaccines during the COVID-19 pandemic. Therapeutic cancer vaccines based on mRNA are well tolerated, and are an attractive choice for future cancer immunotherapy. Ideal personalized tumor-dependent mRNA vaccines could stimulate both humoral and cellular immunity by overcoming cancer-induced immune suppression and tumor relapse. The stability, structure, and distribution strategies of mRNA-based vaccines have been improved by technological innovations, and patients with diverse tumor types are now being enrolled in numerous clinical trials investigating mRNA vaccine therapy. Despite the fact that therapeutic mRNA-based cancer vaccines have not yet received clinical approval, early clinical trials with mRNA vaccines as monotherapy and in conjunction with checkpoint inhibitors have shown promising results. In this review, we analyze the most recent clinical developments in mRNA-based cancer vaccines and discuss the optimal platforms for the creation of mRNA vaccines. We also discuss the development of the cancer vaccines' clinical research, paying particular attention to their clinical use and therapeutic efficacy, which could facilitate the design of mRNA-based vaccines in the near future.
Collapse
Affiliation(s)
- Theodora Katopodi
- Laboratory of Medical Biology and Genetics, Department of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (T.K.); (E.G.)
| | - Savvas Petanidis
- Laboratory of Medical Biology and Genetics, Department of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (T.K.); (E.G.)
- Department of Pulmonology, I.M. Sechenov First Moscow State Medical University, Moscow 119992, Russia
| | - Eirini Grigoriadou
- Laboratory of Medical Biology and Genetics, Department of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (T.K.); (E.G.)
| | - Doxakis Anestakis
- Department of Anatomy, Medical School, University of Cyprus, Nicosia 1678, Cyprus; (D.A.); (C.C.)
| | | | | | - George Floros
- Department of Electrical and Computer Engineering, University of Thessaly, 38334 Volos, Greece;
| | - Panagiotis Eskitzis
- Department of Obstetrics, University of Western Macedonia, 50100 Kozani, Greece;
| | - Paul Zarogoulidis
- Third Department of Surgery, “AHEPA” University Hospital, Aristotle University of Thessaloniki, 55236 Thessaloniki, Greece; (P.Z.); (C.K.); (C.S.); (K.P.); (S.M.); (M.D.); (A.V.K.); (C.K.)
| | - Charilaos Koulouris
- Third Department of Surgery, “AHEPA” University Hospital, Aristotle University of Thessaloniki, 55236 Thessaloniki, Greece; (P.Z.); (C.K.); (C.S.); (K.P.); (S.M.); (M.D.); (A.V.K.); (C.K.)
| | - Christina Sevva
- Third Department of Surgery, “AHEPA” University Hospital, Aristotle University of Thessaloniki, 55236 Thessaloniki, Greece; (P.Z.); (C.K.); (C.S.); (K.P.); (S.M.); (M.D.); (A.V.K.); (C.K.)
| | - Konstantinos Papadopoulos
- Third Department of Surgery, “AHEPA” University Hospital, Aristotle University of Thessaloniki, 55236 Thessaloniki, Greece; (P.Z.); (C.K.); (C.S.); (K.P.); (S.M.); (M.D.); (A.V.K.); (C.K.)
| | - Panagiota Roulia
- Third Department of Surgery, “AHEPA” University Hospital, Aristotle University of Thessaloniki, 55236 Thessaloniki, Greece; (P.Z.); (C.K.); (C.S.); (K.P.); (S.M.); (M.D.); (A.V.K.); (C.K.)
| | - Stylianos Mantalovas
- Third Department of Surgery, “AHEPA” University Hospital, Aristotle University of Thessaloniki, 55236 Thessaloniki, Greece; (P.Z.); (C.K.); (C.S.); (K.P.); (S.M.); (M.D.); (A.V.K.); (C.K.)
| | - Marios Dagher
- Third Department of Surgery, “AHEPA” University Hospital, Aristotle University of Thessaloniki, 55236 Thessaloniki, Greece; (P.Z.); (C.K.); (C.S.); (K.P.); (S.M.); (M.D.); (A.V.K.); (C.K.)
| | - Alexandros Vasileios Karakousis
- Third Department of Surgery, “AHEPA” University Hospital, Aristotle University of Thessaloniki, 55236 Thessaloniki, Greece; (P.Z.); (C.K.); (C.S.); (K.P.); (S.M.); (M.D.); (A.V.K.); (C.K.)
| | | | - Konstantinos Vlassopoulos
- Department of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.V.); (V.T.); (C.M.M.)
| | - Vasiliki Theodorou
- Department of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.V.); (V.T.); (C.M.M.)
| | - Chrysi Maria Mystakidou
- Department of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (K.V.); (V.T.); (C.M.M.)
| | - Nikolaos Iason Katsios
- Medical School, Faculty of Health Sciences, University of Ioannina, 45110 Ioannina, Greece;
| | - Konstantinos Farmakis
- Pediatric Surgery Clinic, General Hospital of Thessaloniki “G. Gennimatas”, Aristotle University of Thessaloniki, 54635 Thessaloniki, Greece;
| | - Christoforos Kosmidis
- Third Department of Surgery, “AHEPA” University Hospital, Aristotle University of Thessaloniki, 55236 Thessaloniki, Greece; (P.Z.); (C.K.); (C.S.); (K.P.); (S.M.); (M.D.); (A.V.K.); (C.K.)
| |
Collapse
|
12
|
Chen L, Zhang D, Chen Y, Zhu H, Liu Z, Yu Z, Xie J. ORC6 acts as an effective prognostic predictor for non‑small cell lung cancer and is closely associated with tumor progression. Oncol Lett 2024; 27:96. [PMID: 38288041 PMCID: PMC10823314 DOI: 10.3892/ol.2024.14229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 12/07/2023] [Indexed: 01/31/2024] Open
Abstract
Origin recognition complexes (ORCs) are vital in the control of DNA replication and the progression of the cell cycle, however the precise function and mechanism of ORC6 in non-small cell lung cancer (NSCLC) is still not well understood. The present study used bioinformatics methods to assess the predictive significance of ORC6 expression in NSCLC. Moreover, the expression of ORC6 was further evaluated using reverse transcription-quantitative PCR and western blotting, and its functional significance in lung cancer was assessed via knockdown experiments using small interfering RNA. A significant association was demonstrated between the expression of ORC6 and the clinical features of NSCLC. In particular, elevated levels of ORC6 were significantly strongly correlated with an unfavorable prognosis. Multivariate analysis demonstrated that increased ORC6 expression independently contributed to the risk of overall survival (HR 1.304; P=0.015) in individuals diagnosed with NSCLC. Analysis of Kaplan-Meier plots demonstrated that ORC6 expression served as a valuable indicator for diagnosing and predicting the prognosis of NSCLC. Moreover, in vitro studies demonstrated that modified ORC6 expression had a significant impact on the proliferation, migration and metastasis of NSCLC cells. NSCLC cell lines (H1299 and mH1650) exhibited markedly higher ORC6 expression than normal lung cell lines. The results of the present study indicated a strong association between the expression of ORC6 and the clinicopathological characteristics of NSCLC, which suggested its potential as a reliable biomarker for predicting NSCLC. Furthermore, ORC6 may have important therapeutic implications in the management of NSCLC.
Collapse
Affiliation(s)
- Letian Chen
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Dongdong Zhang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Yujuan Chen
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Huilan Zhu
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Zhipeng Liu
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Zhiping Yu
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| | - Junping Xie
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330000, P.R. China
| |
Collapse
|
13
|
Tai Y, Chen M, Wang F, Fan Y, Zhang J, Cai B, Yan L, Luo Y, Li Y. The role of dendritic cells in cancer immunity and therapeutic strategies. Int Immunopharmacol 2024; 128:111548. [PMID: 38244518 DOI: 10.1016/j.intimp.2024.111548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/03/2024] [Accepted: 01/12/2024] [Indexed: 01/22/2024]
Abstract
Dendritic cells (DCs) are asserted as the most potent antigen-presenting cells (APCs) that orchestrate both innate and adaptive immunity, being extremely effective in the induction of robust anti-cancer T cell responses. Hence, the modulation of DCs function represents an attractive target for improving cancer immunotherapy efficacy. A better understanding of the immunobiology of DCs, the interaction among DCs, immune effector cells and tumor cells in tumor microenvironment (TME) and the latest advances in biomedical engineering technology would be required for the design of optimal DC-based immunotherapy. In this review, we focus on elaborating the immunobiology of DCs in healthy and cancer environments, the recent advances in the development of enhancing endogenous DCs immunocompetence via immunomodulators as well as DC-based vaccines. The rapidly developing field of applying nanotechnology to improve DC-based immunotherapy is also highlighted.
Collapse
Affiliation(s)
- Yunze Tai
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Man Chen
- Hebei Yanda Lu Daopei Hospital, Langfang 065201, China
| | - Fang Wang
- Department of Medical Laboratory, The Second Affiliated Hospital of Guizhou Medical University, Kaili, Guizhou 556000, China
| | - Yu Fan
- Department of Urology, National Clinical Research Center for Geriatrics and Organ Transplantation Center, West China Hospital, Sichuan University, No. 37 Guoxue Xiang, Chengdu 610041, China
| | - Junlong Zhang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bei Cai
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lin Yan
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yao Luo
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Yi Li
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
14
|
Qiu Q, Chen S, He H, Chen J, Ding X, Wang D, Yang J, Guo P, Li Y, Kim J, Sheng J, Gao C, Yin B, Zheng S, Wang J. An injectable signal-amplifying device elicits a specific immune response against malignant glioblastoma. Acta Pharm Sin B 2023; 13:5091-5106. [PMID: 38045037 PMCID: PMC10692361 DOI: 10.1016/j.apsb.2023.06.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 06/04/2023] [Accepted: 06/05/2023] [Indexed: 12/05/2023] Open
Abstract
Despite exciting achievements with some malignancies, immunotherapy for hypoimmunogenic cancers, especially glioblastoma (GBM), remains a formidable clinical challenge. Poor immunogenicity and deficient immune infiltrates are two major limitations to an effective cancer-specific immune response. Herein, we propose that an injectable signal-amplifying nanocomposite/hydrogel system consisting of granulocyte-macrophage colony-stimulating factor and imiquimod-loaded antigen-capturing nanoparticles can simultaneously amplify the chemotactic signal of antigen-presenting cells and the "danger" signal of GBM. We demonstrated the feasibility of this strategy in two scenarios of GBM. In the first scenario, we showed that this simultaneous amplification system, in conjunction with local chemotherapy, enhanced both the immunogenicity and immune infiltrates in a recurrent GBM model; thus, ultimately making a cold GBM hot and suppressing postoperative relapse. Encouraged by excellent efficacy, we further exploited this signal-amplifying system to improve the efficiency of vaccine lysate in the treatment of refractory multiple GBM, a disease with limited clinical treatment options. In general, this biomaterial-based immune signal amplification system represents a unique approach to restore GBM-specific immunity and may provide a beneficial preliminary treatment for other clinically refractory malignancies.
Collapse
Affiliation(s)
- Qiujun Qiu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Sunhui Chen
- Department of Pharmacy, Fujian Provincial Hospital & Provincial Clinical Medical College of Fujian Medical University, Fuzhou 350001, China
| | - Huining He
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Jixiang Chen
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Xinyi Ding
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Dongdong Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Jiangang Yang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Pengcheng Guo
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Yang Li
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Jisu Kim
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Jianyong Sheng
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
| | - Chao Gao
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
- Institute of Tropical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Bo Yin
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Shihao Zheng
- Department of Neurosurgery, Fujian Provincial Hospital & Provincial Clinical Medical College of Fujian Medical University, Fuzhou 350001, China
| | - Jianxin Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai 201203, China
- Institute of Materia Medica, Academy of Chinese and Western Integrative Medicine, Fudan University, Shanghai 201203, China
| |
Collapse
|
15
|
Li C, Hou Y, He M, Lv L, Zhang Y, Sun S, Zhao Y, Liu X, Ma P, Wang X, Zhou Q, Zhan L. Laponite Lights Calcium Flickers by Reprogramming Lysosomes to Steer DC Migration for An Effective Antiviral CD8 + T-Cell Response. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303006. [PMID: 37638719 PMCID: PMC10602536 DOI: 10.1002/advs.202303006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/13/2023] [Indexed: 08/29/2023]
Abstract
Immunotherapy using dendritic cell (DC)-based vaccination is an established approach for treating cancer and infectious diseases; however, its efficacy is limited. Therefore, targeting the restricted migratory capacity of the DCs may enhance their therapeutic efficacy. In this study, the effect of laponite (Lap) on DCs, which can be internalized into lysosomes and induce cytoskeletal reorganization via the lysosomal reprogramming-calcium flicker axis, is evaluated, and it is found that Lap dramatically improves the in vivo homing ability of these DCs to lymphoid tissues. In addition, Lap improves antigen cross-presentation by DCs and increases DC-T-cell synapse formation, resulting in enhanced antigen-specific CD8+ T-cell activation. Furthermore, a Lap-modified cocktail (Lap@cytokine cocktail [C-C]) is constructed based on the gold standard, C-C, as an adjuvant for DC vaccines. Lap@C-C-adjuvanted DCs initiated a robust cytotoxic T-cell immune response against hepatitis B infection, resulting in > 99.6% clearance of viral DNA and successful hepatitis B surface antigen seroconversion. These findings highlight the potential value of Lap as a DC vaccine adjuvant that can regulate DC homing, and provide a basis for the development of effective DC vaccines.
Collapse
Affiliation(s)
- Chenyan Li
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P. R. China
- BGI college, Henan Institute of Medical and Pharmaceutical Science, Zhengzhou University, Zhengzhou, 450001, P. R. China
| | - Yangyang Hou
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P. R. China
| | - Minwei He
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P. R. China
| | - Liping Lv
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P. R. China
| | - Yulong Zhang
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P. R. China
| | - Sujing Sun
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P. R. China
| | - Yan Zhao
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P. R. China
| | - Xingzhao Liu
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P. R. China
| | - Ping Ma
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P. R. China
| | - Xiaohui Wang
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P. R. China
| | - Qianqian Zhou
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P. R. China
| | - Linsheng Zhan
- Institute of Health Service and Transfusion Medicine, Beijing, 100850, P. R. China
- BGI college, Henan Institute of Medical and Pharmaceutical Science, Zhengzhou University, Zhengzhou, 450001, P. R. China
| |
Collapse
|
16
|
Wang MM, Coupland SE, Aittokallio T, Figueiredo CR. Resistance to immune checkpoint therapies by tumour-induced T-cell desertification and exclusion: key mechanisms, prognostication and new therapeutic opportunities. Br J Cancer 2023; 129:1212-1224. [PMID: 37454231 PMCID: PMC10575907 DOI: 10.1038/s41416-023-02361-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 06/26/2023] [Accepted: 06/29/2023] [Indexed: 07/18/2023] Open
Abstract
Immune checkpoint therapies (ICT) can reinvigorate the effector functions of anti-tumour T cells, improving cancer patient outcomes. Anti-tumour T cells are initially formed during their first contact (priming) with tumour antigens by antigen-presenting cells (APCs). Unfortunately, many patients are refractory to ICT because their tumours are considered to be 'cold' tumours-i.e., they do not allow the generation of T cells (so-called 'desert' tumours) or the infiltration of existing anti-tumour T cells (T-cell-excluded tumours). Desert tumours disturb antigen processing and priming of T cells by targeting APCs with suppressive tumour factors derived from their genetic instabilities. In contrast, T-cell-excluded tumours are characterised by blocking effective anti-tumour T lymphocytes infiltrating cancer masses by obstacles, such as fibrosis and tumour-cell-induced immunosuppression. This review delves into critical mechanisms by which cancer cells induce T-cell 'desertification' and 'exclusion' in ICT refractory tumours. Filling the gaps in our knowledge regarding these pro-tumoral mechanisms will aid researchers in developing novel class immunotherapies that aim at restoring T-cell generation with more efficient priming by APCs and leukocyte tumour trafficking. Such developments are expected to unleash the clinical benefit of ICT in refractory patients.
Collapse
Affiliation(s)
- Mona Meng Wang
- Medical Immune Oncology Research Group (MIORG), Institute of Biomedicine, Faculty of Medicine, University of Turku, Turku, Finland
- Singapore National Eye Centre and Singapore Eye Research Institute, Singapore, Singapore
| | - Sarah E Coupland
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Liverpool Ocular Oncology Research Group (LOORG), Institute of Systems Molecular and Integrative Biology, Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, UK
| | - Tero Aittokallio
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Institute for Cancer Research, Department of Cancer Genetics, Oslo University Hospital, Oslo, Norway
- Oslo Centre for Biostatistics and Epidemiology (OCBE), Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Carlos R Figueiredo
- Medical Immune Oncology Research Group (MIORG), Institute of Biomedicine, Faculty of Medicine, University of Turku, Turku, Finland.
- InFLAMES Research Flagship Center, University of Turku, Turku, Finland.
- Turku Bioscience Centre, University of Turku, Turku, Finland.
| |
Collapse
|
17
|
Gong Z, Li H, Qian M, Bai Y, Jin H, Sun J, Zhang M, Jiao C, Huang P, Li Y, Zhang H, Wang H. Incorporation of Escherichia coli heat-labile enterotoxin B subunit into rabies virus particles enhances its immunogenicity in mice and dogs. BIOSAFETY AND HEALTH 2023; 5:308-319. [PMID: 40078911 PMCID: PMC11894999 DOI: 10.1016/j.bsheal.2023.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 03/14/2025] Open
Abstract
Although inactivated vaccines against rabies have the advantage of high safety, effective protection against rabies virus (RABV) infection often requires multiple, high-dose immunization. Incorporating a molecular adjuvant into the viral particles has been found to be a useful strategy to promote the immune effectiveness of inactivated vaccines. In this study, we constructed a recombinant virus, rCVS11-LTB, which chimerically expresses a molecular adjuvant heat-labile enterotoxin B subunit (LTB) protein on the surface of the RABV particles. Immunogenicity in vivo was found to be promoted by rCVS11-LTB through the activation of dendritic cells (DCs). Our results demonstrated that inactivated rCVS11-LTB was able to induce higher levels of virus-neutralizing antibodies (VNAs) in both mice and dogs than the parent virus rCVS11, to enhance the cellular immune response and T cell immune memory in mice, and was also able to provide 100% protection in mice from lethal doses of rabies virus, indicating its potential as a safe and effective inactivated rabies vaccine candidate.
Collapse
Affiliation(s)
- Zhiyuan Gong
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Hailun Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Meichen Qian
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yujie Bai
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Hongli Jin
- Changchun Sino Biotechnology Co., Ltd., Changchun 130012, China
| | - Jingxuan Sun
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Mengyao Zhang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Cuicui Jiao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Pei Huang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yuanyuan Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Haili Zhang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Hualei Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| |
Collapse
|
18
|
Rocca C, Soda T, De Francesco EM, Fiorillo M, Moccia F, Viglietto G, Angelone T, Amodio N. Mitochondrial dysfunction at the crossroad of cardiovascular diseases and cancer. J Transl Med 2023; 21:635. [PMID: 37726810 PMCID: PMC10507834 DOI: 10.1186/s12967-023-04498-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 09/01/2023] [Indexed: 09/21/2023] Open
Abstract
A large body of evidence indicates the existence of a complex pathophysiological relationship between cardiovascular diseases and cancer. Mitochondria are crucial organelles whose optimal activity is determined by quality control systems, which regulate critical cellular events, ranging from intermediary metabolism and calcium signaling to mitochondrial dynamics, cell death and mitophagy. Emerging data indicate that impaired mitochondrial quality control drives myocardial dysfunction occurring in several heart diseases, including cardiac hypertrophy, myocardial infarction, ischaemia/reperfusion damage and metabolic cardiomyopathies. On the other hand, diverse human cancers also dysregulate mitochondrial quality control to promote their initiation and progression, suggesting that modulating mitochondrial homeostasis may represent a promising therapeutic strategy both in cardiology and oncology. In this review, first we briefly introduce the physiological mechanisms underlying the mitochondrial quality control system, and then summarize the current understanding about the impact of dysregulated mitochondrial functions in cardiovascular diseases and cancer. We also discuss key mitochondrial mechanisms underlying the increased risk of cardiovascular complications secondary to the main current anticancer strategies, highlighting the potential of strategies aimed at alleviating mitochondrial impairment-related cardiac dysfunction and tumorigenesis. It is hoped that this summary can provide novel insights into precision medicine approaches to reduce cardiovascular and cancer morbidities and mortalities.
Collapse
Affiliation(s)
- Carmine Rocca
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, E and E.S. (DiBEST), University of Calabria, Arcavacata di Rende, 87036, Cosenza, Italy
| | - Teresa Soda
- Department of Health Science, University Magna Graecia of Catanzaro, 88100, Catanzaro, Italy
| | - Ernestina Marianna De Francesco
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Catania, Garibaldi-Nesima Hospital, 95122, Catania, Italy
| | - Marco Fiorillo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036, Rende, Italy
| | - Francesco Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100, Pavia, Italy
| | - Giuseppe Viglietto
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100, Catanzaro, Italy
| | - Tommaso Angelone
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, E and E.S. (DiBEST), University of Calabria, Arcavacata di Rende, 87036, Cosenza, Italy.
- National Institute of Cardiovascular Research (I.N.R.C.), 40126, Bologna, Italy.
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100, Catanzaro, Italy.
| |
Collapse
|
19
|
Zhou H, Ma Y, Liu F, Li B, Qiao D, Ren P, Wang M. Current advances in cancer vaccines targeting NY-ESO-1 for solid cancer treatment. Front Immunol 2023; 14:1255799. [PMID: 37731507 PMCID: PMC10508181 DOI: 10.3389/fimmu.2023.1255799] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 08/22/2023] [Indexed: 09/22/2023] Open
Abstract
New York-esophageal cancer 1 (NY-ESO-1) belongs to the cancer testis antigen (CTA) family, and has been identified as one of the most immunogenic tumor-associated antigens (TAAs) among the family members. Given its ability to trigger spontaneous humoral and cellular immune response and restricted expression, NY-ESO-1 has emerged as one of the most promising targets for cancer immunotherapy. Cancer vaccines, an important element of cancer immunotherapy, function by presenting an exogenous source of TAA proteins, peptides, and antigenic epitopes to CD4+ T cells via major histocompatibility complex class II (MHC-II) and to CD8+ T cells via major histocompatibility complex class I (MHC-I). These mechanisms further enhance the immune response against TAAs mediated by cytotoxic T lymphocytes (CTLs) and helper T cells. NY-ESO-1-based cancer vaccines have a history of nearly two decades, starting from the first clinical trial conducted in 2003. The current cancer vaccines targeting NY-ESO-1 have various types, including Dendritic cells (DC)-based vaccines, peptide vaccines, protein vaccines, viral vaccines, bacterial vaccines, therapeutic whole-tumor cell vaccines, DNA vaccines and mRNA vaccines, which exhibit their respective benefits and obstacles in the development and application. Here, we summarized the current advances in cancer vaccines targeting NY-ESO-1 for solid cancer treatment, aiming to provide perspectives for future research.
Collapse
Affiliation(s)
- Hong Zhou
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- Department of Research and Development, Shenzhen Innovation Immunotechnology Co., Ltd, Shenzhen, China
- Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen, China
| | - Yipeng Ma
- Department of Research and Development, Shenzhen Innovation Immunotechnology Co., Ltd, Shenzhen, China
- Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen, China
| | - Fenglan Liu
- Department of Research and Development, Shenzhen Innovation Immunotechnology Co., Ltd, Shenzhen, China
- Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen, China
| | - Bin Li
- Department of Research and Development, Shenzhen Innovation Immunotechnology Co., Ltd, Shenzhen, China
- Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen, China
| | - Dongjuan Qiao
- Department of Research and Development, Shenzhen Innovation Immunotechnology Co., Ltd, Shenzhen, China
- Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen, China
| | - Peigen Ren
- Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Mingjun Wang
- Department of Research and Development, Shenzhen Innovation Immunotechnology Co., Ltd, Shenzhen, China
- Department of Research and Development, Shenzhen Institute for Innovation and Translational Medicine, Shenzhen, China
| |
Collapse
|
20
|
Lee KW, Yam JWP, Mao X. Dendritic Cell Vaccines: A Shift from Conventional Approach to New Generations. Cells 2023; 12:2147. [PMID: 37681880 PMCID: PMC10486560 DOI: 10.3390/cells12172147] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/21/2023] [Accepted: 08/23/2023] [Indexed: 09/09/2023] Open
Abstract
In the emerging era of cancer immunotherapy, immune checkpoint blockades (ICBs) and adoptive cell transfer therapies (ACTs) have gained significant attention. However, their therapeutic efficacies are limited due to the presence of cold type tumors, immunosuppressive tumor microenvironment, and immune-related side effects. On the other hand, dendritic cell (DC)-based vaccines have been suggested as a new cancer immunotherapy regimen that can address the limitations encountered by ICBs and ACTs. Despite the success of the first generation of DC-based vaccines, represented by the first FDA-approved DC-based therapeutic cancer vaccine Provenge, several challenges remain unsolved. Therefore, new DC vaccine strategies have been actively investigated. This review addresses the limitations of the currently most adopted classical DC vaccine and evaluates new generations of DC vaccines in detail, including biomaterial-based, immunogenic cell death-inducing, mRNA-pulsed, DC small extracellular vesicle (sEV)-based, and tumor sEV-based DC vaccines. These innovative DC vaccines are envisioned to provide a significant breakthrough in cancer immunotherapy landscape and are expected to be supported by further preclinical and clinical studies.
Collapse
Affiliation(s)
- Kyu-Won Lee
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; (K.-W.L.); (J.W.P.Y.)
| | - Judy Wai Ping Yam
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong; (K.-W.L.); (J.W.P.Y.)
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong
| | - Xiaowen Mao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao
| |
Collapse
|
21
|
Wang R, Zhu T, Hou B, Huang X. An iPSC-derived exosome-pulsed dendritic cell vaccine boosts antitumor immunity in melanoma. Mol Ther 2023; 31:2376-2390. [PMID: 37312452 PMCID: PMC10422017 DOI: 10.1016/j.ymthe.2023.06.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 03/23/2023] [Accepted: 06/08/2023] [Indexed: 06/15/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) express a broad spectrum of tumor-associated antigens and exert prophylactic effects on various tumors. However, some problems remain, such as potential tumorigenicity, challenges in transport to the lymph nodes and spleen, and limited antitumor effects. Thus, designing a safe and effective iPSC-based tumor vaccine is necessary. We prepared iPSC-derived exosomes and incubated them with DCs (dendritic cells) for pulsing to explore their antitumor effects in murine melanoma models. The antitumor immune response induced by the DC vaccine pulsed with iPSC exosomes (DC + EXO) was assessed in vitro and in vivo. After DC + EXO vaccination, extracted spleen T cells effectively killed a variety of tumor cells (melanoma, lung cancer, breast cancer, and colorectal cancer) in vitro. In addition, DC + EXO vaccination significantly inhibited melanoma growth and lung metastasis in mouse models. Furthermore, DC + EXO vaccination induced long-term T cell responses and prevented melanoma rechallenge. Finally, biocompatibility studies showed that the DC vaccine did not significantly alter the viability of normal cells and mouse viscera. Hence, our research may provide a prospective strategy of a safe and effective iPSC-based tumor vaccine for clinical use.
Collapse
Affiliation(s)
- Ronghao Wang
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China; Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China
| | - Tianchuan Zhu
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China
| | - Bingzong Hou
- Department of Gastrointestinal Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China.
| | - Xi Huang
- Center for Infection and Immunity and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 519000, Guangdong, China; Southern Marine Science and Engineering Guangdong Laboratory, Zhuhai 519000, Guangdong, China.
| |
Collapse
|
22
|
Zhao J, Liao B, Gong L, Yang H, Li S, Li Y. Knowledge mapping of therapeutic cancer vaccine from 2013 to 2022: A bibliometric and visual analysis. Hum Vaccin Immunother 2023; 19:2254262. [PMID: 37728107 PMCID: PMC10512878 DOI: 10.1080/21645515.2023.2254262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 08/29/2023] [Indexed: 09/21/2023] Open
Abstract
The investigation of therapeutic cancer vaccines has been ongoing for the past century. Herein, we used VOSviewer and CiteSpace to perform the first global bibliometric analysis of the literature on therapeutic cancer vaccines from 2013 to 2022 aiming to explore the current status and potential research trends. The findings revealed a consistent upward trend in both publication counts and citations. The United States emerged as the leading contributor with the highest number of published papers. Additionally, the analysis of references and keywords indicated that therapeutic cancer vaccines have long been popular topics, whereas neoantigen vaccines, mRNA vaccines, combination strategies, and vaccine delivery systems are emerging research hotspots. This bibliometric study provides a comprehensive and important overview of the current knowledge and potential developments in therapeutic cancer vaccines from 2013 to 2022, which may serve as a valuable reference for scholars interested in further exploring this promising field.
Collapse
Affiliation(s)
- Juan Zhao
- Department of Phase I Clinical Trial Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Bin Liao
- Department of Phase I Clinical Trial Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Li Gong
- Department of Phase I Clinical Trial Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Huiyao Yang
- Department of Phase I Clinical Trial Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Sha Li
- Department of Phase I Clinical Trial Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Yongsheng Li
- Department of Phase I Clinical Trial Center, Chongqing University Cancer Hospital, Chongqing, China
| |
Collapse
|
23
|
Qiao X, Zhu L, Song R, Shang C, Guo Y. A novel oncogene trigger transposable element derived-1 promotes oral squamous cell carcinoma progression via evoking immune inhibition. Mol Carcinog 2023. [PMID: 37144838 DOI: 10.1002/mc.23557] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/22/2023] [Accepted: 04/25/2023] [Indexed: 05/06/2023]
Abstract
Oral squamous cell carcinoma (OSCC) is one of the most common head and neck squamous cell carcinomas (HNSCC) globally. Its incidence rate is rapidly increasing, and its 5-year survival rate remains at 50%, despite advances in medical science. Trigger transposable element-derived 1 (TIGD1) has been found to be upregulated in various cancer types. However, its biological function in OSCC requires further investigation. We searched the Cancer Genome Atlas database using CIBERSORT and TIMER 2.0 to predict the significance of TIGD1 and evaluate its effect on immune cell infiltration. Gene set enrichment analysis was performed to determine the biological functions of TIGD1. Gain/loss of function techniques were used to explore the biological behavior of TIGD1 in Cal27 and HSC4 cells. Finally, flow cytometry was used to detect dendritic cell markers in an OSCC and dendritic cell co-culture model. Our results show that TIGD1 is upregulated significantly in OSCC and is closely associated with tumor progression and prognosis. TIGD1 functions as an oncogene by increasing cells proliferation, inhibiting apoptosis, promoting cell invasion and migration. TIGD1 is also involved in tumor immune cell infiltration. Its overexpression can inhibit dendritic cell maturation, leading to immune suppression and tumor progression. High TIGD1 expression, which promotes OSCC progression, might be related to decreased dendritic cell maturation and activation. These findings suggest that TIGD1-specific small interfering RNA synthesized in vitro could be a new target for OSCC immunotherapy.
Collapse
Affiliation(s)
- Xue Qiao
- Department of Central Laboratory, Liaoning Provincial Key Laboratory of Oral Disease, School and Hospital of Stomatology, China Medical University, Shenyang, Liaoning, China
- Department of Oral Biology, Liaoning Provincial Key Laboratory of Oral Disease, School and Hospital of Stomatology, China Medical University, Shenyang, Liaoning, China
| | - Li Zhu
- Department of Central Laboratory, Liaoning Provincial Key Laboratory of Oral Disease, School and Hospital of Stomatology, China Medical University, Shenyang, Liaoning, China
| | - Rongbo Song
- Department of Central Laboratory, Liaoning Provincial Key Laboratory of Oral Disease, School and Hospital of Stomatology, China Medical University, Shenyang, Liaoning, China
| | - Chao Shang
- Department of Neurobiology, China Medical University, Shenyang, Liaoning, China
| | - Yan Guo
- Department of Central Laboratory, Liaoning Provincial Key Laboratory of Oral Disease, School and Hospital of Stomatology, China Medical University, Shenyang, Liaoning, China
- Department of Oral Biology, Liaoning Provincial Key Laboratory of Oral Disease, School and Hospital of Stomatology, China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
24
|
Gong Z, Huang P, Jin H, Bai Y, Li H, Qian M, Sun J, Jiao C, Zhang M, Li Y, Zhang H, Wang H. A recombinant rabies virus chimera expressing the DC-targeting molecular MAB2560 shows enhanced vaccine immunogenicity through activation of dendritic cells. PLoS Negl Trop Dis 2023; 17:e0011254. [PMID: 37093869 PMCID: PMC10124880 DOI: 10.1371/journal.pntd.0011254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 03/20/2023] [Indexed: 04/25/2023] Open
Abstract
BACKGROUND Rabies, caused by the rabies virus (RABV), is an ancient and neglected zoonotic disease posing a large public health threat to humans and animals in developing countries. Immunization of animals with a rabies vaccine is the most effective way to control the epidemic and the occurrence of the disease in humans. Therefore, the development of cost-effective and efficient rabies vaccines is urgently needed. The activation of dendritic cells (DCs) is known to play an important role in improving the host immune response induced by rabies vaccines. METHODOLOGY/PRINCIPAL FINDINGS In this study, we constructed a recombinant virus, rCVS11-MAB2560, based on the reverse genetic system of the RABV CVS11 strain. The MAB2560 protein (a DC-targeting molecular) was chimeric expressed on the surface of the viral particles to help target and activate the DCs when this virus was used as inactivated vaccine. Our results demonstrated that inactivated rCVS11-MAB2560 was able to promote the recruitment and/or proliferation of DC cells, T cells and B cells in mice, and induce good immune memory after two immunizations. Moreover, the inactivated recombinant virus rCVS11-MAB2560 could produce higher levels of virus-neutralizing antibodies (VNAs) in both mice and dogs more quickly than rCVS11 post immunization. CONCLUSIONS/SIGNIFICANCE In summary, the recombinant virus rCVS11-MAB2560 chimeric-expressing the molecular adjuvant MAB2560 can stimulate high levels of humoral and cellular immune responses in vivo and can be used as an effective inactivated rabies vaccine candidate.
Collapse
Affiliation(s)
- Zhiyuan Gong
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Pei Huang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Hongli Jin
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
- Changchun Sino Biotechnology Co., Ltd., Changchun, China
| | - Yujie Bai
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Hailun Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Meichen Qian
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jingxuan Sun
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Cuicui Jiao
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Mengyao Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yuanyuan Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Haili Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Hualei Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
25
|
Wang Q, Wang K, Tan X, Li Z, Wang H. Immunomodulatory role of metalloproteases in cancers: Current progress and future trends. Front Immunol 2022; 13:1064033. [PMID: 36591235 PMCID: PMC9800621 DOI: 10.3389/fimmu.2022.1064033] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
Metalloproteinases (MPs) is a large family of proteinases with metal ions in their active centers. According to the different domains metalloproteinases can be divided into a variety of subtypes mainly including Matrix Metalloproteinases (MMPs), A Disintegrin and Metalloproteases (ADAMs) and ADAMs with Thrombospondin Motifs (ADAMTS). They have various functions such as protein hydrolysis, cell adhesion and remodeling of extracellular matrix. Metalloproteinases expressed in multiple types of cancers and participate in many pathological processes involving tumor genesis and development, invasion and metastasis by regulating signal transduction and tumor microenvironment. In this review, based on the current research progress, we summarized the structure of MPs, their expression and especially immunomodulatory role and mechanisms in cancers. Additionally, a relevant and timely update of recent advances and future directions were provided for the diagnosis and immunotherapy targeting MPs in cancers.
Collapse
Affiliation(s)
- Qi Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Kai Wang
- Key Laboratory of Epigenetics and Oncology, Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China
| | - Xiaojing Tan
- Department of Oncology, Dongying People's Hospital, Dongying, China
| | - Zhenxiang Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China,*Correspondence: Zhenxiang Li, ; Haiyong Wang,
| | - Haiyong Wang
- Department of Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China,*Correspondence: Zhenxiang Li, ; Haiyong Wang,
| |
Collapse
|
26
|
Qiao DR, Shan GY, Wang S, Cheng JY, Yan WQ, Li HJ. The mononuclear phagocyte system in hepatocellular carcinoma. World J Gastroenterol 2022; 28:6345-6355. [PMID: 36533105 PMCID: PMC9753057 DOI: 10.3748/wjg.v28.i45.6345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 10/10/2022] [Accepted: 11/16/2022] [Indexed: 12/02/2022] Open
Abstract
The mononuclear phagocyte system (MPS) consists of monocytes, dendritic cells and macrophages, which play vital roles in innate immune defense against cancer. Hepatocellular carcinoma (HCC) is a complex disease that is affected or initiated by many factors, including chronic hepatitis B virus infection, hepatitis C virus infection, metabolic disorders or alcohol consumption. Liver function, tumor stage and the performance status of patients affect HCC clinical outcomes. Studies have shown that targeted treatment of tumor microenvironment disorders may improve the efficacy of HCC treatments. Cytokines derived from the innate immune response can regulate T-cell differentiation, thereby shaping adaptive immunity, which is associated with the prognosis of HCC. Therefore, it is important to elucidate the function of the MPS in the progression of HCC. In this review, we outline the impact of HCC on the MPS. We illustrate how HCC reshapes MPS cell phenotype remodeling and the production of associated cytokines and characterize the function and impairment of the MPS in HCC.
Collapse
Affiliation(s)
- Duan-Rui Qiao
- Department of Bioengineering, Pharmacy School of Jilin University, Changchun 130021, Jilin Province, China
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Guan-Yue Shan
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Shuai Wang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
- Department of Students Affairs, China-Japan Union Hospital of Jilin University, Changchun 130031, Jilin Province, China
| | - Jun-Ya Cheng
- Department of Bioengineering, Pharmacy School of Jilin University, Changchun 130021, Jilin Province, China
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| | - Wei-Qun Yan
- Department of Bioengineering, Pharmacy School of Jilin University, Changchun 130021, Jilin Province, China
| | - Hai-Jun Li
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
- Institute of Liver Diseases, The First Hospital of Jilin University, Changchun 130061, Jilin Province, China
| |
Collapse
|
27
|
Van Delen M, Janssens I, Dams A, Roosens L, Ogunjimi B, Berneman ZN, Derdelinckx J, Cools N. Tolerogenic Dendritic Cells Induce Apoptosis-Independent T Cell Hyporesponsiveness of SARS-CoV-2-Specific T Cells in an Antigen-Specific Manner. Int J Mol Sci 2022; 23:15201. [PMID: 36499533 PMCID: PMC9740551 DOI: 10.3390/ijms232315201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Although the global pandemic caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is still ongoing, there are currently no specific and highly efficient drugs for COVID-19 available, particularly in severe cases. Recent findings demonstrate that severe COVID-19 disease that requires hospitalization is associated with the hyperactivation of CD4+ and CD8+ T cell subsets. In this study, we aimed to counteract this high inflammatory state by inducing T-cell hyporesponsiveness in a SARS-CoV-2-specific manner using tolerogenic dendritic cells (tolDC). In vitro-activated SARS-CoV-2-specific T cells were isolated and stimulated with SARS-CoV-2 peptide-loaded monocyte-derived tolDC or with SARS-CoV-2 peptide-loaded conventional (conv) DC. We demonstrate a significant decrease in the number of interferon (IFN)-γ spot-forming cells when SARS-CoV-2-specific T cells were stimulated with tolDC as compared to stimulation with convDC. Importantly, this IFN-γ downmodulation in SARS-CoV-2-specific T cells was antigen-specific, since T cells retain their capacity to respond to an unrelated antigen and are not mediated by T cell deletion. Altogether, we have demonstrated that SARS-CoV-2 peptide-pulsed tolDC induces SARS-CoV-2-specific T cell hyporesponsiveness in an antigen-specific manner as compared to stimulation with SARS-CoV-2-specific convDC. These observations underline the clinical potential of tolDC to correct the immunological imbalance in the critically ill.
Collapse
Affiliation(s)
- Mats Van Delen
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Ibo Janssens
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Amber Dams
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
| | - Laurence Roosens
- Laboratory of Clinical Biology, Antwerp University Hospital, 2650 Edegem, Belgium
| | - Benson Ogunjimi
- Centre for Health Economics Research & Modeling Infectious Diseases (CHERMID), VAXINFECTIO, University of Antwerp, 2610 Antwerp, Belgium
- Department of Paediatrics, Antwerp University Hospital, 2650 Edegem, Belgium
- Antwerp Center for Translational Immunology and Virology (ACTIV), VAXINFECTIO, University of Antwerp, 2610 Antwerp, Belgium
- Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, 2020 Antwerp, Belgium
| | - Zwi N. Berneman
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, 2650 Edegem, Belgium
| | - Judith Derdelinckx
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
- Department of Neurology, Antwerp University Hospital, 2650 Edegem, Belgium
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, 2650 Edegem, Belgium
| |
Collapse
|
28
|
Yu G, He X, Li X, Wu Y. Driving neoantigen-based cancer vaccines for personalized immunotherapy into clinic: A burdensome journey to promising land. Biomed Pharmacother 2022; 153:113464. [DOI: 10.1016/j.biopha.2022.113464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/15/2022] [Accepted: 07/21/2022] [Indexed: 11/02/2022] Open
|
29
|
Barshidi A, Karpisheh V, Noukabadi FK, Kiani FK, Mohammadi M, Afsharimanesh N, Ebrahimi F, Kiaie SH, Navashenaq JG, Hojjat-Farsangi M, Zolbanin NM, Mahmoodpoor A, Hassannia H, Nami S, Jalali P, Jafari R, Jadidi-Niaragh F. Dual Blockade of PD-1 and LAG3 Immune Checkpoints Increases Dendritic Cell Vaccine Mediated T Cell Responses in Breast Cancer Model. Pharm Res 2022; 39:1851-1866. [PMID: 35715669 DOI: 10.1007/s11095-022-03297-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/18/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE Increasing the efficiency of unsuccessful immunotherapy methods is one of the most important research fields. Therefore, the use of combination therapy is considered as one of the ways to increase the effectiveness of the dendritic cell (DC) vaccine. In this study, the inhibition of immune checkpoint receptors such as LAG3 and PD-1 on T cells was investigated to increase the efficiency of T cells in response to the DC vaccine. METHODS We used trimethyl chitosan-dextran sulfate-lactate (TMC-DS-L) nanoparticles (NPs) loaded with siRNA molecules to quench the PD-1 and LAG3 checkpoints' expression. RESULTS Appropriate physicochemical characteristics of the generated NPs led to efficient inhibition of LAG3 and PD-1 on T cells, which was associated with increased survival and activity of T cells, ex vivo. Also, treating mice with established breast tumors (4T1) using NPs loaded with siRNA molecules in combination with DC vaccine pulsed with tumor lysate significantly inhibited tumor growth and increased survival in mice. These ameliorative effects were associated with increased anti-tumor T cell responses and downregulation of immunosuppressive cells in the tumor microenvironment and spleen. CONCLUSION These findings strongly suggest that TMC-DS-L NPs loaded with siRNA could act as a novel tool in inhibiting the expression of immune checkpoints in the tumor microenvironment. Also, combination therapy based on inhibition of PD-1 and LAG3 in combination with DC vaccine is an effective method in treating cancer that needs to be further studied.
Collapse
Affiliation(s)
- Asal Barshidi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Karpisheh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Fariba Karoon Kiani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Mohammadi
- Department of Cell and Molecular Biology, School of Advanced Sciences, Islamic Azad University, Tehran Medical Branch, Tehran, , Iran
| | - Negin Afsharimanesh
- Department of Microbiology, Faculty of Basic Sciences, Hamedan Branch, Islamic Azad University, Hamedan, Iran
| | - Farbod Ebrahimi
- Nanoparticle Process Technology, Faculty of Engineering, University of Duisburg-Essen, Duisburg, Germany
| | - Seyed Hossein Kiaie
- Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
- Nano Drug Delivery Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | | | - Mohammad Hojjat-Farsangi
- Bioclinicum, Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
- The Persian Gulf Marine Biotechnology Medicine Research Center, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Naime Majidi Zolbanin
- Experimental and Applied Pharmaceutical Sciences Research Center,, Urmia University of Medical Sciences, Urmia, Iran
- Department of Pharmacology and Toxicology, School of Pharmacy, , Urmia University of Medical Sciences, Urmia, Iran
| | - Ata Mahmoodpoor
- Department of Anesthesiology, School of Medicine, Imam Reza Medical Research & Training Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Hassannia
- Immunogenetic Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Sanam Nami
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pooya Jalali
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Jafari
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran.
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
30
|
Chen WH, Chen QW, Chen Q, Cui C, Duan S, Kang Y, Liu Y, Liu Y, Muhammad W, Shao S, Tang C, Wang J, Wang L, Xiong MH, Yin L, Zhang K, Zhang Z, Zhen X, Feng J, Gao C, Gu Z, He C, Ji J, Jiang X, Liu W, Liu Z, Peng H, Shen Y, Shi L, Sun X, Wang H, Wang J, Xiao H, Xu FJ, Zhong Z, Zhang XZ, Chen X. Biomedical polymers: synthesis, properties, and applications. Sci China Chem 2022; 65:1010-1075. [PMID: 35505924 PMCID: PMC9050484 DOI: 10.1007/s11426-022-1243-5] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/01/2022] [Indexed: 02/07/2023]
Abstract
Biomedical polymers have been extensively developed for promising applications in a lot of biomedical fields, such as therapeutic medicine delivery, disease detection and diagnosis, biosensing, regenerative medicine, and disease treatment. In this review, we summarize the most recent advances in the synthesis and application of biomedical polymers, and discuss the comprehensive understanding of their property-function relationship for corresponding biomedical applications. In particular, a few burgeoning bioactive polymers, such as peptide/biomembrane/microorganism/cell-based biomedical polymers, are also introduced and highlighted as the emerging biomaterials for cancer precision therapy. Furthermore, the foreseeable challenges and outlook of the development of more efficient, healthier and safer biomedical polymers are discussed. We wish this systemic and comprehensive review on highlighting frontier progress of biomedical polymers could inspire and promote new breakthrough in fundamental research and clinical translation.
Collapse
Affiliation(s)
- Wei-Hai Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072 China
| | - Qi-Wen Chen
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072 China
| | - Qian Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123 China
| | - Chunyan Cui
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300350 China
| | - Shun Duan
- Key Laboratory of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029 China
| | - Yongyuan Kang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027 China
| | - Yang Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin, 300071 China
| | - Yun Liu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China
- Jinhua Institute of Zhejiang University, Jinhua, 321299 China
| | - Wali Muhammad
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027 China
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart BioMaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027 China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 311215 China
| | - Chengqiang Tang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science and Laboratory of Advanced Materials, Fudan University, Shanghai, 200438 China
| | - Jinqiang Wang
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China
- Jinhua Institute of Zhejiang University, Jinhua, 321299 China
| | - Lei Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nano-science, National Center for Nanoscience and Technology (NCNST), Beijing, 100190 China
| | - Meng-Hua Xiong
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006 China
| | - Lichen Yin
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Collaborative Innovation Center of Suzhou Nano Science & Technology, Soochow University, Suzhou, 215123 China
| | - Kuo Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nano-science, National Center for Nanoscience and Technology (NCNST), Beijing, 100190 China
| | - Zhanzhan Zhang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin, 300071 China
| | - Xu Zhen
- Department of Polymer Science and Engineering, College of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093 China
| | - Jun Feng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072 China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027 China
| | - Zhen Gu
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058 China
- Jinhua Institute of Zhejiang University, Jinhua, 321299 China
| | - Chaoliang He
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022 China
| | - Jian Ji
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027 China
| | - Xiqun Jiang
- Department of Polymer Science and Engineering, College of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093 China
| | - Wenguang Liu
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300350 China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou, 215123 China
| | - Huisheng Peng
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science and Laboratory of Advanced Materials, Fudan University, Shanghai, 200438 China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart BioMaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027 China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 311215 China
| | - Linqi Shi
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Functional Polymer Materials of Ministry of Education, College of Chemistry, Nankai University, Tianjin, 300071 China
| | - Xuemei Sun
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science and Laboratory of Advanced Materials, Fudan University, Shanghai, 200438 China
| | - Hao Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nano-science, National Center for Nanoscience and Technology (NCNST), Beijing, 100190 China
| | - Jun Wang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 510006 China
| | - Haihua Xiao
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Polymer Physics and Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190 China
| | - Fu-Jian Xu
- Key Laboratory of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, 100029 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
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123 China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan, 430072 China
| | - Xuesi Chen
- CAS Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022 China
| |
Collapse
|
31
|
Sun L, Shen F, Xiong Z, Yang H, Dong Z, Xiang J, Gu Q, Ji Q, Fan C, Liu Z. DNA Engineered Lymphocyte-Based Homologous Targeting Artificial Antigen-Presenting Cells for Personalized Cancer Immunotherapy. J Am Chem Soc 2022; 144:7634-7645. [PMID: 35438987 DOI: 10.1021/jacs.1c09316] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Artificial antigen-presenting cells (aAPCs) constructed by integrating T cell activation ligands on biocompatible materials hold great potential in tumor immunotherapy. However, it remains challenging to develop aAPCs, which could mimic the characteristics of natural APCs, thereby realizing antigen-specific T cells activation in vivo. Here, we report the first effort to construct natural lymphocyte-based homologous targeting aAPCs (LC-aAPCs) with lipid-DNA-mediated noninvasive live cell surface engineering. Through a predesigned bottom-up self-assembly path, we achieved natural-APC-mimicking distribution of T cell activation ligands on LC-aAPCs, which would enable the optimized T cell activation. Moreover, the lipid-DNA-mediated self-assembly occurring on lipid bilayers would not affect the functions of homing receptors expressed on lymphocyte. Therefore, such LC-aAPCs could actively migrate to peripheral lymphatic organs and then effectively activate antigen-specific T cells. Combined with an immune checkpoint inhibitor, such LC-aAPCs could effectively inhibit the growth of different tumor models. Thus, our work provides a new design of aAPCs for in vivo applications in tumor immunotherapy, and the lipid-DNA-mediated noninvasive live cell surface engineering would be a powerful tool for designing cell-based therapeutics.
Collapse
Affiliation(s)
- Lele Sun
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices, Soochow University, Suzhou 215123, Jiangsu, China
| | - Fengyun Shen
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices, Soochow University, Suzhou 215123, Jiangsu, China
| | - Zijian Xiong
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices, Soochow University, Suzhou 215123, Jiangsu, China
| | - He Yang
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices, Soochow University, Suzhou 215123, Jiangsu, China
| | - Ziliang Dong
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices, Soochow University, Suzhou 215123, Jiangsu, China
| | - Jian Xiang
- WuXi AppTec (Suzhou) Co., Ltd., 1336 Wuzhong Avenue, Wuzhong District, Suzhou 215104, Jiangsu, China
| | - Qingyang Gu
- WuXi AppTec (Suzhou) Co., Ltd., 1336 Wuzhong Avenue, Wuzhong District, Suzhou 215104, Jiangsu, China
| | - Qunsheng Ji
- WuXi AppTec (Suzhou) Co., Ltd., 1336 Wuzhong Avenue, Wuzhong District, Suzhou 215104, Jiangsu, China
| | - Chunhai Fan
- Natl Ctr Translat Med, Frontiers Sci Ctr Transformat Mol, Sch Chem & Chem Engn, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM) Jiangsu Key Lab Carbon Based Functional Materials and Devices, Soochow University, Suzhou 215123, Jiangsu, China
| |
Collapse
|
32
|
Kalantar K, Gholijani N, Martinuzzi E, Culina S, Kabelitz D, Amirghofran Z. Accelerated co-cultured dendritic cell (acDC) loaded with autologous apoptotic bodies might be a promising approach for antigen delivery. J Immunoassay Immunochem 2022; 43:467-479. [PMID: 35301912 DOI: 10.1080/15321819.2022.2048010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Antigens derived from engulfed apoptotic bodies that are presented by dendritic cells can amplify Ag-specific T-cells. Accelerated co-cultured DC (acDC) strategy keeps lymphocytes in contact with differentiating DCs. Therefore, Ag-specific T-cell activation can occur during DC maturation. Our aim was to prepare DCs by acDC method and check the subsequent engulfment of the apoptotic body by acDC. We have proposed that this method could be feasible if we transfect the apoptotic bodies with the antigen. DCs were prepared using acDC method and their maturation markers were confirmed by flow cytometry. Ultraviolet was used for inducing apoptosis in the PBMCs and induction of apoptosis checked by propidium iodide and 7-aminoactinomycin D staining. Flow cytometry and immunohistochemistry were used for checking the uptake of apoptotic bodies by the DCs. The alloreactivity against apoptotic bodies was examined by enzyme-linked immunospot (ELISPOT) assay. Results showed that 40.4% of DCs could efficiently engulf the apoptotic bodies. The results indicated that acDC method is capable to isolate a high yield of DCs, and these cells could properly engulf the apoptotic bodies, more works should be performed to use this method for Ag discovery through delivering the Ag by apoptotic bodies into the DCs.
Collapse
Affiliation(s)
- Kurosh Kalantar
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,Autoimmune Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nasser Gholijani
- Autoimmune Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Emanuela Martinuzzi
- Système immunitaire, cerveau et nerfs périphériques, Institute de Pharmacologie Moléculaire et Cellulaire (IPMC) CNRS, Valbonne, France
| | - Slobodan Culina
- Université de Paris, Institut Cochin, CNRS, INSERM, Paris, France
| | | | - Zahra Amirghofran
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.,Autoimmune Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
33
|
Ruan S, Greenberg Z, Pan X, Zhuang P, Erwin N, He M. Extracellular Vesicles as an Advanced Delivery Biomaterial for Precision Cancer Immunotherapy. Adv Healthc Mater 2022; 11:e2100650. [PMID: 34197051 PMCID: PMC8720116 DOI: 10.1002/adhm.202100650] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 06/22/2021] [Indexed: 12/11/2022]
Abstract
In recent years, cancer immunotherapy has been observed in numerous preclinical and clinical studies for showing benefits. However, due to the unpredictable outcomes and low response rates, novel targeting delivery approaches and modulators are needed for being effective to more broader patient populations and cancer types. Compared to synthetic biomaterials, extracellular vesicles (EVs) specifically open a new avenue for improving the efficacy of cancer immunotherapy by offering targeted and site-specific immunity modulation. In this review, the molecular understanding of EV cargos and surface receptors, which underpin cell targeting specificity and precisely modulating immunogenicity, are discussed. Unique properties of EVs are reviewed in terms of their surface markers, intravesicular contents, intrinsic immunity modulatory functions, and pharmacodynamic behavior in vivo with tumor tissue models, highlighting key indications of improved precision cancer immunotherapy. Novel molecular engineered strategies for reprogramming and directing cancer immunotherapeutics, and their unique challenges are also discussed to illuminate EV's future potential as a cancer immunotherapeutic biomaterial.
Collapse
Affiliation(s)
- Shaobo Ruan
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Zachary Greenberg
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Xiaoshu Pan
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Pei Zhuang
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Nina Erwin
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Mei He
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| |
Collapse
|
34
|
Meng Z, Zhang Y, Zhou X, Ji J, Liu Z. Nanovaccines with cell-derived components for cancer immunotherapy. Adv Drug Deliv Rev 2022; 182:114107. [PMID: 34995678 DOI: 10.1016/j.addr.2021.114107] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 10/16/2021] [Accepted: 12/29/2021] [Indexed: 12/13/2022]
Abstract
Cancer nanovaccines as one of immunotherapeutic approaches are able to attack tumors by stimulating tumor-specific immunological responses. However, there still exist multiple challenges to be tackled for cancer nanovaccines to evoke potent antitumor immunity. Particularly, the administration of exogenous materials may cause the off-target immunotherapy responses. In recent years, biomimetic nanovaccines by using cell lysates, cell-derived nanovesicles, or extracted cell membranes as the functional components have received extensive attention. Such nanovaccines based on cell-derived components would show many unique advantages including inherent biocompatibility and the ability to trigger immune responses against a range of tumor-associated antigens. In this review article, we will introduce the recent research progresses of those cell-derived biomimetic nanovaccines for cancer immunotherapy, and discuss the perspectives and challenges associated with the future clinical translation of these emerging vaccine platforms.
Collapse
|
35
|
Emerging strategies for biomaterial-assisted cancer immunotherapy. KOREAN J CHEM ENG 2022. [DOI: 10.1007/s11814-021-0985-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
36
|
Shafaati M, Saidijam M, Soleimani M, Hazrati F, Mirzaei R, Amirheidari B, Tanzadehpanah H, Karampoor S, Kazemi S, Yavari B, Mahaki H, Safaei M, Rahbarizadeh F, Samadi P, Ahmadyousefi Y. A brief review on DNA vaccines in the era of COVID-19. Future Virol 2021; 17:10.2217/fvl-2021-0170. [PMID: 34858516 PMCID: PMC8629371 DOI: 10.2217/fvl-2021-0170] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 11/05/2021] [Indexed: 02/08/2023]
Abstract
This article provides a brief overview of DNA vaccines. First, the basic DNA vaccine design strategies are described, then specific issues related to the industrial production of DNA vaccines are discussed, including the production and purification of DNA products such as plasmid DNA, minicircle DNA, minimalistic, immunologically defined gene expression (MIDGE) and Doggybone™. The use of adjuvants to enhance the immunogenicity of DNA vaccines is then discussed. In addition, different delivery routes and several physical and chemical methods to increase the efficacy of DNA delivery into cells are explained. Recent preclinical and clinical trials of DNA vaccines for COVID-19 are then summarized. Lastly, the advantages and obstacles of DNA vaccines are discussed.
Collapse
Affiliation(s)
- Maryam Shafaati
- Department of Microbiology, Faculty of Sciences, Jahrom Branch, Islamic Azad University, Jahrom, Iran
| | - Massoud Saidijam
- Department of Medical Biotechnology, School of Advanced Medical Sciences & Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
- Research Center for Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Meysam Soleimani
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Fereshte Hazrati
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rasoul Mirzaei
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Bagher Amirheidari
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
- Extremophile and Productive Microorganisms Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamid Tanzadehpanah
- Department of Medical Biotechnology, School of Advanced Medical Sciences & Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
- Research Center for Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sajad Karampoor
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sima Kazemi
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Bahram Yavari
- Department of Medical Biotechnology, School of Advanced Medical Sciences & Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
- Research Center for Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hanie Mahaki
- Vascular & Endovascular Surgery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Safaei
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Fatemeh Rahbarizadeh
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Pouria Samadi
- Department of Medical Biotechnology, School of Advanced Medical Sciences & Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
- Research Center for Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Yaghoub Ahmadyousefi
- Department of Medical Biotechnology, School of Advanced Medical Sciences & Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
- Research Center for Molecular Medicine, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
37
|
Cao J, Bhatnagar S, Wang J, Qi X, Prabha S, Panyam J. Cancer stem cells and strategies for targeted drug delivery. Drug Deliv Transl Res 2021; 11:1779-1805. [PMID: 33095384 PMCID: PMC8062588 DOI: 10.1007/s13346-020-00863-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2020] [Indexed: 12/23/2022]
Abstract
Cancer stem cells (CSCs) are a small proportion of cancer cells with high tumorigenic activity, self-renewal ability, and multilineage differentiation potential. Standard anti-tumor therapies including conventional chemotherapy, radiation therapy, and molecularly targeted therapies are not effective against CSCs, and often lead to enrichment of CSCs that can result in tumor relapse. Therefore, it is hypothesized that targeting CSCs is key to increasing the efficacy of cancer therapies. In this review, CSC properties including CSC markers, their role in tumor growth, invasiveness, metastasis, and drug resistance, as well as CSC microenvironment are discussed. Further, CSC-targeted strategies including the use of targeted drug delivery systems are examined.
Collapse
Affiliation(s)
- Jin Cao
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
- College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Shubhmita Bhatnagar
- College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA
- School of Pharmacy, Temple University, Philadelphia, PA, 19140, USA
| | - Jiawei Wang
- College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA
- College of Pharmacy, University of Texas at Austin, Austin, TX, 78712, USA
| | - Xueyong Qi
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Swayam Prabha
- College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA
- Cancer Research & Molecular Biology and Department of Pharmacology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Jayanth Panyam
- College of Pharmacy, University of Minnesota, Minneapolis, MN, 55455, USA.
- School of Pharmacy, Temple University, Philadelphia, PA, 19140, USA.
| |
Collapse
|
38
|
Ye T, Li F, Ma G, Wei W. Enhancing therapeutic performance of personalized cancer vaccine via delivery vectors. Adv Drug Deliv Rev 2021; 177:113927. [PMID: 34403752 DOI: 10.1016/j.addr.2021.113927] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 07/29/2021] [Accepted: 08/10/2021] [Indexed: 12/21/2022]
Abstract
In recent years, personalized cancer vaccines have gained increasing attention as emerging immunotherapies with the capability to overcome interindividual differences and show great benefits for individual patients in the clinic due to the highly tailored vaccine formulations. A large number of materials have been studied as delivery vectors to enhance the therapeutic performance of personalized cancer vaccines, including artificial materials, engineered microorganisms, cells and cell derivatives. These delivery vectors with distinct features are employed to change antigen biodistributions and to facilitate antigen uptake, processing and presentation, improving the strength, velocity, and duration of the immune response when delivered by different strategies. Here, we provide an overview of personalized cancer vaccine delivery vectors, describing their materials, physicochemical properties, delivery strategies and challenges for clinical transformation.
Collapse
|
39
|
Maiorano BA, Schinzari G, Ciardiello D, Rodriquenz MG, Cisternino A, Tortora G, Maiello E. Cancer Vaccines for Genitourinary Tumors: Recent Progresses and Future Possibilities. Vaccines (Basel) 2021; 9:623. [PMID: 34207536 PMCID: PMC8228524 DOI: 10.3390/vaccines9060623] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/27/2021] [Accepted: 06/04/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND In the last years, many new treatment options have widened the therapeutic scenario of genitourinary malignancies. Immunotherapy has shown efficacy, especially in the urothelial and renal cell carcinomas, with no particular relevance in prostate cancer. However, despite the use of immune checkpoint inhibitors, there is still high morbidity and mortality among these neoplasms. Cancer vaccines represent another way to activate the immune system. We sought to summarize the most recent advances in vaccine therapy for genitourinary malignancies with this review. METHODS We searched PubMed, Embase and Cochrane Database for clinical trials conducted in the last ten years, focusing on cancer vaccines in the prostate, urothelial and renal cancer. RESULTS Various therapeutic vaccines, including DNA-based, RNA-based, peptide-based, dendritic cells, viral vectors and modified tumor cells, have been demonstrated to induce specific immune responses in a variable percentage of patients. However, these responses rarely corresponded to significant survival improvements. CONCLUSIONS Further preclinical and clinical studies will improve the knowledge about cancer vaccines in genitourinary malignancies to optimize dosage, select targets with a driver role for tumor development and growth, and finally overcome resistance mechanisms. Combination strategies represent possibly more effective and long-lasting treatments.
Collapse
Affiliation(s)
- Brigida Anna Maiorano
- Oncology Unit, Foundation Casa Sollievo della Sofferenza IRCCS, 73013 San Giovanni Rotondo, Italy; (D.C.); (M.G.R.); (E.M.)
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy; (G.S.); (G.T.)
| | - Giovanni Schinzari
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy; (G.S.); (G.T.)
- Medical Oncology Unit, Comprehensive Cancer Center, Foundation A. Gemelli Policlinic IRCCS, 00168 Rome, Italy
| | - Davide Ciardiello
- Oncology Unit, Foundation Casa Sollievo della Sofferenza IRCCS, 73013 San Giovanni Rotondo, Italy; (D.C.); (M.G.R.); (E.M.)
- Medical Oncology, Department of Precision Medicine, Luigi Vanvitelli University of Campania, 80131 Naples, Italy
| | - Maria Grazia Rodriquenz
- Oncology Unit, Foundation Casa Sollievo della Sofferenza IRCCS, 73013 San Giovanni Rotondo, Italy; (D.C.); (M.G.R.); (E.M.)
| | - Antonio Cisternino
- Urology Unit, Foundation Casa Sollievo della Sofferenza IRCCS, 73013 San Giovanni Rotondo, Italy;
| | - Giampaolo Tortora
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy; (G.S.); (G.T.)
- Medical Oncology Unit, Comprehensive Cancer Center, Foundation A. Gemelli Policlinic IRCCS, 00168 Rome, Italy
| | - Evaristo Maiello
- Oncology Unit, Foundation Casa Sollievo della Sofferenza IRCCS, 73013 San Giovanni Rotondo, Italy; (D.C.); (M.G.R.); (E.M.)
| |
Collapse
|
40
|
Hussein YM, Hendawy DM, Alghamdy AN, Raafat N. Phenotypic and genetic evaluation of human monocyte-derived dendritic cells generated from whole blood for immunotherapy. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2021. [DOI: 10.1186/s43042-021-00168-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Dendritic cells (DCs) recognize different pathogens and cancer cells and activate the adaptive immune response. The generation of effective DC-based cancer vaccines depends on the appropriate differentiation of monocytes in vitro. This study aimed to standardize a protocol for the in vitro differentiation of human peripheral blood monocytes into immature DCs upon treatment with growth factors and generate monocyte-derived DCs (MoDCs). Peripheral blood mononuclear cells were separated from peripheral blood. After monocyte enrichment by plastic adhesion, monocytes were cultured for 6 days in the presence of granulocyte-macrophage colony-stimulating factor and interleukin-4 to generate immature DCs. The cells were examined by microscopy. Using flow cytometry, DCs were evaluated for the expression of the CD83 and HLA-DR surface antigens, for the uptake of fluorescein isothiocyanate conjugated dextran, and also for the expression of CD80 and CD86 mRNA.
Results
CD80 and CD86 genes expression was upregulated at day six and exhibited a significant difference (P < 0.05). DCs showed positive expression of the CD83 and HLA-DR surface antigens by flow cytometry and FITC-conjugated dextran uptake.
Conclusion
This study represents a preliminary trial to generate immature MoDCs in vitro from blood monocytes collected by the flask adherence method. It offers a panel of surface markers for DCs characterization and provides Immature DCs for experimental procedures after 6 incubation days.
Collapse
|
41
|
Rahimian N, Miraei HR, Amiri A, Ebrahimi MS, Nahand JS, Tarrahimofrad H, Hamblin MR, Khan H, Mirzaei H. Plant-based vaccines and cancer therapy: Where are we now and where are we going? Pharmacol Res 2021; 169:105655. [PMID: 34004270 DOI: 10.1016/j.phrs.2021.105655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 10/21/2022]
Abstract
Therapeutic vaccines are an effective approach in cancer therapy for treating the disease at later stages. The Food and Drug Administration (FDA) recently approved the first therapeutic cancer vaccine, and further studies are ongoing in clinical trials. These are expected to result in the future development of vaccines with relatively improved efficacy. Several vaccination approaches are being studied in pre-clinical and clinical trials, including the generation of anti-cancer vaccines by plant expression systems.This approach has advantages, such as high safety and low costs, especially for the synthesis of recombinant proteins. Nevertheless, the development of anti-cancer vaccines in plants is faced with some technical obstacles.Herein, we summarize some vaccines that have been used in cancer therapy, with an emphasis on plant-based vaccines.
Collapse
Affiliation(s)
- Neda Rahimian
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Hamid Reza Miraei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Atefeh Amiri
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashahd, Iran
| | | | - Javid Sadri Nahand
- Department of Virology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Tarrahimofrad
- Department of Animal Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 20282028, South Africa
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, 23200, Pakistan.
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
42
|
Yao L, Hu Q, Chen S, Zhou T, Yu X, Ma H, H. Ghonaim A, Wu H, Sun Q, Fan S, He Q. Recombinant Pseudorabies Virus with TK/gE Gene Deletion and Flt3L Co-Expression Enhances the Innate and Adaptive Immune Response via Activating Dendritic Cells. Viruses 2021; 13:v13040691. [PMID: 33923590 PMCID: PMC8072707 DOI: 10.3390/v13040691] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/08/2021] [Accepted: 04/09/2021] [Indexed: 12/23/2022] Open
Abstract
Owing to viral evolution and recombination, emerging pseudorabies virus (PRV) strains have caused unprecedented outbreaks in swine farms even when the pigs were previously vaccinated, which might indicate that traditional vaccines were unable to provide effective protection. The development of safe and efficacious vaccines presents prospects to minimize the clinical signs and eventually eradicate the infection. In this study, we used an emerging PRV strain, HNX, as the parental strain to construct a recombinant PRV with TK/gE gene deletion and Fms-related tyrosine kinase 3 ligand (Flt3L) expression, named HNX-TK−/gE−-Flt3L. HNX-TK−/gE−-Flt3L enhanced the maturation of bone marrow derived dendritic cells (DCs) in vitro. Significantly more activated DCs were detected in HNX-TK−/gE−-Flt3L-immunized mice compared with those immunized with HNX-TK−/gE−. Subsequently, a remarkable increase of neutralizing antibodies, gB-specific IgG antibodies, and interferon-gamma (IFN-γ) was observed in mice vaccinated with HNX-TK−/gE−-Flt3L. In addition, a lower mortality and less histopathological damage were observed in HNX-TK−/gE−-Flt3L vaccinated mice with upon PRV lethal challenge infection. Taken together, our results revealed the potential of Flt3L as an ideal adjuvant that can activate DCs and enhance protective immune responses and support the further evaluation of HNX-TK−/gE−-Flt3L as a promising PRV vaccine candidate.
Collapse
Affiliation(s)
- Lun Yao
- State Key Laboratory of Agricultural Microbiology College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (L.Y.); (Q.H.); (S.C.); (T.Z.); (X.Y.); (H.M.); (A.H.G.); (H.W.); (Q.S.); (S.F.)
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430000, China
| | - Qiao Hu
- State Key Laboratory of Agricultural Microbiology College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (L.Y.); (Q.H.); (S.C.); (T.Z.); (X.Y.); (H.M.); (A.H.G.); (H.W.); (Q.S.); (S.F.)
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430000, China
| | - Siqi Chen
- State Key Laboratory of Agricultural Microbiology College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (L.Y.); (Q.H.); (S.C.); (T.Z.); (X.Y.); (H.M.); (A.H.G.); (H.W.); (Q.S.); (S.F.)
| | - Tong Zhou
- State Key Laboratory of Agricultural Microbiology College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (L.Y.); (Q.H.); (S.C.); (T.Z.); (X.Y.); (H.M.); (A.H.G.); (H.W.); (Q.S.); (S.F.)
| | - Xuexiang Yu
- State Key Laboratory of Agricultural Microbiology College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (L.Y.); (Q.H.); (S.C.); (T.Z.); (X.Y.); (H.M.); (A.H.G.); (H.W.); (Q.S.); (S.F.)
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430000, China
| | - Hailong Ma
- State Key Laboratory of Agricultural Microbiology College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (L.Y.); (Q.H.); (S.C.); (T.Z.); (X.Y.); (H.M.); (A.H.G.); (H.W.); (Q.S.); (S.F.)
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430000, China
| | - Ahmed. H. Ghonaim
- State Key Laboratory of Agricultural Microbiology College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (L.Y.); (Q.H.); (S.C.); (T.Z.); (X.Y.); (H.M.); (A.H.G.); (H.W.); (Q.S.); (S.F.)
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430000, China
- Desert Research Center, Cairo 11435, Egypt
| | - Hao Wu
- State Key Laboratory of Agricultural Microbiology College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (L.Y.); (Q.H.); (S.C.); (T.Z.); (X.Y.); (H.M.); (A.H.G.); (H.W.); (Q.S.); (S.F.)
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430000, China
| | - Qi Sun
- State Key Laboratory of Agricultural Microbiology College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (L.Y.); (Q.H.); (S.C.); (T.Z.); (X.Y.); (H.M.); (A.H.G.); (H.W.); (Q.S.); (S.F.)
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430000, China
| | - Shengxian Fan
- State Key Laboratory of Agricultural Microbiology College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (L.Y.); (Q.H.); (S.C.); (T.Z.); (X.Y.); (H.M.); (A.H.G.); (H.W.); (Q.S.); (S.F.)
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430000, China
| | - Qigai He
- State Key Laboratory of Agricultural Microbiology College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; (L.Y.); (Q.H.); (S.C.); (T.Z.); (X.Y.); (H.M.); (A.H.G.); (H.W.); (Q.S.); (S.F.)
- The Cooperative Innovation Center for Sustainable Pig Production, Huazhong Agricultural University, Wuhan 430000, China
- Correspondence: ; Tel.: +86-27-8728-6974; Fax: +86-27-8728-7288
| |
Collapse
|
43
|
Xu X, Zhou Z, Li H, Fan Y. Towards customized cancer vaccines: a promising filed in personalized cancer medicine. Expert Rev Vaccines 2021; 20:545-557. [PMID: 33769185 DOI: 10.1080/14760584.2021.1909479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Cancer remains a major source of disease burden worldwide. Although cancer vaccines have been developed, most currently available cancer vaccines have limited therapeutic efficacy. Recent research using novel sequencing and bioinformatic tools has led scientists to realize that each tumor harbors a unique set of genetic mutations that can manifest as tumor-specific neoantigens. Therefore, it would be useful to develop personalized cancer vaccines that target neoantigens, which might improve the efficacy of these cancer treatments. AREAS COVERED This review covers cancer vaccine development and the emerging field of personalized cancer vaccines, with a discussion of future clinical trials for this promising treatment strategy. EXPERT OPINION Developing vaccines to treat tumors is one of the most promising and exciting fields in cancer research. However, cancer vaccines have shown limited efficacy in clinical trials for several decades, which may be related to the unique and complex processes underlying tumor development and progression. Recent studies have indicated that tumors express highly specific neoantigens, which are distinct from self-antigens. Thus, developing cancer vaccines that target these tumor-specific neoantigens is a promising strategy for developing personalized cancer vaccines.
Collapse
Affiliation(s)
- Xiaoling Xu
- Department of Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital),Hangzhou City, China.,Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences,Hangzhou City, China
| | - Zichao Zhou
- Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences,Hangzhou City, China.,Department of Thoracic Surgery, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou City, China
| | - Hui Li
- Department of Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital),Hangzhou City, China.,Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences,Hangzhou City, China
| | - Yun Fan
- Department of Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital),Hangzhou City, China.,Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences,Hangzhou City, China
| |
Collapse
|
44
|
Fu C, Tian G, Duan J, Liu K, Zhang C, Yan W, Wang Y. Therapeutic Antitumor Efficacy of Cancer Stem Cell-Derived DRibble Vaccine on Colorectal Carcinoma. Int J Med Sci 2021; 18:3249-3260. [PMID: 34400894 PMCID: PMC8364449 DOI: 10.7150/ijms.61510] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 07/12/2021] [Indexed: 12/13/2022] Open
Abstract
Dendritic cell (DC)-based immunotherapy has been a promising strategy for colon cancer therapy, but the efficacy of dendritic cell vaccines is in part limited by immunogenicity of loaded antigens. In this study, we aimed to identify a putative tumor antigen that can generate or enhance anti-tumor immune responses against colon cancer. CD44+ colon cancer stem cells (CCSCs) were isolated from mouse colorectal carcinoma CT-26 cell cultures and induced to form defective ribosomal products-containing autophagosome-rich blebs (DRibbles) by treatment with rapamycin, bortezomib, and ammonium chloride. DRibbles were characterized by western blot and transmission electron microscopy. DCs generated from the mice bone marrow monocytes were cocultured with DRibbles, then surface markers of DCs were analyzed by flow cytometry. Meanwhile, the efficacy of DRibble-DCs was examined in vivo. Our results showed that CCSC-derived DRibbles upregulated CD80, CD86, major histocompatibility complex (MHC)-I, and MHC-II on DCs and induced proliferation of mouse splenic lymphocytes and CD8+ T cells. In a model of colorectal carcinoma using BALB/c mice with robust tumor growth and mortality, DC vaccine pulsed with CCSC-derived DRibbles suppressed tumor growth and extended survival. A lactate dehydrogenase test indicated a strong cytolytic activity of cytotoxic T-cells derived from mice vaccinated with CCSC-derived DRibbles against CT-26 cells. Furthermore, flow cytometry analyses showed that the percentages of IFN-γ-producing CD8+ T-cells were increased in SD-DC group compare with the other groups. These findings provide a rationale for novel immunotherapeutic anti-tumor approaches based on DRibbles derived from colon cancer stem cells.
Collapse
Affiliation(s)
- Changhao Fu
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, China.,Stanford University Medical School, VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Geer Tian
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Jinyue Duan
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Kun Liu
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Chen Zhang
- Institute of Oceanography, Minjiang University, Fuzhou, Fujian 350108, China
| | - Weiqun Yan
- Medical Institute of Regeneration Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Yi Wang
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|
45
|
Jang GY, Kim YS, Lee SE, Lee JW, Han HD, Kang TH, Park YM. Improvement of DC-based vaccines using adjuvant TLR4-binding 60S acidic ribosomal protein P2 and immune checkpoint inhibitors. Cancer Immunol Immunother 2020; 70:1075-1088. [PMID: 33113002 DOI: 10.1007/s00262-020-02759-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 10/15/2020] [Indexed: 12/25/2022]
Abstract
Cancer immunotherapy has fewer side effects and higher efficiency than conventional methods. Dendritic cell (DC)-based vaccine, a cancer immunotherapeutic, is prepared by processing mature DCs and pulsing with tumor antigen peptide ex vivo, to induce the activation of tumor-specific T lymphocytes followed by tumor clearance in vivo. Unfortunately, clinical trials of this method mostly failed due to low patient response, possibly due to the absence of novel adjuvants that induce DC maturation through Toll-like receptor (TLR) signals. Interestingly, immune checkpoint inhibitor (ICI) therapy has shown remarkable anti-tumor efficacy when combined with cancer vaccines. In this study, we identified 60S acidic ribosomal protein P2 (RPLP2) through pull-down assay using human cancer cells derived proteins that binds to Toll-like receptor 4 (TLR4). Recombinant RPLP2 induced maturation and activation of DCs in vitro. This DC-based vaccine, followed by pulsing with tumor-specific antigen, has shown to significantly increase tumor-specific CD8+IFN-γ+ T cells, and improved both tumor prevention and tumor treatment effects in vivo. The adjuvant effects of RPLP2 were shown to be dependent on TLR4 using TLR4 knockout mice. Moreover, ICIs that suppress the tumor evasion mechanism showed synergistic effects on tumor treatment when combined with these vaccines.
Collapse
Affiliation(s)
- Gun-Young Jang
- Department of Immunology, College of Medicine, Konkuk University, 268 Chungwon-daero Chungju-si Chungcheongbuk-do 27478, Seoul, South Korea
| | - Young Seob Kim
- Department of Immunology, College of Medicine, Konkuk University, 268 Chungwon-daero Chungju-si Chungcheongbuk-do 27478, Seoul, South Korea
| | - Sung Eun Lee
- Department of Immunology, College of Medicine, Konkuk University, 268 Chungwon-daero Chungju-si Chungcheongbuk-do 27478, Seoul, South Korea
| | - Ji Won Lee
- Department of Immunology, College of Medicine, Konkuk University, 268 Chungwon-daero Chungju-si Chungcheongbuk-do 27478, Seoul, South Korea
| | - Hee Dong Han
- Department of Immunology, College of Medicine, Konkuk University, 268 Chungwon-daero Chungju-si Chungcheongbuk-do 27478, Seoul, South Korea
| | - Tae Heung Kang
- Department of Immunology, College of Medicine, Konkuk University, 268 Chungwon-daero Chungju-si Chungcheongbuk-do 27478, Seoul, South Korea.
| | - Yeong-Min Park
- Department of Immunology, College of Medicine, Konkuk University, 268 Chungwon-daero Chungju-si Chungcheongbuk-do 27478, Seoul, South Korea.
| |
Collapse
|
46
|
Moku G, Vangala S, Gulla SK, Yakati V. In vivo Targeting of DNA Vaccines to Dendritic Cells via the Mannose Receptor Induces Long-Lasting Immunity against Melanoma. Chembiochem 2020; 22:523-531. [PMID: 32909670 DOI: 10.1002/cbic.202000364] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/24/2020] [Indexed: 12/15/2022]
Abstract
Herein, we report effective, C-type lectin mannose receptor (MR)-selective, in vivo dendritic cell (DC)-targeting lipid nanoparticles (LNPs) of a novel lipid-containing mannose-mimicking di-shikimoyl- and guanidine head group and two n-hexadecyl hydrophobic tails (DSG). Subcutaneous administration of LNPs of the DSG/p-CMV-GFP complex showed a significant expression of green fluorescence protein in the CD11c+ DCs of the neighboring lymph nodes compared to the control LNPs of the BBG/p-CMV-GFP complex. Mannose receptor-facilitated in vivo DC-targeted vaccination (s.c.) with the electrostatic complex of LNPs of DSG/pCMV-MART1 stimulated long-lasting (270 days post B16F10 tumor challenge) antimelanoma immunity under prophylactic conditions. Remarkably, under therapeutic settings, vaccination (s.c.) with LNPs of the DSG/pCMV-MART1 complex significantly delayed melanoma growth and improved the survival of mice with melanoma. These findings demonstrate that this nonviral delivery system offers a resilient and potential approach to deliver DNA vaccines encoding tumor antigens to DCs in vivo with high efficacy.
Collapse
Affiliation(s)
- Gopikrishna Moku
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology (CSIR-IICT) Tarnaka, Uppal Road, Hyderabad, 500 007, India.,Present address: Department of Physical Sciences, Kakatiya Institute of Technology and Science, Yerragattu Gutta, Warangal, 506 015, Telangana, India
| | - Swathi Vangala
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology (CSIR-IICT) Tarnaka, Uppal Road, Hyderabad, 500 007, India.,Present address: Telangana Social Welfare Residential Degree College for Women, Bhupalapally 506 168, Telangana, India
| | - Suresh Kumar Gulla
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology (CSIR-IICT) Tarnaka, Uppal Road, Hyderabad, 500 007, India
| | - Venu Yakati
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology (CSIR-IICT) Tarnaka, Uppal Road, Hyderabad, 500 007, India
| |
Collapse
|
47
|
Chen Y, De Koker S, De Geest BG. Engineering Strategies for Lymph Node Targeted Immune Activation. Acc Chem Res 2020; 53:2055-2067. [PMID: 32910636 DOI: 10.1021/acs.accounts.0c00260] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Development of vaccine technology that induces long lasting and potent adaptive immune responses is of vital importance to combat emerging pathogens and to design the next generation of cancer immunotherapies. Advanced biomaterials such as nanoparticle carriers are intensively explored to increase the efficacy and safety of vaccines and immunotherapies, based on their intrinsic potential to focus the therapeutic payload onto the relevant immune cells and to limit systemic distribution. With adaptive immune responses being primarily initiated in lymph nodes, the potency of nanoparticle vaccines in turn is tightly linked to their capacity to reach and accumulate in the lymph nodes draining the immunization site. Here, we discuss the main strategies applied to increase nanoparticle delivery to lymph nodes: (1) direct lymph node injection, (2) active cell-mediated transport through targeting of peripheral dendritic cells, and (3) exploiting passive transport through the afferent lymphatics.The intralymph nodal injection is obviously the most direct way for nanoparticles to reach lymph nodes, and multiple studies have demonstrated its capability in enhancing immunostimulant drugs' immune activation and increasing the therapeutic window. However, the requirement of using ultrasound guidance for mapping lymph nodes in patients renders intranodal administration unsuited for mass vaccination campaigns. As lymph nodes are fine structured organs with lymphocytes and chemokine gradients arrayed in a highly ordered fashion, the breakdown of such formats by the intralymph nodal injection is another concern. The exploitation of dendritic cells as live vectors for transporting nanoparticles to lymph nodes has intensively been studied both ex vivo and in vivo. While ex vivo engineering of dendritic cells in theory can achieve 100% dendritic cell-specific selectivity, a scenario impossible to be achieved in vivo, this procedure is usually laborious and complicated and entails the participation of professional staff and equipment. In addition, the poor efficiency of dendritic cell migration to the draining lymph node is another significant limitation following the injection of ex vivo cultured dendritic cells. Thus, in vivo targeting of surface receptors, particularly C-type lectin receptors, on dendritic cells by conjugating nanoparticles with antibodies or ligands is intensively studied by both academia and industry. Although such nanoparticles in vivo still face nonspecific engulfment by various phagocytes, multiple studies have shown its feasibility in targeting dendritic cells with high selectivity. Moreover, through optimizing the physicochemical properties of nanoparticles, nanoparticles can passively drain to lymph nodes carried by the interstitial flow. Compared to dendritic cell-mediated transport, passive draining is much faster and of higher efficiency. Of all such properties, size is the most important parameter as large particles (>500 nm) can only reach lymph nodes by an active cell-mediated transport. Other surface properties, such as the charge and the balance of hydrophobicity-vs-hydrophilicity, strongly influence the mobility of nanoparticles in the extracellular space. In addition, albumin, a natural fatty acid transporter, has recently been demonstrated capable of binding the amphiphiles through their lipid moiety and subsequent transporting them to lymph nodes.
Collapse
Affiliation(s)
- Yong Chen
- Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghemt, Belgium
| | | | - Bruno G. De Geest
- Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghemt, Belgium
| |
Collapse
|
48
|
Chakravarty D, Huang L, Kahn M, Tewari AK. Immunotherapy for Metastatic Prostate Cancer: Current and Emerging Treatment Options. Urol Clin North Am 2020; 47:487-510. [PMID: 33008499 DOI: 10.1016/j.ucl.2020.07.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The advent of immunotherapy has revolutionized cancer treatment. Prostate cancer has an immunosuppressive microenvironment and a low tumor mutation burden, resulting in low neoantigen expression. The consensus was that immunotherapy would be less effective in prostate cancer. However, recent studies have reported that prostate cancer does have a high number of DNA damage and repair gene defects. Immunotherapies that have been tested in prostate cancer so far have been mainly vaccines and checkpoint inhibitors. A combination of genomically targeted therapies, with approaches to alleviate immune response and thereby make the tumor microenvironment immunologically hot, is promising.
Collapse
Affiliation(s)
- Dimple Chakravarty
- Department of Urology and the Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Li Huang
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, New York, USA; Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Matthew Kahn
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ashutosh K Tewari
- Department of Urology and the Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
49
|
Zhai J, Gao W, Zhao L, Lu C. Integrated transcriptomic and quantitative proteomic analysis identifies potential RNA sensors that respond to the Ag85A DNA vaccine. Microb Pathog 2020; 149:104487. [PMID: 32920150 DOI: 10.1016/j.micpath.2020.104487] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 09/04/2020] [Accepted: 09/05/2020] [Indexed: 01/25/2023]
Abstract
OBJECTIVE DNA vaccine has emerged as a promising approach with potential for Tuberculosis (TB) prevention in adults. However, the mechanism behind DNA vaccines is still largely unknown. MATERIALS AND METHODS Utilizing the CRISPR/Cas9 technique, we engineered Ag85A mutated dendritic cells (Ag85A-M-DCs) in which the Ag85A mRNA derived from Mycobacterium tuberculosis was expressed but not the corresponding protein. Control cells (Ag85A-DCs) expressed both Ag85A mRNA and protein. To better understand the mechanism of antigen presentation following DNA vaccination, integrated transcriptomic and proteomic analysis of dendritic cells (DCs), Ag85A-DCs, and Ag85A-M-DCs were performed. RESULTS A total of 723, 278, and 933 differentially expressed genes (DEGs), and 209, 134, and 509 differentially expressed proteins (DEPs) were identified between Ag85A-M-DCs and DCs, Ag85A-DCs and DCs, and Ag85A-M-DCs and Ag85A-DCs, respectively. Integration analysis detected 59, 15, and 64 associated DEGs/DEPs with the same expression trend between Ag85A-M-DCs and DCs, Ag85A-DCs and DCs, and Ag85A-M-DCs and Ag85A-DCs, respectively. KEGG pathway analysis showed that chemokine signaling pathway and MAPK signaling pathway were enriched in all three pairs of comparisons. The protein and protein interaction network revealed that ANXA1 was in the top 10 high-degree hub genes closely related to other genes in all three pairs of comparisons. CONCLUSION The results indicated that Ag85A DNA vaccine might transmit immunogenicity information and induce immune responses by activating chemokine signaling pathway and MAPK signaling pathway. ANXA1 may serve as a key target molecule of the Ag85A vaccine with additional potential for TB prevention.
Collapse
Affiliation(s)
- Jingbo Zhai
- Brucellosis Institute of Inner Mongolia University for the Nationalities, Tongliao, 028000, China; Department of Immunology, China Medical University, Shenyang, 110122, China; Brucellosis Prevention and Treatment Engineering Research Center of Inner Mongolia Autonomous Region, Tongliao, 028042, China
| | - Wei Gao
- Brucellosis Institute of Inner Mongolia University for the Nationalities, Tongliao, 028000, China
| | - Leheng Zhao
- Brucellosis Institute of Inner Mongolia University for the Nationalities, Tongliao, 028000, China
| | - Changlong Lu
- Brucellosis Institute of Inner Mongolia University for the Nationalities, Tongliao, 028000, China; Department of Immunology, China Medical University, Shenyang, 110122, China; Brucellosis Prevention and Treatment Engineering Research Center of Inner Mongolia Autonomous Region, Tongliao, 028042, China.
| |
Collapse
|
50
|
Cardiotoxic mechanisms of cancer immunotherapy - A systematic review. Int J Cardiol 2020; 323:179-187. [PMID: 32800915 DOI: 10.1016/j.ijcard.2020.08.033] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/26/2020] [Accepted: 08/07/2020] [Indexed: 02/08/2023]
Abstract
Cancer immunotherapy is a success story of translational medicine that has led to improved survival in patients with different difficult-to-treat types of cancer, such as metastasized melanoma, non-small cell lung cancer or renal cell carcinoma. These novel therapeutic agents exert their antitumor effects by activating the patients' immune system against cancer cells. Immunotherapy can be divided into active agents, such as anti-tumour vaccines or adoptive T-cell transfer, and passive immunotherapies like monoclonal antibodies, checkpoint inhibitors, cytokine therapy, bispecific T-cell engagers. After initial experimental use, broad clinical application revealed a number of important cardiovascular side effects of immunotherapeutics, which limit treatment options and decrease patients' prognosis and quality of life. With the rising rate of new immunotherapeutics at a hand, the number of patients receiving cancer immunotherapy will constantly increase, resulting in improved long-term survival rates. This review aims to summarize available cancer immunotherapies, their mechanism of action, currently known cardiovascular toxicities and their treatment. Further optimization of patient care will depend on the combined efforts by oncologists, cardiologists and cardiac surgeons to identify patients at risk and the implementation of interdisciplinary screening and treatment strategies. It is therefore crucial to familiarize heart specialists with novel cancer therapeutics and their potential adverse effects.
Collapse
|