1
|
Yang L, Wang Q, He L, Sun X. The critical role of tumor microbiome in cancer immunotherapy. Cancer Biol Ther 2024; 25:2301801. [PMID: 38241173 PMCID: PMC10802201 DOI: 10.1080/15384047.2024.2301801] [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: 10/24/2023] [Accepted: 01/01/2024] [Indexed: 01/21/2024] Open
Abstract
In recent years, the microbiome has shown an integral role in cancer immunotherapy and has become a prominent and widely studied topic. A full understanding of the interactions between the tumor microbiome and various immunotherapies offers opportunities for immunotherapy of cancer. This review scrutinizes the composition of the tumor microbiome, the mechanism of microbial immune regulation, the influence of tumor microorganisms on tumor metastasis, and the interaction between tumor microorganisms and immunotherapy. In addition, this review also summarizes the challenges and opportunities of immunotherapy through tumor microbes, as well as the prospects and directions for future related research. In conclusion, the potential of microbial immunotherapy to enhance treatment outcomes for cancer patients should not be underestimated. Through this review, it is hoped that more research on tumor microbial immunotherapy will be done to better solve the treatment problems of cancer patients.
Collapse
Affiliation(s)
- Liu Yang
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Qi Wang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Jiangsu University, Zhenjiang, China
| | - Lijuan He
- Department of Health Management Center, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Xingyu Sun
- Department of Gynecology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
| |
Collapse
|
2
|
Stilpeanu RI, Secara BS, Cretu-Stancu M, Bucur O. Oncolytic Viruses as Reliable Adjuvants in CAR-T Cell Therapy for Solid Tumors. Int J Mol Sci 2024; 25:11127. [PMID: 39456909 PMCID: PMC11508774 DOI: 10.3390/ijms252011127] [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: 08/18/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024] Open
Abstract
Although impactful scientific advancements have recently been made in cancer therapy, there remains an opportunity for future improvements. Immunotherapy is perhaps one of the most cutting-edge categories of therapies demonstrating potential in the clinical setting. Genetically engineered T cells express chimeric antigen receptors (CARs), which can detect signals expressed by the molecules present on the surface of cancer cells, also called tumor-associated antigens (TAAs). Their effectiveness has been extensively demonstrated in hematological cancers; therefore, these results can establish the groundwork for their applications on a wide range of requirements. However, the application of CAR-T cell technology for solid tumors has several challenges, such as the existence of an immune-suppressing tumor microenvironment and/or inadequate tumor infiltration. Consequently, combining therapies such as CAR-T cell technology with other approaches has been proposed. The effectiveness of combining CAR-T cell with oncolytic virus therapy, with either genetically altered or naturally occurring viruses, to target tumor cells is currently under investigation, with several clinical trials being conducted. This narrative review summarizes the current advancements, opportunities, benefits, and limitations in using each therapy alone and their combination. The use of oncolytic viruses offers an opportunity to address the existing challenges of CAR-T cell therapy, which appear in the process of trying to overcome solid tumors, through the combination of their strengths. Additionally, utilizing oncolytic viruses allows researchers to modify the virus, thus enabling the targeted delivery of specific therapeutic agents within the tumor environment. This, in turn, can potentially enhance the cytotoxic effect and therapeutic potential of CAR-T cell technology on solid malignancies, with impactful results in the clinical setting.
Collapse
MESH Headings
- Humans
- Neoplasms/therapy
- Neoplasms/immunology
- Oncolytic Viruses/genetics
- Oncolytic Viruses/immunology
- Immunotherapy, Adoptive/methods
- Oncolytic Virotherapy/methods
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Animals
- Tumor Microenvironment/immunology
- T-Lymphocytes/immunology
- Combined Modality Therapy/methods
- Adjuvants, Immunologic
- Antigens, Neoplasm/immunology
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
Collapse
Affiliation(s)
- Ruxandra Ilinca Stilpeanu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania (B.S.S.)
| | - Bianca Stefania Secara
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania (B.S.S.)
| | | | - Octavian Bucur
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania (B.S.S.)
- Genomics Research and Development Institute, 020021 Bucharest, Romania
- Viron Molecular Medicine Institute, Boston, MA 02108, USA
| |
Collapse
|
3
|
Hu Z, Li Y, Yang J, Liu J, Zhou H, Sun C, Tian C, Zhu C, Shao M, Wang S, Wei L, Liu M, Li S, Wang J, Xu H, Zhu W, Li X, Li J. Improved antitumor effectiveness of oncolytic HSV-1 viruses engineered with IL-15/IL-15Rα complex combined with oncolytic HSV-1-aPD1 targets colon cancer. Sci Rep 2024; 14:23671. [PMID: 39389985 PMCID: PMC11467195 DOI: 10.1038/s41598-024-72888-w] [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: 02/28/2024] [Accepted: 09/11/2024] [Indexed: 10/12/2024] Open
Abstract
Oncolytic virotherapy is emerging as a promising therapeutic avenue for cancer treatment, harnessing both innate and tumor-specific immune responses for targeted tumor elimination. In this study, we present a novel oncolytic virus (oHSV1-IL15B) derived from herpes simplex virus-1 (HSV-1), armed with IL-15/IL-15Rα complex, with a focus on treating colon cancer combined with oncolytic HSV-1 expressing anti-PD-1 antibody (oHSV1-aPD1). Results from our study reveal that recombinant oHSV-1 virus equipped with IL-15/IL-15Rα complex exhibited significant anti-tumor effects in a murine CT26 colon adenocarcinoma model. Notably, oHSV1-IL15B combined with oHSV-1-aPD1 demonstrates superior tumor inhibition and prolonged overall survival compared to oHSV1-mock and monotherapy groups. Further exploration highlights the impact of oHSV1-IL15B, oHSV-1-aPD1 and combined group on antitumor capacity, revealing a substantial increase in CD8+ T and CD4+ T cell proportions of CT26-bearing BALB/c mice and promoting apoptosis in tumor tissue. The study emphasizes the pivotal role of cytotoxic CD8+T cells in oncolytic virotherapy, demonstrating that recombinant oHSV1-IL15B combined with oncolytic HSV-1-aPD1 induces a robust tumor-specific T cell response. RNA sequence analysis highlighted oHSV1-IL15B combined with oHSV1-aPD1 improved tumors immune microenvironment on immune response, antiviral response-related genes and apoptosis-related genes, which contributed to anti-tumor immunotherapy. The findings underscore the promising antitumor activity achieved through the combination of IL-15/IL-15Rα complex and anti-PD-1 antibody with oHSV-1. This research opens avenues for diverse therapeutic strategies, suggesting the potential of synergistically utilizing cytokines and anti-PD-1 antibody with oncolytic viruses to enhance immunotherapy for cancer management.
Collapse
Affiliation(s)
- Zongfeng Hu
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Yixiao Li
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | | | - Jiajia Liu
- Beijing WellGene Company, Ltd, Beijing, 100085, China
| | - Hua Zhou
- Beijing WellGene Company, Ltd, Beijing, 100085, China
| | - Chunyang Sun
- Beijing WellGene Company, Ltd, Beijing, 100085, China
| | - Chao Tian
- Beijing WellGene Company, Ltd, Beijing, 100085, China
| | - Chengyang Zhu
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Mingxia Shao
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Shengrun Wang
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Lijun Wei
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Min Liu
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Shuzhen Li
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Jinyu Wang
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Haitian Xu
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Wei Zhu
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China
| | - Xiaopeng Li
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China.
- Beijing WellGene Company, Ltd, Beijing, 100085, China.
| | - Jingfeng Li
- School of Pharmacy, Yantai University, Yantai, 264005, Shandong, China.
| |
Collapse
|
4
|
Wang G, Mu M, Zhang Z, Chen Y, Yang N, Zhong K, Li Y, Lu F, Guo G, Tong A. Systemic delivery of tannic acid-ferric-masked oncolytic adenovirus reprograms tumor microenvironment for improved therapeutic efficacy in glioblastoma. Cancer Gene Ther 2024:10.1038/s41417-024-00839-8. [PMID: 39385009 DOI: 10.1038/s41417-024-00839-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/11/2024]
Abstract
Glioblastoma (GBM) represents the most aggressive primary brain tumor, and urgently requires effective treatments. Oncolytic adenovirus (OA) shows promise as a potential candidate for clinical antitumor therapy, including in the treatment of GBM. Nevertheless, the systemic delivery of OA continues to face challenges, leading to significantly compromised antitumor efficacy. In this study, we developed an innovative approach by encapsulating CXCL11-armed OA with tannic acid and Fe3+ (TA-Fe3+) to realize the systemic delivery of OA. The nanocarrier's ability to protect the OA from elimination by host immune response was evaluated in vitro and in vivo. We evaluated the antitumor effect and safety profile of OA@TA-Fe3+ in a GBM-bearing mice model. OA@TA-Fe3+ effectively safeguarded the virus from host immune clearance and extended its circulation in vivo. After targeting tumor sites, TA-Fe3+ could dissolve and release Fe3+ and OA. Fe3+-induced O2 production from H2O2 relieved the hypoxic state, and promoted OA replication, leading to a remarkable alteration of tumor immune microenvironment and enhancement in antitumor efficacy. Moreover, the systemic delivery of OA@TA-Fe3+ was safe without inflammation or organ damage. Our findings demonstrated the promising potential of systemically delivering the engineered OA for effective oncolytic virotherapy against GBM.
Collapse
Affiliation(s)
- Guoqing Wang
- Department of Ophthalmology, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, PR China
| | - Min Mu
- Laboratory of Liquid Biopsy and Single Cell Research, Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Zongliang Zhang
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Yongdong Chen
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Nian Yang
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Kunhong Zhong
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Yanfang Li
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Fang Lu
- Department of Ophthalmology, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, PR China.
| | - Gang Guo
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China.
| | - Aiping Tong
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, PR China.
| |
Collapse
|
5
|
Li Z, Qian W, Zhang Y, Liao C, Chen J, Ding K, Yu Q, Jia Y, He L. Enhanced Oncolytic Potential of Engineered Newcastle Disease Virus Lasota Strain through Modification of Its F Protein Cleavage Site. Microorganisms 2024; 12:2029. [PMID: 39458338 PMCID: PMC11510066 DOI: 10.3390/microorganisms12102029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/28/2024] Open
Abstract
Newcastle disease virus (NDV) is an oncolytic virus whose F protein cleavage activity is associated with viral infectivity. To explore the potential of modifying F protein cleavage activity to enhance antitumor effects, we constructed a recombinant NDV LaSota strain by replacing its F protein cleavage site with that from the mesogenic Beaudette C (BC) strain using reverse genetics techniques. The resulting virus, rLaSota-BC-RFP, demonstrated significantly enhanced infectivity and tumor cell suppression on the murine melanoma B16F10 cell, characterized by higher cytotoxicity and increased apoptosis compared to its parental strain, rLaSota-RFP. In vivo, rLaSota-BC-RFP treatment of B16F10 tumors in C57BL/6 mice resulted in significant tumor growth inhibition, improved survival rate, and induction of tumor-specific apoptosis and necrosis. Additionally, the rLaSota-BC-RFP treatment enhanced immunostimulatory effects within the tumor microenvironment (TME), characterized by increased infiltration of CD4+ and CD8+ T cells and elevated levels of antitumor immune modulator cytokines, including mouse IL-12, IFN-γ, IL-15, and TNF-α, in the rLaSota-BC-RFP-treated tumor tissues. Collectively, these findings demonstrate that the mesogenic F protein cleavage site enhances the oncolytic potential of the NDV LaSota strain, suggesting that rLaSota-BC-RFP is a promising oncolytic viral vector for gene delivery in cancer immunotherapy.
Collapse
Affiliation(s)
- Zedian Li
- The Key Lab of Animal Disease and Public Health/Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang 471023, China; (Z.L.); (W.Q.); (Y.Z.); (C.L.); (J.C.); (K.D.)
| | - Weifeng Qian
- The Key Lab of Animal Disease and Public Health/Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang 471023, China; (Z.L.); (W.Q.); (Y.Z.); (C.L.); (J.C.); (K.D.)
| | - Yuhao Zhang
- The Key Lab of Animal Disease and Public Health/Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang 471023, China; (Z.L.); (W.Q.); (Y.Z.); (C.L.); (J.C.); (K.D.)
| | - Chengshui Liao
- The Key Lab of Animal Disease and Public Health/Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang 471023, China; (Z.L.); (W.Q.); (Y.Z.); (C.L.); (J.C.); (K.D.)
| | - Jian Chen
- The Key Lab of Animal Disease and Public Health/Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang 471023, China; (Z.L.); (W.Q.); (Y.Z.); (C.L.); (J.C.); (K.D.)
| | - Ke Ding
- The Key Lab of Animal Disease and Public Health/Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang 471023, China; (Z.L.); (W.Q.); (Y.Z.); (C.L.); (J.C.); (K.D.)
| | - Qingzhong Yu
- Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, United States Department of Agriculture, Athens, GA 30605, USA;
| | - Yanyan Jia
- The Key Lab of Animal Disease and Public Health/Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang 471023, China; (Z.L.); (W.Q.); (Y.Z.); (C.L.); (J.C.); (K.D.)
| | - Lei He
- The Key Lab of Animal Disease and Public Health/Luoyang Key Laboratory of Live Carrier Biomaterial and Animal Disease Prevention and Control, Henan University of Science and Technology, Luoyang 471023, China; (Z.L.); (W.Q.); (Y.Z.); (C.L.); (J.C.); (K.D.)
- Southeast Poultry Research Laboratory, US National Poultry Research Center, Agricultural Research Service, United States Department of Agriculture, Athens, GA 30605, USA;
| |
Collapse
|
6
|
Khan M, Dong Y, Ullah R, Li M, Huang Q, Hu Y, Yang L, Luo Z. Recent Advances in Bacterium-Based Therapeutic Modalities for Melanoma Treatment. Adv Healthc Mater 2024:e2401076. [PMID: 39375965 DOI: 10.1002/adhm.202401076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/16/2024] [Indexed: 10/09/2024]
Abstract
Melanoma is one of the most severe skin cancer indications with rapid progression and a high risk of metastasis. However, despite the accumulated advances in melanoma treatment including adjuvant radiation, chemotherapy, and immunotherapy, the overall melanoma treatment efficacy in the clinics is still not satisfactory. Interestingly, bacterial therapeutics have demonstrated unique properties for tumor-related therapeutic applications, such as tumor-targeted motility, tailorable cytotoxicity, and immunomodulatory capacity of the tumor microenvironment, which have emerged as a promising platform for melanoma therapy. Indeed, the recent advances in genetic engineering and nanotechnologies have boosted the application potential of bacterium-based therapeutics for treating melanoma by further enhancing their tumor-homing, cell-killing, drug delivery, and immunostimulatory capacities. This review provides a comprehensive summary of the state-of-the-art bacterium-based anti-melanoma modalities, which are categorized according to their unique functional merits, including tumor-specific cytotoxins, tumor-targeted drug delivery platforms, and immune-stimulatory agents. Furthermore, a perspective is provided discussing the potential challenges and breakthroughs in this area. The insights in this review may facilitate the development of more advanced bacterium-based therapeutic modalities for improved melanoma treatment efficacy.
Collapse
Affiliation(s)
- Mubassir Khan
- Key Laboratory of Biorheological Science and Technology Ministry of Education College of Bioengineering Chongqing University Chongqing, Chongqing, 400044, P. R. China
| | - Yilong Dong
- Ruian People's Hospital, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325016, P. R. China
| | - Razi Ullah
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Lab for Vascular Implants College of Bioengineering Chongqing University, Chongqing, 400030, P. R. China
| | - Menghuan Li
- School of Life Science, Chongqing University, Chongqing, 400044, P. R. China
| | - Qiping Huang
- Key Laboratory of Biorheological Science and Technology Ministry of Education College of Bioengineering Chongqing University Chongqing, Chongqing, 400044, P. R. China
| | - Yan Hu
- Key Laboratory of Biorheological Science and Technology Ministry of Education College of Bioengineering Chongqing University Chongqing, Chongqing, 400044, P. R. China
| | - Li Yang
- Key Laboratory of Biorheological Science and Technology Ministry of Education College of Bioengineering Chongqing University Chongqing, Chongqing, 400044, P. R. China
| | - Zhong Luo
- School of Life Science, Chongqing University, Chongqing, 400044, P. R. China
| |
Collapse
|
7
|
Thoidingjam S, Bhatnagar AR, Sriramulu S, Siddiqui F, Nyati S. Optimizing Pancreatic Cancer Therapy: The Promise of Immune Stimulatory Oncolytic Viruses. Int J Mol Sci 2024; 25:9912. [PMID: 39337402 PMCID: PMC11432658 DOI: 10.3390/ijms25189912] [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: 08/27/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Pancreatic cancer presents formidable challenges due to rapid progression and resistance to conventional treatments. Oncolytic viruses (OVs) selectively infect cancer cells and cause cancer cells to lyse, releasing molecules that can be identified by the host's immune system. Moreover, OV can carry immune-stimulatory payloads such as interleukin-12, which when delivered locally can enhance immune system-mediated tumor killing. OVs are very well tolerated by cancer patients due to their ability to selectively target tumors without affecting surrounding normal tissues. OVs have recently been combined with other therapies, including chemotherapy and immunotherapy, to improve clinical outcomes. Several OVs including adenovirus, herpes simplex viruses (HSVs), vaccinia virus, parvovirus, reovirus, and measles virus have been evaluated in preclinical and clinical settings for the treatment of pancreatic cancer. We evaluated the safety and tolerability of a replication-competent oncolytic adenoviral vector carrying two suicide genes (thymidine kinase, TK; and cytosine deaminase, CD) and human interleukin-12 (hIL12) in metastatic pancreatic cancer patients in a phase 1 trial. This vector was found to be safe and well-tolerated at the highest doses tested without causing any significant adverse events (SAEs). Moreover, long-term follow-up studies indicated an increase in the overall survival (OS) in subjects receiving the highest dose of the OV. Our encouraging long-term survival data provide hope for patients with advanced pancreatic cancer, a disease that has not seen a meaningful increase in OS in the last five decades. In this review article, we highlight several preclinical and clinical studies and discuss future directions for optimizing OV therapy in pancreatic cancer. We envision OV-based gene therapy to be a game changer in the near future with the advent of newer generation OVs that have higher specificity and selectivity combined with personalized treatment plans developed under AI guidance.
Collapse
Affiliation(s)
| | | | | | - Farzan Siddiqui
- Department of Radiation Oncology, Henry Ford Health, Detroit, MI 48202, USA
- Henry Ford Health + Michigan State University Health Sciences, Detroit, MI 48202, USA
- Department of Medicine, Michigan State University, East Lansing, MI 48824, USA
| | - Shyam Nyati
- Department of Radiation Oncology, Henry Ford Health, Detroit, MI 48202, USA
- Henry Ford Health + Michigan State University Health Sciences, Detroit, MI 48202, USA
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
8
|
Haller SD, Essani K. Oncolytic Tanapoxvirus Variants Expressing mIL-2 and mCCL-2 Regress Human Pancreatic Cancer Xenografts in Nude Mice. Biomedicines 2024; 12:1834. [PMID: 39200298 PMCID: PMC11351728 DOI: 10.3390/biomedicines12081834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/01/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the fifth leading cause of cancer-related death and presents the lowest 5-year survival rate of any form of cancer in the US. Only 20% of PDAC patients are suitable for surgical resection and adjuvant chemotherapy, which remains the only curative treatment. Chemotherapeutic and gene therapy treatments are associated with adverse effects and lack specificity/efficacy. In this study, we assess the oncolytic potential of immuno-oncolytic tanapoxvirus (TPV) recombinants expressing mouse monocyte chemoattractant protein (mMCP-1 or mCCL2) and mouse interleukin (mIL)-2 in human pancreatic BxPc-3 cells using immunocompromised and CD-3+ T-cell-reconstituted mice. Intratumoral treatment with TPV/∆66R/mCCL2 and TPV/∆66R/mIL-2 resulted in a regression in BxPc-3 xenograft volume compared to control in immunocompromised mice; mCCL-2 expressing TPV OV resulted in a significant difference from control at p < 0.05. Histological analysis of immunocompromised mice treated with TPV/∆66R/mCCL2 or TPV/∆66R/mIL-2 demonstrated multiple biomarkers indicative of increased severity of chronic, active inflammation compared to controls. In conclusion, TPV recombinants expressing mCCL2 and mIL-2 demonstrated a therapeutic effect via regression in BxPc-3 tumor xenografts. Considering the enhanced oncolytic potency of TPV recombinants demonstrated against PDAC in this study, further investigation as an alternative or combination treatment option for human PDAC may be warranted.
Collapse
Affiliation(s)
| | - Karim Essani
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI 49008-5410, USA;
| |
Collapse
|
9
|
Brandenburg A, Heine A, Brossart P. Next-generation cancer vaccines and emerging immunotherapy combinations. Trends Cancer 2024; 10:749-769. [PMID: 39048489 DOI: 10.1016/j.trecan.2024.06.003] [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: 02/27/2024] [Revised: 06/06/2024] [Accepted: 06/12/2024] [Indexed: 07/27/2024]
Abstract
Therapeutic cancer vaccines have been a subject of research for several decades as potential new weapons to tackle malignancies. Their goal is to induce a long-lasting and efficient antitumour-directed immune response, capable of mediating tumour regression, preventing tumour progression, and eradicating minimal residual disease, while avoiding major adverse effects. Development of new vaccine technologies and antigen prediction methods has led to significant improvements in cancer vaccine efficacy. However, for their successful clinical application, certain obstacles still need to be overcome, especially tumour-mediated immunosuppression and escape mechanisms. In this review, we introduce therapeutic cancer vaccines and subsequently discuss combination approaches of next-generation cancer vaccines and existing immunotherapies, particularly immune checkpoint inhibitors (ICIs) and adoptive cell transfer/cell-based immunotherapies.
Collapse
Affiliation(s)
- Anne Brandenburg
- Medical Clinic III of Oncology, Hematology, Rheumatology and Immune-Oncology, University Hospital Bonn, Venusberg Campus 1, 53127 Bonn, Germany
| | - Annkristin Heine
- Medical Clinic III of Oncology, Hematology, Rheumatology and Immune-Oncology, University Hospital Bonn, Venusberg Campus 1, 53127 Bonn, Germany
| | - Peter Brossart
- Medical Clinic III of Oncology, Hematology, Rheumatology and Immune-Oncology, University Hospital Bonn, Venusberg Campus 1, 53127 Bonn, Germany.
| |
Collapse
|
10
|
Hodgins JJ, Abou-Hamad J, O’Dwyer CE, Hagerman A, Yakubovich E, Tanese de Souza C, Marotel M, Buchler A, Fadel S, Park MM, Fong-McMaster C, Crupi MF, Makinson OJ, Kurdieh R, Rezaei R, Dhillon HS, Ilkow CS, Bell JC, Harper ME, Rotstein BH, Auer RC, Vanderhyden BC, Sabourin LA, Bourgeois-Daigneault MC, Cook DP, Ardolino M. PD-L1 promotes oncolytic virus infection via a metabolic shift that inhibits the type I IFN pathway. J Exp Med 2024; 221:e20221721. [PMID: 38869480 PMCID: PMC11176258 DOI: 10.1084/jem.20221721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 02/04/2024] [Accepted: 03/14/2024] [Indexed: 06/14/2024] Open
Abstract
While conventional wisdom initially postulated that PD-L1 serves as the inert ligand for PD-1, an emerging body of literature suggests that PD-L1 has cell-intrinsic functions in immune and cancer cells. In line with these studies, here we show that engagement of PD-L1 via cellular ligands or agonistic antibodies, including those used in the clinic, potently inhibits the type I interferon pathway in cancer cells. Hampered type I interferon responses in PD-L1-expressing cancer cells resulted in enhanced efficacy of oncolytic viruses in vitro and in vivo. Consistently, PD-L1 expression marked tumor explants from cancer patients that were best infected by oncolytic viruses. Mechanistically, PD-L1 promoted a metabolic shift characterized by enhanced glycolysis rate that resulted in increased lactate production. In turn, lactate inhibited type I IFN responses. In addition to adding mechanistic insight into PD-L1 intrinsic function, our results will also help guide the numerous ongoing efforts to combine PD-L1 antibodies with oncolytic virotherapy in clinical trials.
Collapse
Affiliation(s)
- Jonathan J. Hodgins
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| | - John Abou-Hamad
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Colin Edward O’Dwyer
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| | - Ash Hagerman
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| | - Edward Yakubovich
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | | | - Marie Marotel
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| | - Ariel Buchler
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Canada
- University of Ottawa Heart Institute, Ottawa, Canada
| | - Saleh Fadel
- The Ottawa Hospital, Ottawa, Canada
- Department of Pathology and Laboratory Medicine, The Ottawa Hospital, Ottawa, Canada
| | - Maria M. Park
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| | - Claire Fong-McMaster
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Ottawa Institute for Systems Biology, Ottawa, Canada
| | - Mathieu F. Crupi
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Olivia Joan Makinson
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| | - Reem Kurdieh
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Reza Rezaei
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| | - Harkirat Singh Dhillon
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| | - Carolina S. Ilkow
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| | - John C. Bell
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
| | - Mary-Ellen Harper
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
- Ottawa Institute for Systems Biology, Ottawa, Canada
| | - Benjamin H. Rotstein
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Canada
- University of Ottawa Heart Institute, Ottawa, Canada
| | - Rebecca C. Auer
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
| | - Barbara C. Vanderhyden
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Luc A. Sabourin
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Marie-Claude Bourgeois-Daigneault
- Department of Microbiology, Infectious Diseases, and Immunology, University of Montreal, Montreal, Canada
- Centre Hospitalier de l’Université de Montréal Research Centre, Cancer and Immunopathology axes, Montreal, Canada
| | - David P. Cook
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Canada
| | - Michele Ardolino
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Canada
- Center for Infection, Immunity, and Inflammation, University of Ottawa, Ottawa, Canada
| |
Collapse
|
11
|
Taheri M, Tehrani HA, Dehghani S, Alibolandi M, Arefian E, Ramezani M. Nanotechnology and bioengineering approaches to improve the potency of mesenchymal stem cell as an off-the-shelf versatile tumor delivery vehicle. Med Res Rev 2024; 44:1596-1661. [PMID: 38299924 DOI: 10.1002/med.22023] [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: 12/08/2022] [Revised: 11/28/2023] [Accepted: 01/10/2024] [Indexed: 02/02/2024]
Abstract
Targeting actionable mutations in oncogene-driven cancers and the evolution of immuno-oncology are the two prominent revolutions that have influenced cancer treatment paradigms and caused the emergence of precision oncology. However, intertumoral and intratumoral heterogeneity are the main challenges in both fields of precision cancer treatment. In other words, finding a universal marker or pathway in patients suffering from a particular type of cancer is challenging. Therefore, targeting a single hallmark or pathway with a single targeted therapeutic will not be efficient for fighting against tumor heterogeneity. Mesenchymal stem cells (MSCs) possess favorable characteristics for cellular therapy, including their hypoimmune nature, inherent tumor-tropism property, straightforward isolation, and multilineage differentiation potential. MSCs can be loaded with various chemotherapeutics and oncolytic viruses. The combination of these intrinsic features with the possibility of genetic manipulation makes them a versatile tumor delivery vehicle that can be used for in vivo selective tumor delivery of various chemotherapeutic and biological therapeutics. MSCs can be used as biofactory for the local production of chemical or biological anticancer agents at the tumor site. MSC-mediated immunotherapy could facilitate the sustained release of immunotherapeutic agents specifically at the tumor site, and allow for the achievement of therapeutic concentrations without the need for repetitive systemic administration of high therapeutic doses. Despite the enthusiasm evoked by preclinical studies that used MSC in various cancer therapy approaches, the translation of MSCs into clinical applications has faced serious challenges. This manuscript, with a critical viewpoint, reviewed the preclinical and clinical studies that have evaluated MSCs as a selective tumor delivery tool in various cancer therapy approaches, including gene therapy, immunotherapy, and chemotherapy. Then, the novel nanotechnology and bioengineering approaches that can improve the potency of MSC for tumor targeting and overcoming challenges related to their low localization at the tumor sites are discussed.
Collapse
Affiliation(s)
- Mojtaba Taheri
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Abdul Tehrani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sadegh Dehghani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ehsan Arefian
- Department of Microbiology, School of Biology, College of Science, University of Tehran, Tehran, Iran
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
12
|
Nelson A, McMullen N, Gebremeskel S, De Antueno R, Mackenzie D, Duncan R, Johnston B. Fusogenic vesicular stomatitis virus combined with natural killer T cell immunotherapy controls metastatic breast cancer. Breast Cancer Res 2024; 26:78. [PMID: 38750591 PMCID: PMC11094881 DOI: 10.1186/s13058-024-01818-5] [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: 12/06/2023] [Accepted: 03/30/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND Metastatic breast cancer is a leading cause of cancer death in woman. Current treatment options are often associated with adverse side effects and poor outcomes, demonstrating the need for effective new treatments. Immunotherapies can provide durable outcomes in many cancers; however, limited success has been achieved in metastatic triple negative breast cancer. We tested whether combining different immunotherapies can target metastatic triple negative breast cancer in pre-clinical models. METHODS Using primary and metastatic 4T1 triple negative mammary carcinoma models, we examined the therapeutic effects of oncolytic vesicular stomatitis virus (VSVΔM51) engineered to express reovirus-derived fusion associated small transmembrane proteins p14 (VSV-p14) or p15 (VSV-p15). These viruses were delivered alone or in combination with natural killer T (NKT) cell activation therapy mediated by adoptive transfer of α-galactosylceramide-loaded dendritic cells. RESULTS Treatment of primary 4T1 tumors with VSV-p14 or VSV-p15 alone increased immunogenic tumor cell death, attenuated tumor growth, and enhanced immune cell infiltration and activation compared to control oncolytic virus (VSV-GFP) treatments and untreated mice. When combined with NKT cell activation therapy, oncolytic VSV-p14 and VSV-p15 reduced metastatic lung burden to undetectable levels in all mice and generated immune memory as evidenced by enhanced in vitro recall responses (tumor killing and cytokine production) and impaired tumor growth upon rechallenge. CONCLUSION Combining NKT cell immunotherapy with enhanced oncolytic virotherapy increased anti-tumor immune targeting of lung metastasis and presents a promising treatment strategy for metastatic breast cancer.
Collapse
Affiliation(s)
- Adam Nelson
- Department of Microbiology and Immunology, Dalhousie University, B3H 4R2, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, B3H 4R2, Halifax, NS, Canada
| | - Nichole McMullen
- Department of Microbiology and Immunology, Dalhousie University, B3H 4R2, Halifax, NS, Canada
| | - Simon Gebremeskel
- Department of Microbiology and Immunology, Dalhousie University, B3H 4R2, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, B3H 4R2, Halifax, NS, Canada
| | - Roberto De Antueno
- Department of Microbiology and Immunology, Dalhousie University, B3H 4R2, Halifax, NS, Canada
| | - Duncan Mackenzie
- Department of Microbiology and Immunology, Dalhousie University, B3H 4R2, Halifax, NS, Canada
| | - Roy Duncan
- Department of Microbiology and Immunology, Dalhousie University, B3H 4R2, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, B3H 4R2, Halifax, NS, Canada
- Department of Biochemistry and Molecular Biology, Dalhousie University, B3H 4R2, Halifax, NS, Canada
- Department of Pediatrics, Dalhousie University, B3H 4R2, Halifax, NS, Canada
| | - Brent Johnston
- Department of Microbiology and Immunology, Dalhousie University, B3H 4R2, Halifax, NS, Canada.
- Beatrice Hunter Cancer Research Institute, B3H 4R2, Halifax, NS, Canada.
- Department of Pathology, Dalhousie University, B3H 4R2, Halifax, NS, Canada.
| |
Collapse
|
13
|
Kurmasheva N, Said A, Wong B, Kinderman P, Han X, Rahimic AHF, Kress A, Carter-Timofte ME, Holm E, van der Horst D, Kollmann CF, Liu Z, Wang C, Hoang HD, Kovalenko E, Chrysopoulou M, Twayana KS, Ottosen RN, Svenningsen EB, Begnini F, Kiib AE, Kromm FEH, Weiss HJ, Di Carlo D, Muscolini M, Higgins M, van der Heijden M, Bardoul A, Tong T, Ozsvar A, Hou WH, Schack VR, Holm CK, Zheng Y, Ruzek M, Kalucka J, de la Vega L, Elgaher WAM, Korshoej AR, Lin R, Hiscott J, Poulsen TB, O'Neill LA, Roy DG, Rinschen MM, van Montfoort N, Diallo JS, Farin HF, Alain T, Olagnier D. Octyl itaconate enhances VSVΔ51 oncolytic virotherapy by multitarget inhibition of antiviral and inflammatory pathways. Nat Commun 2024; 15:4096. [PMID: 38750019 PMCID: PMC11096414 DOI: 10.1038/s41467-024-48422-x] [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: 06/07/2023] [Accepted: 04/23/2024] [Indexed: 05/18/2024] Open
Abstract
The presence of heterogeneity in responses to oncolytic virotherapy poses a barrier to clinical effectiveness, as resistance to this treatment can occur through the inhibition of viral spread within the tumor, potentially leading to treatment failures. Here we show that 4-octyl itaconate (4-OI), a chemical derivative of the Krebs cycle-derived metabolite itaconate, enhances oncolytic virotherapy with VSVΔ51 in various models including human and murine resistant cancer cell lines, three-dimensional (3D) patient-derived colon tumoroids and organotypic brain tumor slices. Furthermore, 4-OI in combination with VSVΔ51 improves therapeutic outcomes in a resistant murine colon tumor model. Mechanistically, we find that 4-OI suppresses antiviral immunity in cancer cells through the modification of cysteine residues in MAVS and IKKβ independently of the NRF2/KEAP1 axis. We propose that the combination of a metabolite-derived drug with an oncolytic virus agent can greatly improve anticancer therapeutic outcomes by direct interference with the type I IFN and NF-κB-mediated antiviral responses.
Collapse
Affiliation(s)
- Naziia Kurmasheva
- Department of Biomedicine, Aarhus University, 8000, Aarhus C, Denmark
| | - Aida Said
- Department of Biochemistry Microbiology and Immunology, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, K1H 8L1, Canada
| | - Boaz Wong
- Department of Biochemistry Microbiology and Immunology, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
- Ottawa Hospital Research Insitute, Ottawa, ON, K1H 8L6, Canada
| | - Priscilla Kinderman
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Xiaoying Han
- Lady Davis Institute, Jewish General Hospital and Department of Medicine, McGill University, Montreal, QC, H3T 1E2, Canada
| | - Anna H F Rahimic
- Department of Biomedicine, Aarhus University, 8000, Aarhus C, Denmark
| | - Alena Kress
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
- Faculty of Biological Sciences, Goethe University, 60438, Frankfurt am Main, Germany
| | | | - Emilia Holm
- Department of Biomedicine, Aarhus University, 8000, Aarhus C, Denmark
| | | | | | - Zhenlong Liu
- Lady Davis Institute, Jewish General Hospital and Department of Medicine, McGill University, Montreal, QC, H3T 1E2, Canada
| | - Chen Wang
- Lady Davis Institute, Jewish General Hospital and Department of Medicine, McGill University, Montreal, QC, H3T 1E2, Canada
| | - Huy-Dung Hoang
- Department of Biochemistry Microbiology and Immunology, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, K1H 8L1, Canada
| | - Elina Kovalenko
- Department of Biomedicine, Aarhus University, 8000, Aarhus C, Denmark
| | | | | | - Rasmus N Ottosen
- Department of Chemistry, Aarhus University, 8000, Aarhus C, Denmark
| | | | - Fabio Begnini
- Department of Chemistry, Aarhus University, 8000, Aarhus C, Denmark
| | - Anders E Kiib
- Department of Chemistry, Aarhus University, 8000, Aarhus C, Denmark
| | | | - Hauke J Weiss
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin 2, Ireland
| | - Daniele Di Carlo
- Pasteur Laboratories, Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, 00161, Italy
| | - Michela Muscolini
- Pasteur Laboratories, Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, 00161, Italy
| | - Maureen Higgins
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Mirte van der Heijden
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Angelina Bardoul
- Cancer Axis, CHUM Research Centre, Montreal, Canada
- Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, University of Montreal, Montreal, Canada
- Institut du Cancer de Montréal, Montreal, QC, Canada
| | - Tong Tong
- Department of Neurosurgery, Aarhus University Hospital, 8200, Aarhus N, Denmark
- Department of Clinical Medicine, Aarhus University, 8200, Aarhus N, Denmark
- DCCC Brain Tumor Center, Copenhagen University Hospital, Copenhagen, Denmark
| | - Attila Ozsvar
- Department of Biomedicine, Aarhus University, 8000, Aarhus C, Denmark
- Department of Clinical Medicine, Aarhus University, 8200, Aarhus N, Denmark
| | - Wen-Hsien Hou
- Department of Biomedicine, Aarhus University, 8000, Aarhus C, Denmark
| | - Vivien R Schack
- Department of Biomedicine, Aarhus University, 8000, Aarhus C, Denmark
| | - Christian K Holm
- Department of Biomedicine, Aarhus University, 8000, Aarhus C, Denmark
| | - Yunan Zheng
- Small Molecule Therapeutics & Platform Technologies, AbbVie Inc., 1 North Waukegon Road, North Chicago, IL, 60064, USA
| | - Melanie Ruzek
- AbbVie, Bioresearch Center, 100 Research Drive, Worcester, MA, 01608, USA
| | - Joanna Kalucka
- Department of Biomedicine, Aarhus University, 8000, Aarhus C, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Laureano de la Vega
- Jacqui Wood Cancer Centre, Division of Cellular Medicine, School of Medicine, University of Dundee, Dundee, UK
| | - Walid A M Elgaher
- Department of Drug Design and Optimization, Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, E8.1, 66123, Saarbrücken, Germany
| | - Anders R Korshoej
- Department of Neurosurgery, Aarhus University Hospital, 8200, Aarhus N, Denmark
- Department of Clinical Medicine, Aarhus University, 8200, Aarhus N, Denmark
- DCCC Brain Tumor Center, Copenhagen University Hospital, Copenhagen, Denmark
| | - Rongtuan Lin
- Lady Davis Institute, Jewish General Hospital and Department of Medicine, McGill University, Montreal, QC, H3T 1E2, Canada
| | - John Hiscott
- Pasteur Laboratories, Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, 00161, Italy
| | - Thomas B Poulsen
- Department of Chemistry, Aarhus University, 8000, Aarhus C, Denmark
| | - Luke A O'Neill
- School of Biochemistry and Immunology, Trinity College Dublin, Trinity Biomedical Sciences Institute, Dublin 2, Ireland
| | - Dominic G Roy
- Cancer Axis, CHUM Research Centre, Montreal, Canada
- Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, University of Montreal, Montreal, Canada
- Institut du Cancer de Montréal, Montreal, QC, Canada
| | - Markus M Rinschen
- Department of Biomedicine, Aarhus University, 8000, Aarhus C, Denmark
- III. Department of Medicine and Hamburg Center for Kidney Health, Hamburg, Germany
- Aarhus Institute of Advanced Studies, Aarhus University, Aarhus, Denmark
| | - Nadine van Montfoort
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jean-Simon Diallo
- Department of Biochemistry Microbiology and Immunology, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
- Ottawa Hospital Research Insitute, Ottawa, ON, K1H 8L6, Canada
| | - Henner F Farin
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Goethe University, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Frankfurt/Mainz partner site and German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Tommy Alain
- Department of Biochemistry Microbiology and Immunology, University of Ottawa, Ottawa, ON, K1H 8M5, Canada
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, K1H 8L1, Canada
| | - David Olagnier
- Department of Biomedicine, Aarhus University, 8000, Aarhus C, Denmark.
| |
Collapse
|
14
|
Vazaios K, van Berkum RE, Calkoen FG, van der Lugt J, Hulleman E. OV Modulators of the Paediatric Brain TIME: Current Status, Combination Strategies, Limitations and Future Directions. Int J Mol Sci 2024; 25:5007. [PMID: 38732225 PMCID: PMC11084613 DOI: 10.3390/ijms25095007] [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: 03/12/2024] [Revised: 04/26/2024] [Accepted: 05/01/2024] [Indexed: 05/13/2024] Open
Abstract
Oncolytic viruses (OVs) are characterised by their preference for infecting and replicating in tumour cells either naturally or after genetic modification, resulting in oncolysis. Furthermore, OVs can elicit both local and systemic anticancer immune responses while specifically infecting and lysing tumour cells. These characteristics render them a promising therapeutic approach for paediatric brain tumours (PBTs). PBTs are frequently marked by a cold tumour immune microenvironment (TIME), which suppresses immunotherapies. Recent preclinical and clinical studies have demonstrated the capability of OVs to induce a proinflammatory immune response, thereby modifying the TIME. In-depth insights into the effect of OVs on different cell types in the TIME may therefore provide a compelling basis for using OVs in combination with other immunotherapy modalities. However, certain limitations persist in our understanding of oncolytic viruses' ability to regulate the TIME to enhance anti-tumour activity. These limitations primarily stem from the translational limitations of model systems, the difficulties associated with tracking reliable markers of efficacy throughout the course of treatment and the role of pre-existing viral immunity. In this review, we describe the different alterations observed in the TIME in PBTs due to OV treatment, combination therapies of OVs with different immunotherapies and the hurdles limiting the development of effective OV therapies while suggesting future directions based on existing evidence.
Collapse
Affiliation(s)
| | | | | | | | - Esther Hulleman
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (K.V.); (F.G.C.); (J.v.d.L.)
| |
Collapse
|
15
|
Iyer M, Ravichandran N, Karuppusamy PA, Gnanarajan R, Yadav MK, Narayanasamy A, Vellingiri B. Molecular insights and promise of oncolytic virus based immunotherapy. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 140:419-492. [PMID: 38762277 DOI: 10.1016/bs.apcsb.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2024]
Abstract
Discovering a therapeutic that can counteract the aggressiveness of this disease's mechanism is crucial for improving survival rates for cancer patients and for better understanding the most different types of cancer. In recent years, using these viruses as an anticancer therapy has been thought to be successful. They mostly work by directly destroying cancer cells, activating the immune system to fight cancer, and expressing exogenous effector genes. For the treatment of tumors, oncolytic viruses (OVs), which can be modified to reproduce only in tumor tissues and lyse them while preserving the healthy non-neoplastic host cells and reinstating antitumor immunity which present a novel immunotherapeutic strategy. OVs can exist naturally or be created in a lab by altering existing viruses. These changes heralded the beginning of a new era of less harmful virus-based cancer therapy. We discuss three different types of oncolytic viruses that have already received regulatory approval to treat cancer as well as clinical research using oncolytic adenoviruses. The primary therapeutic applications, mechanism of action of oncolytic virus updates, future views of this therapy will be covered in this chapter.
Collapse
Affiliation(s)
- Mahalaxmi Iyer
- Department of Microbiology, Central University of Punjab, Bathinda, India
| | - Nandita Ravichandran
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | | | - Roselin Gnanarajan
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Mukesh Kumar Yadav
- Department of Microbiology, Central University of Punjab, Bathinda, India
| | - Arul Narayanasamy
- Disease Proteomics Laboratory, Department of Zoology, Bharathiar University, Coimbatore, Tamil Nadu, India.
| | - Balachandar Vellingiri
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India.
| |
Collapse
|
16
|
Chen Y, Chen X, Bao W, Liu G, Wei W, Ping Y. An oncolytic virus-T cell chimera for cancer immunotherapy. Nat Biotechnol 2024:10.1038/s41587-023-02118-7. [PMID: 38336902 DOI: 10.1038/s41587-023-02118-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 12/21/2023] [Indexed: 02/12/2024]
Abstract
The efficacy of oncolytic adenoviruses (OAs) for cancer therapy has been limited by insufficient delivery to tumors after systemic injection and the propensity of OAs to induce the expression of immune checkpoints. To address these limitations, we use T cells to deliver OAs into tumors and engineer the OA to express a Cas9 system targeting the PDL1 gene encoding the immune checkpoint protein PD-L1. By cloaking OAs with cell membranes presenting T cell-specific antigens, we physically conjugated OAs onto T cell surfaces by antigen-receptor interaction. We tested the oncolytic virus-T cell chimera (ONCOTECH) via intravenous delivery in mouse cancer models, including models of melanoma, pancreatic adenocarcinoma, lung cancer and glioblastoma. In the melanoma model, the in vivo delivery of ONCOTECH resulted in a strong accumulation of OAs in tumor cells, where PD-L1 expression was reduced by 50% and the single administration of ONCOTECH enabled 80% survival over 70 days. Collectively, ONCOTECH represents a promising translational technology to combine virotherapy and cell therapy.
Collapse
Affiliation(s)
- Yuxuan Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China
| | - Xiaohong Chen
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
| | - Weier Bao
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, China
| | - Wei Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Yuan Ping
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China.
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, China.
| |
Collapse
|
17
|
Volovat SR, Scripcariu DV, Vasilache IA, Stolniceanu CR, Volovat C, Augustin IG, Volovat CC, Ostafe MR, Andreea-Voichița SG, Bejusca-Vieriu T, Lungulescu CV, Sur D, Boboc D. Oncolytic Virotherapy: A New Paradigm in Cancer Immunotherapy. Int J Mol Sci 2024; 25:1180. [PMID: 38256250 PMCID: PMC10816814 DOI: 10.3390/ijms25021180] [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: 11/19/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 01/24/2024] Open
Abstract
Oncolytic viruses (OVs) are emerging as potential treatment options for cancer. Natural and genetically engineered viruses exhibit various antitumor mechanisms. OVs act by direct cytolysis, the potentiation of the immune system through antigen release, and the activation of inflammatory responses or indirectly by interference with different types of elements in the tumor microenvironment, modification of energy metabolism in tumor cells, and antiangiogenic action. The action of OVs is pleiotropic, and they show varied interactions with the host and tumor cells. An important impediment in oncolytic virotherapy is the journey of the virus into the tumor cells and the possibility of its binding to different biological and nonbiological vectors. OVs have been demonstrated to eliminate cancer cells that are resistant to standard treatments in many clinical trials for various cancers (melanoma, lung, and hepatic); however, there are several elements of resistance to the action of viruses per se. Therefore, it is necessary to evaluate the combination of OVs with other standard treatment modalities, such as chemotherapy, immunotherapy, targeted therapies, and cellular therapies, to increase the response rate. This review provides a comprehensive update on OVs, their use in oncolytic virotherapy, and the future prospects of this therapy alongside the standard therapies currently used in cancer treatment.
Collapse
Affiliation(s)
- Simona Ruxandra Volovat
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania; (S.R.V.); (M.-R.O.); (S.-G.A.-V.); (T.B.-V.)
| | - Dragos Viorel Scripcariu
- Department of Surgery, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania;
| | - Ingrid Andrada Vasilache
- Department of Obstetrics and Gynecology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Cati Raluca Stolniceanu
- Department of Biophysics and Medical Physics—Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania;
| | - Constantin Volovat
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania; (S.R.V.); (M.-R.O.); (S.-G.A.-V.); (T.B.-V.)
| | | | | | - Madalina-Raluca Ostafe
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania; (S.R.V.); (M.-R.O.); (S.-G.A.-V.); (T.B.-V.)
| | - Slevoacă-Grigore Andreea-Voichița
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania; (S.R.V.); (M.-R.O.); (S.-G.A.-V.); (T.B.-V.)
| | - Toni Bejusca-Vieriu
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania; (S.R.V.); (M.-R.O.); (S.-G.A.-V.); (T.B.-V.)
| | | | - Daniel Sur
- 11th Department of Medical Oncology, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania;
| | - Diana Boboc
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str., 700115 Iasi, Romania; (S.R.V.); (M.-R.O.); (S.-G.A.-V.); (T.B.-V.)
| |
Collapse
|
18
|
Alwithenani A, Taha Z, Thomson M, Chen A, Wong B, Arulanandam R, Diallo JS. Unlocking the potential of dimethyl fumarate: enhancing oncolytic HSV-1 efficacy for wider cancer applications. Front Immunol 2023; 14:1332929. [PMID: 38169670 PMCID: PMC10758402 DOI: 10.3389/fimmu.2023.1332929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024] Open
Abstract
Immunotherapy and specifically oncolytic virotherapy has emerged as a promising option for cancer patients, with oncolytic herpes simplex virus-1 (oHSV-1) expressing granulocyte macrophage colony stimulating factor being the first OV to be approved by the FDA for treatment of melanoma. However, not all cancers are sensitive and responsive to oncolytic viruses (OVs). Our group has demonstrated that fumaric and maleic acid esters (FMAEs) are very effective in sensitizing cancer cells to OV infection. Of note, these FMAEs include dimethyl fumarate (DMF, also known as Tecfidera®), an approved treatment for multiple sclerosis and psoriasis. This study aimed to assess the efficacy of DMF in combination with oncolytic HSV-1 in preclinical cancer models. We demonstrate herewith that pre-treatment with DMF or other FMAEs leads to a significant increase in viral growth of oHSV-1 in several cancer cell lines, including melanoma, while decreasing cell viability. Additionally, DMF was able to enhance ex vivo oHSV-1 infection of mouse-derived tumor cores as well as human patient tumor samples but not normal tissue. We further reveal that the increased viral spread and oncolysis of the combination therapy occurs via inhibition of type I IFN production and response. Finally, we demonstrate that DMF in combination with oHSV-1 can improve therapeutic outcomes in aggressive syngeneic murine cancer models. In sum, this study demonstrates the synergistic potential of two approved therapies for clinical evaluation in cancer patients.
Collapse
Affiliation(s)
- Akram Alwithenani
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, ON, Canada
- Department of Clinical Laboratory Science, Faculty of Applied Medical Science, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Zaid Taha
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, ON, Canada
| | - Max Thomson
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Andrew Chen
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Boaz Wong
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, ON, Canada
| | - Rozanne Arulanandam
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Jean-Simon Diallo
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, ON, Canada
| |
Collapse
|
19
|
Zhao Z, Wang W, Wang G, Huang Z, Zhou L, Lin L, Ou Y, Huang W, Zhang X, Wu C, Tao L, Wang Q. Dual peptides-modified cationic liposomes for enhanced Lung cancer gene therapy by a gap junction regulating strategy. J Nanobiotechnology 2023; 21:473. [PMID: 38066528 PMCID: PMC10709977 DOI: 10.1186/s12951-023-02242-1] [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: 07/06/2023] [Accepted: 12/03/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Gene therapy for lung cancer has emerged as a novel tumor-combating strategy for its superior tumor specificity, low systematical toxicity and huge clinical translation potential. Especially, the applications of microRNA shed led on effective tumor ablation by directly interfering with the crucial gene expression, making it one of the most promising gene therapy agents. However, for lung cancer therapy, the microRNA treatment confronted three bottlenecks, the poor tumor tissue penetration effect, the insufficient lung drug accumulation and unsatisfied gene transfection efficiency. To address these issues, an inhalable RGD-TAT dual peptides-modified cationic liposomes loaded with microRNA miR-34a and gap junction (GJ) regulation agent all-trans retinoic acid (ATRA) was proposed, which was further engineered into dry powder inhalers (DPIs). RESULTS Equipped with a rough particle surface and appropriate aerodynamic size, the proposed RGD-TAT-CLPs/ARTA@miR-34a DPIs were expected to deposit into the deep lung and reach lung tumor lesions guided by targeting peptide RGD. Assisted by cellular transmembrane peptides TAT, the RGD-TAT-CLPs/ARTA@miR-34a was proven to be effectively internalized by cancer cells, enhancing gene transfection efficiency. Then, the GJ between tumor cells was upregulated by ARTA, facilitating the intercellular transport of miR-34a and boosting the gene expression in the deep tumor. CONCLUSION Overall, the proposed RGD-TAT-CLPs/ARTA@miR-34a DPIs could enhance tumor tissue penetration, elevate lung drug accumulation and boost gene transfection efficiency, breaking the three bottlenecks to enhancing tumor elimination in vitro and in vivo. We believe that the proposed RGD-TAT-CLPs/ARTA@miR-34a DPIs could serve as a promising pulmonary gene delivery platform for multiple lung local disease treatments.
Collapse
Affiliation(s)
- Ziyu Zhao
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, PR China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, 510632, PR China
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510632, PR China
| | - Wenhao Wang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, 510006, PR China
| | - Guanlin Wang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, Guangdong, 510006, PR China
| | - Zhengwei Huang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, 510632, PR China
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510632, PR China
| | - Liping Zhou
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, PR China
| | - Li Lin
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, PR China
| | - Yueling Ou
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, PR China
| | - Wanzhen Huang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, PR China
| | - Xuejuan Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, 510632, PR China.
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510632, PR China.
| | - Chuanbin Wu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou, Guangdong, 510632, PR China
- College of Pharmacy, Jinan University, Guangzhou, Guangdong, 510632, PR China
| | - Liang Tao
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, PR China.
- Nanchang Research Institute, Sun Yat-Sen University, Nanchang, Jiangxi, 330096, PR China.
| | - Qin Wang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, PR China.
- Nanchang Research Institute, Sun Yat-Sen University, Nanchang, Jiangxi, 330096, PR China.
| |
Collapse
|
20
|
Li H, Zhu Y, Wang X, Feng Y, Qian Y, Ma Q, Li X, Chen Y, Chen K. Joining Forces: The Combined Application of Therapeutic Viruses and Nanomaterials in Cancer Therapy. Molecules 2023; 28:7679. [PMID: 38005401 PMCID: PMC10674375 DOI: 10.3390/molecules28227679] [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: 09/29/2023] [Revised: 11/10/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
Cancer, on a global scale, presents a monumental challenge to our healthcare systems, posing a significant threat to human health. Despite the considerable progress we have made in the diagnosis and treatment of cancer, realizing precision cancer therapy, reducing side effects, and enhancing efficacy remain daunting tasks. Fortunately, the emergence of therapeutic viruses and nanomaterials provides new possibilities for tackling these issues. Therapeutic viruses possess the ability to accurately locate and attack tumor cells, while nanomaterials serve as efficient drug carriers, delivering medication precisely to tumor tissues. The synergy of these two elements has led to a novel approach to cancer treatment-the combination of therapeutic viruses and nanomaterials. This advantageous combination has overcome the limitations associated with the side effects of oncolytic viruses and the insufficient tumoricidal capacity of nanomedicines, enabling the oncolytic viruses to more effectively breach the tumor's immune barrier. It focuses on the lesion site and even allows for real-time monitoring of the distribution of therapeutic viruses and drug release, achieving a synergistic effect. This article comprehensively explores the application of therapeutic viruses and nanomaterials in tumor treatment, dissecting their working mechanisms, and integrating the latest scientific advancements to predict future development trends. This approach, which combines viral therapy with the application of nanomaterials, represents an innovative and more effective treatment strategy, offering new perspectives in the field of tumor therapy.
Collapse
Affiliation(s)
- Hongyu Li
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
- Ocean College, Beibu Gulf University, Qinzhou 535011, China
| | - Yunhuan Zhu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
| | - Xin Wang
- Center of Infectious Disease Research, School of Life Science, Westlake University, Hangzhou 310024, China;
| | - Yilu Feng
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
| | - Yuncheng Qian
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
| | - Qiman Ma
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
| | - Xinyuan Li
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
| | - Yihan Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
| | - Keda Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (Y.Z.); (Y.F.); (Y.Q.); (Q.M.); (X.L.); (Y.C.)
| |
Collapse
|
21
|
Wong B, Bergeron A, Maznyi G, Ng K, Jirovec A, Birdi HK, Serrano D, Spinelli M, Thomson M, Taha Z, Alwithenani A, Chen A, Lorimer I, Vanderhyden B, Arulanandam R, Diallo JS. Pevonedistat, a first-in-class NEDD8-activating enzyme inhibitor, sensitizes cancer cells to VSVΔ51 oncolytic virotherapy. Mol Ther 2023; 31:3176-3192. [PMID: 37766429 PMCID: PMC10638453 DOI: 10.1016/j.ymthe.2023.09.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/23/2023] [Accepted: 09/23/2023] [Indexed: 09/29/2023] Open
Abstract
The clinical efficacy of VSVΔ51 oncolytic virotherapy has been limited by tumor resistance to viral infection, so strategies to transiently repress antiviral defenses are warranted. Pevonedistat is a first-in-class NEDD8-activating enzyme (NAE) inhibitor currently being tested in clinical trials for its antitumor potential. In this study, we demonstrate that pevonedistat sensitizes human and murine cancer cells to increase oncolytic VSVΔ51 infection, increase tumor cell death, and improve therapeutic outcomes in resistant syngeneic murine cancer models. Increased VSVΔ51 infectivity was also observed in clinical human tumor samples. We further identify the mechanism of this effect to operate via blockade of the type 1 interferon (IFN-1) response through neddylation-dependent interferon-stimulated growth factor 3 (ISGF3) repression and neddylation-independent inhibition of NF-κB nuclear translocation. Together, our results identify a role for neddylation in regulating the innate immune response and demonstrate that pevonedistat can improve the therapeutic outcomes of strategies using oncolytic virotherapy.
Collapse
Affiliation(s)
- Boaz Wong
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Anabel Bergeron
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Glib Maznyi
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Kristy Ng
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Anna Jirovec
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Harsimrat K Birdi
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Daniel Serrano
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Marcus Spinelli
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Max Thomson
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Zaid Taha
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Akram Alwithenani
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Andrew Chen
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Ian Lorimer
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Barbara Vanderhyden
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Rozanne Arulanandam
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada.
| | - Jean-Simon Diallo
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada; Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
22
|
Liu S, Wang W, Hu S, Jia B, Tuo B, Sun H, Wang Q, Liu Y, Sun Z. Radiotherapy remodels the tumor microenvironment for enhancing immunotherapeutic sensitivity. Cell Death Dis 2023; 14:679. [PMID: 37833255 PMCID: PMC10575861 DOI: 10.1038/s41419-023-06211-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 09/22/2023] [Accepted: 10/02/2023] [Indexed: 10/15/2023]
Abstract
Cancer immunotherapy has transformed traditional treatments, with immune checkpoint blockade being particularly prominent. However, immunotherapy has minimal benefit for patients in most types of cancer and is largely ineffective in some cancers (such as pancreatic cancer and glioma). A synergistic anti-tumor response may be produced through the combined application with traditional tumor treatment methods. Radiotherapy (RT) not only kills tumor cells but also triggers the pro-inflammatory molecules' release and immune cell infiltration, which remodel the tumor microenvironment (TME). Therefore, the combination of RT and immunotherapy is expected to achieve improved efficacy. In this review, we summarize the effects of RT on cellular components of the TME, including T cell receptor repertoires, different T cell subsets, metabolism, tumor-associated macrophages and other myeloid cells (dendritic cells, myeloid-derived suppressor cells, neutrophils and eosinophils). Meanwhile, non-cellular components such as lactate and extracellular vesicles are also elaborated. In addition, we discuss the impact of different RT modalities on tumor immunity and issues related to the clinical practice of combination therapy.
Collapse
Affiliation(s)
- Senbo Liu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Wenkang Wang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Shengyun Hu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Bin Jia
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Baojing Tuo
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Haifeng Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Qiming Wang
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 450001, Zhengzhou, China.
| | - Yang Liu
- Department of Radiotherapy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, 450001, Zhengzhou, China.
| | - Zhenqiang Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China.
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China.
| |
Collapse
|
23
|
Liu D, Che X, Wang X, Ma C, Wu G. Tumor Vaccines: Unleashing the Power of the Immune System to Fight Cancer. Pharmaceuticals (Basel) 2023; 16:1384. [PMID: 37895855 PMCID: PMC10610367 DOI: 10.3390/ph16101384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 09/25/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
This comprehensive review delves into the rapidly evolving arena of cancer vaccines. Initially, we examine the intricate constitution of the tumor microenvironment (TME), a dynamic factor that significantly influences tumor heterogeneity. Current research trends focusing on harnessing the TME for effective tumor vaccine treatments are also discussed. We then provide a detailed overview of the current state of research concerning tumor immunity and the mechanisms of tumor vaccines, describing the complex immunological processes involved. Furthermore, we conduct an exhaustive analysis of the contemporary research landscape of tumor vaccines, with a particular focus on peptide vaccines, DNA/RNA-based vaccines, viral-vector-based vaccines, dendritic-cell-based vaccines, and whole-cell-based vaccines. We analyze and summarize these categories of tumor vaccines, highlighting their individual advantages, limitations, and the factors influencing their effectiveness. In our survey of each category, we summarize commonly used tumor vaccines, aiming to provide readers with a more comprehensive understanding of the current state of tumor vaccine research. We then delve into an innovative strategy combining cancer vaccines with other therapies. By studying the effects of combining tumor vaccines with immune checkpoint inhibitors, radiotherapy, chemotherapy, targeted therapy, and oncolytic virotherapy, we establish that this approach can enhance overall treatment efficacy and offset the limitations of single-treatment approaches, offering patients more effective treatment options. Following this, we undertake a meticulous analysis of the entire process of personalized cancer vaccines, elucidating the intricate process from design, through research and production, to clinical application, thus helping readers gain a thorough understanding of its complexities. In conclusion, our exploration of tumor vaccines in this review aims to highlight their promising potential in cancer treatment. As research in this field continues to evolve, it undeniably holds immense promise for improving cancer patient outcomes.
Collapse
Affiliation(s)
- Dequan Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (D.L.); (X.C.)
| | - Xiangyu Che
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (D.L.); (X.C.)
| | - Xiaoxi Wang
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China;
| | - Chuanyu Ma
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (D.L.); (X.C.)
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; (D.L.); (X.C.)
| |
Collapse
|
24
|
Pu T, Peddle A, Zhu J, Tejpar S, Verbandt S. Neoantigen identification: Technological advances and challenges. Methods Cell Biol 2023; 183:265-302. [PMID: 38548414 DOI: 10.1016/bs.mcb.2023.06.005] [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] [Indexed: 04/02/2024]
Abstract
Neoantigens have emerged as promising targets for cutting-edge immunotherapies, such as cancer vaccines and adoptive cell therapy. These neoantigens are unique to tumors and arise exclusively from somatic mutations or non-genomic aberrations in tumor proteins. They encompass a wide range of alterations, including genomic mutations, post-transcriptomic variants, and viral oncoproteins. With the advancements in technology, the identification of immunogenic neoantigens has seen rapid progress, raising new opportunities for enhancing their clinical significance. Prediction of neoantigens necessitates the acquisition of high-quality samples and sequencing data, followed by mutation calling. Subsequently, the pipeline involves integrating various tools that can predict the expression, processing, binding, and recognition potential of neoantigens. However, the continuous improvement of computational tools is constrained by the availability of datasets which contain validated immunogenic neoantigens. This review article aims to provide a comprehensive summary of the current knowledge as well as limitations in neoantigen prediction and validation. Additionally, it delves into the origin and biological role of neoantigens, offering a deeper understanding of their significance in the field of cancer immunotherapy. This article thus seeks to contribute to the ongoing efforts to harness neoantigens as powerful weapons in the fight against cancer.
Collapse
Affiliation(s)
- Ting Pu
- Digestive Oncology Unit, KULeuven, Leuven, Belgium
| | | | - Jingjing Zhu
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | | | | |
Collapse
|
25
|
Tojjari A, Saeed A, Singh M, Cavalcante L, Sahin IH, Saeed A. A Comprehensive Review on Cancer Vaccines and Vaccine Strategies in Hepatocellular Carcinoma. Vaccines (Basel) 2023; 11:1357. [PMID: 37631925 PMCID: PMC10459477 DOI: 10.3390/vaccines11081357] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/04/2023] [Accepted: 08/09/2023] [Indexed: 08/29/2023] Open
Abstract
HCC, the most prevalent form of primary liver cancer, presents a substantial global health challenge due to its high mortality and limited therapeutic options. This review delves into the potential of cancer vaccines as a novel therapeutic avenue for HCC. We examine the various categories of cancer vaccines, including peptide-based, dendritic cell-based, viral vector-based, DNA, and mRNA vaccines, and their potential application in HCC management. This review also addresses the inherent challenges in vaccine development, such as tumor heterogeneity and the need for identifying tumor-specific antigens. We underscore the role of cancer vaccines in reshaping the immune environment within HCC, fostering durable immune memory, and their potential in combination therapies. The review also evaluates clinical trials and emphasizes the necessity for more extensive research to optimize vaccine design and patient selection criteria. We conclude with future perspectives, highlighting the significance of personalized therapies, innovative antigen delivery platforms, immune modulatory agents, and predictive biomarkers in revolutionizing HCC treatment. Simple Summary: This review explores the potential of cancer vaccines as a promising therapeutic strategy for hepatocellular carcinoma (HCC), a prevalent and deadly liver cancer. The authors discuss various types of cancer vaccines, their challenges, and their role in modulating the immune response within HCC. They also highlight clinical trials and future perspectives, emphasizing the importance of personalized therapies, novel antigen delivery platforms, and predictive biomarkers. The findings from this research could significantly impact the research community by providing a comprehensive understanding of the current state of cancer vaccines for HCC, thereby guiding future research and potentially transforming HCC treatment strategies.
Collapse
Affiliation(s)
- Alireza Tojjari
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA 15213, USA; (A.T.); (M.S.); (I.H.S.)
| | - Ahmed Saeed
- Sarah Cannon Cancer Institute, HCA Midwest Health, Kansas City, MO 64131, USA;
| | - Meghana Singh
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA 15213, USA; (A.T.); (M.S.); (I.H.S.)
| | | | - Ibrahim Halil Sahin
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA 15213, USA; (A.T.); (M.S.); (I.H.S.)
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| | - Anwaar Saeed
- Department of Medicine, Division of Hematology & Oncology, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA 15213, USA; (A.T.); (M.S.); (I.H.S.)
- UPMC Hillman Cancer Center, Pittsburgh, PA 15213, USA
| |
Collapse
|
26
|
Chen P, Jiang Y, Liang J, Cai J, Zhuo Y, Fan H, Yuan R, Cheng S, Zhang Y. SLC1A5 is a novel biomarker associated with ferroptosis and the tumor microenvironment: a pancancer analysis. Aging (Albany NY) 2023; 15:7451-7475. [PMID: 37566748 PMCID: PMC10457057 DOI: 10.18632/aging.204911] [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: 02/20/2023] [Accepted: 07/06/2023] [Indexed: 08/13/2023]
Abstract
Solute carrier family 1 member 5 (SLC1A5) is a member of the solute carrier (SLC) superfamily of transporters and plays an important role in tumors as a key transporter of glutamine into cells. However, the relationship between SLC1A5, which is involved in immune regulation, and immune cell infiltration in the tumor microenvironment has not been elucidated, and the relationship between SLC1A5 and ferroptosis is rarely reported. Therefore, we comprehensively analyzed the expression level of SLC1A5 across cancers and compared it with that in normal tissues. Then, the relationship between SLC1A5 expression and the tumor immune microenvironment was analyzed by single-cell analysis, gene set enrichment analysis (GSEA), and Tumor Immune Estimation Resource (TIMER). Next, the correlations of the SLC1A5 expression level with immunotherapy response, immunomodulator expression, tumor mutation burden (TMB) and microsatellite instability (MSI) were evaluated. Finally, in vitro experiments verified that SLC1A5 participates in ferroptosis of glioma cells to regulate tumor progression. Our results indicated that SLC1A5 is aberrantly expressed in most cancer types and closely associated with prognosis. The GSEA results showed that SLC1A5 is involved in immune activation processes and closely related to the infiltration levels of different immune cells in different cancer types. Upon further investigation, we found that SLC1A5 is a suppressor of ferroptosis in glioma, and SLC1A5 knockdown inhibited the proliferation and migration of glioma cells in vitro. In conclusion, we conducted a pancancer analysis of SLC1A5, demonstrated its role as a prognostic biomarker in cancer patients and explored its potential biological functions.
Collapse
Affiliation(s)
- Peng Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
- Department of Medical, Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - YongAn Jiang
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
- Department of Medical, Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - JiaWei Liang
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
- Department of Medical, Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - JiaHong Cai
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
- Department of Medical, Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Yi Zhuo
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
- Department of Medical, Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - HengYi Fan
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - RaoRao Yuan
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - ShiQi Cheng
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| | - Yan Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, P.R. China
| |
Collapse
|
27
|
Wang X, Shen Y, Wan X, Hu X, Cai WQ, Wu Z, Xin Q, Liu X, Gui J, Xin HY, Xin HW. Oncolytic virotherapy evolved into the fourth generation as tumor immunotherapy. J Transl Med 2023; 21:500. [PMID: 37491263 PMCID: PMC10369732 DOI: 10.1186/s12967-023-04360-8] [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: 12/28/2022] [Accepted: 07/16/2023] [Indexed: 07/27/2023] Open
Abstract
BACKGROUND Oncolytic virotherapy (OVT) is a promising anti-tumor modality that utilizes oncolytic viruses (OVs) to preferentially attack cancers rather than normal tissues. With the understanding particularly in the characteristics of viruses and tumor cells, numerous innovative OVs have been engineered to conquer cancers, such as Talimogene Laherparepvec (T-VEC) and tasadenoturev (DNX-2401). However, the therapeutic safety and efficacy must be further optimized and balanced to ensure the superior safe and efficient OVT in clinics, and reasonable combination therapy strategies are also important challenges worthy to be explored. MAIN BODY Here we provided a critical review of the development history and status of OVT, emphasizing the mechanisms of enhancing both safety and efficacy. We propose that oncolytic virotherapy has evolved into the fourth generation as tumor immunotherapy. Particularly, to arouse T cells by designing OVs expressing bi-specific T cell activator (BiTA) is a promising strategy of killing two birds with one stone. Amazing combination of therapeutic strategies of OVs and immune cells confers immense potential for managing cancers. Moreover, the attractive preclinical OVT addressed recently, and the OVT in clinical trials were systematically reviewed. CONCLUSION OVs, which are advancing into clinical trials, are being envisioned as the frontier clinical anti-tumor agents coming soon.
Collapse
Affiliation(s)
- Xianwang Wang
- Department of Biochemistry and Molecular Biology, Health Science Center, Yangtze University, Jingzhou, 434023, Hubei, China.
| | - Yihua Shen
- The Second School of Clinical Medicine, Yangtze University, Jingzhou, 434023, Hubei, China
| | - Xingxia Wan
- College of Arts and Sciences, Yangtze University, Jingzhou, 434023, Hubei, China
| | - Xiaoqing Hu
- The Second School of Clinical Medicine, Yangtze University, Jingzhou, 434023, Hubei, China
| | - Wen-Qi Cai
- Xinzhou Traditional Chinese Medicine Hospital, Zhongnan Hospital of Wuhan University (Xinzhou), Wuhan, 430000, Hubei, China
| | - Zijun Wu
- The Second School of Clinical Medicine, Yangtze University, Jingzhou, 434023, Hubei, China
| | - Qiang Xin
- School of Graduate Students, Inner Mongolia Medical University, Inner Mongolian Autonomous Region, Hohhot, 010110, China
| | - Xiaoqing Liu
- College of Arts and Sciences, Yangtze University, Jingzhou, 434023, Hubei, China
| | - Jingang Gui
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Hong-Yi Xin
- The Doctoral Scientific Research Center, People's Hospital of Lianjiang, Guangdong, 524400, China.
- The Doctoral Scientific Research Center, Affiliated People's Hospital of Lianjiang, Guangdong Medical University, Guangdong, 524400, China.
| | - Hong-Wu Xin
- Department of Biochemistry and Molecular Biology, Health Science Center, Yangtze University, Jingzhou, 434023, Hubei, China.
| |
Collapse
|
28
|
Kaur SD, Singh AD, Kapoor DN. Current perspectives on Vaxinia virus: an immuno-oncolytic vector in cancer therapy. Med Oncol 2023; 40:205. [PMID: 37318642 DOI: 10.1007/s12032-023-02068-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 05/14/2023] [Indexed: 06/16/2023]
Abstract
Viruses are being researched as cutting-edge therapeutic agents in cancer due to their selective oncolytic action against malignancies. Immuno-oncolytic viruses are a potential category of anticancer treatments because they have natural features that allow viruses to efficiently infect, replicate, and destroy cancer cells. Oncolytic viruses may be genetically modified; engineers can use them as a platform to develop additional therapy modalities that overcome the limitations of current treatment approaches. In recent years, researchers have made great strides in the understanding relationship between cancer and the immune system. An increasing corpus of research is functioning on the immunomodulatory functions of oncolytic virus (OVs). Several clinical studies are currently underway to determine the efficacy of these immuno-oncolytic viruses. These studies are exploring the design of these platforms to elicit the desired immune response and to supplement the available immunotherapeutic modalities to render immune-resistant malignancies amenable to treatment. This review will discuss current research and clinical developments on Vaxinia immuno-oncolytic virus.
Collapse
Affiliation(s)
- Simran Deep Kaur
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, Himachal Pradesh, 173229, India
| | - Aman Deep Singh
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, 142048, India
| | - Deepak N Kapoor
- School of Pharmaceutical Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, Himachal Pradesh, 173229, India.
| |
Collapse
|
29
|
Bahreyni A, Liu H, Mohamud Y, Xue YC, Fan YM, Zhang YL, Luo H. A combination of genetically engineered oncolytic virus and melittin-CpG for cancer viro-chemo-immunotherapy. BMC Med 2023; 21:193. [PMID: 37226233 DOI: 10.1186/s12916-023-02901-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 05/12/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND Immunotherapy has emerged as an efficient therapeutic approach for cancer management. However, stimulation of host immune system against cancer cells often fails to achieve promising clinical outcomes mainly owing to the immunosuppressive characteristics of the tumor microenvironment (TME). Combination therapeutics that can trigger sustained immunogenic cell death (ICD) have provided new opportunities for cancer treatment. METHODS In this study, we designed and applied an ICD inducer regimen, including a genetically engineered oncolytic virus (miRNA-modified coxsackieviruses B3, miR-CVB3), a pore-forming lytic peptide (melittin, found in bee venom), and a synthetic toll-like receptor 9 ligand (CpG oligodeoxynucleotides), for breast cancer and melanoma treatment. We compared the anti-tumor efficacy of miR-CVB3 and CpG-melittin (CpGMel) alone and in combination (miR-CVB3 + CpGMel) and investigated possible mechanisms involved. RESULTS We demonstrated that miR-CVB3 + CpGMel had no major impact on viral growth, while enhancing the cellular uptake of CpGMel in vitro. We further showed that combination therapy led to significant increases in tumor cell death and release of damage-associated molecular patterns compared with individual treatment. In vivo studies in 4T1 tumor-bearing Balb/c mice revealed that both primary and distant tumors were significantly suppressed, and the survival rate was significantly prolonged after administration of miR-CVB3 + CpGMel compared with single treatment. This anti-tumor effect was accompanied by increased ICD and immune cell infiltration into the TME. Safety analysis showed no significant pathological abnormalities in Balb/c mice. Furthermore, the developed therapeutic regimen also demonstrated a great anti-tumor activity in B16F10 melanoma tumor-bearing C57BL/6 J mice. CONCLUSIONS Overall, our findings indicate that although single treatment using miR-CVB3 or CpGMel can efficiently delay tumor growth, combining oncolytic virus-based therapy can generate even stronger anti-tumor immunity, leading to a greater reduction in tumor size.
Collapse
Affiliation(s)
- Amirhossein Bahreyni
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, V6Z 1Y6, Canada
- Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC, V6Z 1Y6, Canada
| | - Huitao Liu
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, V6Z 1Y6, Canada
- Department of Experimental Medicine, University of British Columbia, Vancouver, BC, V6Z 1Y6, Canada
| | - Yasir Mohamud
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, V6Z 1Y6, Canada
- Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC, V6Z 1Y6, Canada
| | - Yuan Chao Xue
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, V6Z 1Y6, Canada
- Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC, V6Z 1Y6, Canada
| | - Yiyun Michelle Fan
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, V6Z 1Y6, Canada
- Department of Cellular and Physiological Sciences, University of British Columbia, Endowment Lands, Canada
| | - Yizhuo Lyanne Zhang
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, V6Z 1Y6, Canada
- Department of Cellular and Physiological Sciences, University of British Columbia, Endowment Lands, Canada
| | - Honglin Luo
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, V6Z 1Y6, Canada.
- Department of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC, V6Z 1Y6, Canada.
| |
Collapse
|
30
|
Vannini A, Parenti F, Forghieri C, Barboni C, Zaghini A, Campadelli-Fiume G, Gianni T. Innovative retargeted oncolytic herpesvirus against nectin4-positive cancers. Front Mol Biosci 2023; 10:1149973. [PMID: 37251078 PMCID: PMC10213976 DOI: 10.3389/fmolb.2023.1149973] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 05/02/2023] [Indexed: 05/31/2023] Open
Abstract
Nectin4 is a recently discovered tumor associated antigen expressed in cancers that constitute relevant unmet clinical needs, including the undruggable triple negative breast cancer, pancreatic ductal carcinoma, bladder/urothelial cancer, cervical cancer, lung carcinoma and melanoma. So far, only one nectin4-specific drug-Enfortumab Vedotin-has been approved and the clinical trials that test novel therapeutics are only five. Here we engineered R-421, an innovative retargeted onco-immunotherapeutic herpesvirus highly specific for nectin4 and unable to infect through the natural herpes receptors, nectin1 or herpesvirus entry mediator. In vitro, R-421 infected and killed human nectin4-positive malignant cells and spared normal cells, e.g., human fibroblasts. Importantly from a safety viewpoint, R-421 failed to infect malignant cells that do not harbor nectin4 gene amplification/overexpression, whose expression level was moderate-to-low. In essence, there was a net threshold value below which cells were spared from infection, irrespective of whether they were malignant or normal; the only cells that R-421 targeted were the malignant overexpressing ones. In vivo, R-421 decreased or abolished the growth of murine tumors made transgenic for human nectin4 and conferred sensitivity to immune checkpoint inhibitors in combination therapies. Its efficacy was augmented by the cyclophosphamide immunomodulator and decreased by depletion of CD8-positive lymphocytes, arguing that it was in part T cell-mediated. R-421 elicited in-situ vaccination that protected from distant challenge tumors. This study provides proof-of-principle specificity and efficacy data justifying nectin4-retargeted onco-immunotherapeutic herpesvirus as an innovative approach against a number of difficult-to-drug clinical indications.
Collapse
Affiliation(s)
- Andrea Vannini
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Federico Parenti
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Cristina Forghieri
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Catia Barboni
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | - Anna Zaghini
- Department of Veterinary Medical Sciences, University of Bologna, Bologna, Italy
| | | | - Tatiana Gianni
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| |
Collapse
|
31
|
Kciuk M, Yahya EB, Mohamed Ibrahim Mohamed M, Rashid S, Iqbal MO, Kontek R, Abdulsamad MA, Allaq AA. Recent Advances in Molecular Mechanisms of Cancer Immunotherapy. Cancers (Basel) 2023; 15:2721. [PMID: 37345057 DOI: 10.3390/cancers15102721] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/09/2023] [Accepted: 05/10/2023] [Indexed: 06/23/2023] Open
Abstract
Cancer is among the current leading causes of death worldwide, despite the novel advances that have been made toward its treatment, it is still considered a major public health concern. Considering both the serious impact of cancer on public health and the significant side effects and complications of conventional therapeutic options, the current strategies towards targeted cancer therapy must be enhanced to avoid undesired toxicity. Cancer immunotherapy has become preferable among researchers in recent years compared to conventional therapeutic options, such as chemotherapy, surgery, and radiotherapy. The understanding of how to control immune checkpoints, develop therapeutic cancer vaccines, genetically modify immune cells as well as enhance the activation of antitumor immune response led to the development of novel cancer treatments. In this review, we address recent advances in cancer immunotherapy molecular mechanisms. Different immunotherapeutic approaches are critically discussed, focusing on the challenges, potential risks, and prospects involving their use.
Collapse
Affiliation(s)
- Mateusz Kciuk
- Department of Molecular Biotechnology and Genetics, University of Lodz, Banacha Street 12/16, 90-237 Lodz, Poland
- Doctoral School of Exact and Natural Sciences, University of Lodz, Banacha Street 12/16, 90-237 Lodz, Poland
| | - Esam Bashir Yahya
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, Penang 11800, Malaysia
| | | | - Summya Rashid
- Department of Pharmacology & Toxicology, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Muhammad Omer Iqbal
- Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Renata Kontek
- Department of Molecular Biotechnology and Genetics, University of Lodz, Banacha Street 12/16, 90-237 Lodz, Poland
| | - Muhanad A Abdulsamad
- Department of Molecular Biology, Faculty of Science, Sabratha University, Sabratha 00218, Libya
| | - Abdulmutalib A Allaq
- Faculty of Applied Science, Universiti Teknologi MARA, Shah Alam 40450, Malaysia
| |
Collapse
|
32
|
Gao H, Qi X, Zhang J, Wang N, Xin J, Jiao D, Liu K, Qi J, Guan Y, Ding D. Smart One-for-All Agent with Adaptive Functions for Improving Photoacoustic /Fluorescence Imaging-Guided Photodynamic Immunotherapy. SMALL METHODS 2023; 7:e2201582. [PMID: 36807567 DOI: 10.1002/smtd.202201582] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/19/2023] [Indexed: 05/17/2023]
Abstract
Multifunctional phototheranostics that integrate several diagnostic and therapeutic strategies into one platform hold great promise for precision medicine. However, it is really difficult for one molecule to possess multimodality optical imaging and therapy properties that all functions are in the optimized mode because the absorbed photoenergy is fixed. Herein, a smart one-for-all nanoagent that the photophysical energy transformation processes can be facilely tuned by external light stimuli is developed for precise multifunctional image-guided therapy. A dithienylethene-based molecule is designed and synthesized because it has two light-switchable forms. In the ring-closed form, most of the absorbed energy dissipates via nonradiative thermal deactivation for photoacoustic (PA) imaging. In the ring-open form, the molecule possesses obvious aggregation-induced emission features with excellent fluorescence and photodynamic therapy properties. In vivo experiments demonstrate that preoperative PA and fluorescence imaging help to delineate tumors in a high-contrast manner, and intraoperative fluorescence imaging is able to sensitively detect tiny residual tumors. Furthermore, the nanoagent can induce immunogenic cell death to elicit antitumor immunity and significantly suppress solid tumors. This work develops a smart one-for-all agent that the photophysical energy transformation and related phototheranostic properties can be optimized by light-driven structure switch, which is promising for multifunctional biomedical applications.
Collapse
Affiliation(s)
- Heqi Gao
- Frontiers Science Center for Cell Responses, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Xinwen Qi
- Frontiers Science Center for Cell Responses, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jingtian Zhang
- Frontiers Science Center for Cell Responses, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Nan Wang
- Frontiers Science Center for Cell Responses, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jingrui Xin
- Frontiers Science Center for Cell Responses, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Di Jiao
- Frontiers Science Center for Cell Responses, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Kaining Liu
- Frontiers Science Center for Cell Responses, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Ji Qi
- Frontiers Science Center for Cell Responses, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Yong Guan
- Department of Urology, Tianjin Children's Hospital /Tianjin University Children's Hospital, Tianjin, 300134, China
| | - Dan Ding
- Frontiers Science Center for Cell Responses, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| |
Collapse
|
33
|
Storozynsky QT, Agopsowicz KC, Noyce RS, Bukhari AB, Han X, Snyder N, Umer BA, Gamper AM, Godbout R, Evans DH, Hitt MM. Radiation combined with oncolytic vaccinia virus provides pronounced antitumor efficacy and induces immune protection in an aggressive glioblastoma model. Cancer Lett 2023; 562:216169. [PMID: 37061120 DOI: 10.1016/j.canlet.2023.216169] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/26/2023] [Accepted: 04/05/2023] [Indexed: 04/17/2023]
Abstract
Glioblastoma (GB) is a malignant and immune-suppressed brain cancer that remains incurable despite the current standard of care. Radiotherapy is a mainstay of GB treatment, however invasive cancer cells outside the irradiated field and radioresistance preclude complete eradication of GB cells. Oncolytic virus therapy harnesses tumor-selective viruses to spread through and destroy tumors while stimulating antitumor immune responses, and thus has potential for use following radiotherapy. We demonstrate that oncolytic ΔF4LΔJ2R vaccinia virus (VACV) replicates in and induces cytotoxicity of irradiated brain tumor initiating cells in vitro. Importantly, a single 10 Gy dose of radiation combined with ΔF4LΔJ2R VACV produced considerably superior anticancer effects relative to either monotherapy when treating immune-competent orthotopic CT2A-luc mouse models-significantly extending survival and curing the majority of mice. Mice cured by the combination displayed significantly increased survival relative to naïve age-matched controls following intracranial tumor challenge, with some complete rejections. Further, the combination therapy was associated with an increased ratio of CD8+ effector T cells to regulatory T cells compared to either monotherapy. This study validates the use of radiation with an oncolytic ΔF4LΔJ2R VACV to improve treatment of this malignant brain cancer.
Collapse
Affiliation(s)
- Quinn T Storozynsky
- Department of Oncology, University of Alberta, Edmonton, AB, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada; Cancer Research Institute of Northern Alberta (CRINA), University of Alberta, Edmonton, AB, Canada
| | | | - Ryan S Noyce
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Amirali B Bukhari
- Department of Oncology, University of Alberta, Edmonton, AB, Canada; Cancer Research Institute of Northern Alberta (CRINA), University of Alberta, Edmonton, AB, Canada
| | - Xuefei Han
- Department of Oncology, University of Alberta, Edmonton, AB, Canada; Department of Neurosurgery, First Hospital of Jilin University, Changchun, China
| | - Natalie Snyder
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
| | - Brittany A Umer
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Armin M Gamper
- Department of Oncology, University of Alberta, Edmonton, AB, Canada; Cancer Research Institute of Northern Alberta (CRINA), University of Alberta, Edmonton, AB, Canada
| | - Roseline Godbout
- Department of Oncology, University of Alberta, Edmonton, AB, Canada; Cancer Research Institute of Northern Alberta (CRINA), University of Alberta, Edmonton, AB, Canada
| | - David H Evans
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, AB, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Mary M Hitt
- Department of Oncology, University of Alberta, Edmonton, AB, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada; Cancer Research Institute of Northern Alberta (CRINA), University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
34
|
Yang A, Zhang Z, Chaurasiya S, Park AK, Jung A, Lu J, Kim SI, Priceman S, Fong Y, Woo Y. Development of the oncolytic virus, CF33, and its derivatives for peritoneal-directed treatment of gastric cancer peritoneal metastases. J Immunother Cancer 2023; 11:e006280. [PMID: 37019471 PMCID: PMC10083877 DOI: 10.1136/jitc-2022-006280] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2023] [Indexed: 04/07/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) that metastasizes to the peritoneum is fatal. CF33 and its genetically modified derivatives show cancer selectivity and oncolytic potency against various solid tumors. CF33-hNIS and CF33-hNIS-antiPDL1 have entered phase I trials for intratumoral and intravenous treatments of unresectable solid tumors (NCT05346484) and triple-negative breast cancer (NCT05081492). Here, we investigated the antitumor activity of CF33-oncolytic viruses (OVs) against GC and CF33-hNIS-antiPDL1 in the intraperitoneal (IP) treatment of GC peritoneal metastases (GCPM). METHODS We infected six human GC cell lines AGS, MKN-45, MKN-74, KATO III, SNU-1, and SNU-16 with CF33, CF33-GFP, or CF33-hNIS-antiPDL1 at various multiplicities of infection (0.01, 0.1, 1.0, and 10.0), and performed viral proliferation and cytotoxicity assays. We used immunofluorescence imaging and flow cytometric analysis to verify virus-encoded gene expression. We evaluated the antitumor activity of CF33-hNIS-antiPDL1 following IP treatment (3×105 pfu × 3 doses) in an SNU-16 human tumor xenograft model using non-invasive bioluminescence imaging. RESULTS CF33-OVs showed dose-dependent infection, replication, and killing of both diffuse and intestinal subtypes of human GC cell lines. Immunofluorescence imaging showed virus-encoded GFP, hNIS, and anti-PD-L1 antibody scFv expression in CF33-OV-infected GC cells. We confirmed GC cell surface PD-L1 blockade by virus-encoded anti-PD-L1 scFv using flow cytometry. In the xenograft model, CF33-hNIS-antiPDL1 (IP; 3×105 pfu × 3 doses) treatment significantly reduced peritoneal tumors (p<0.0001), decreased amount of ascites (62.5% PBS vs 25% CF33-hNIS-antiPDL1) and prolonged animal survival. At day 91, seven out of eight mice were alive in the virus-treated group versus one out of eight in the control group (p<0.01). CONCLUSIONS Our results show that CF33-OVs can deliver functional proteins and demonstrate effective antitumor activity in GCPM models when delivered intraperitoneally. These preclinical results will inform the design of future peritoneal-directed therapy in GCPM patients.
Collapse
Affiliation(s)
- Annie Yang
- Department of Surgery, City of Hope National Medical Center, Duarte, California, USA
| | - Zhifang Zhang
- Department of Surgery, City of Hope National Medical Center, Duarte, California, USA
| | - Shyambabu Chaurasiya
- Department of Surgery, City of Hope National Medical Center, Duarte, California, USA
| | - Anthony K Park
- Cancer Immunotherapeutics Program, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Audrey Jung
- Department of Surgery, City of Hope National Medical Center, Duarte, California, USA
| | - Jianming Lu
- Department of Surgery, City of Hope National Medical Center, Duarte, California, USA
| | - Sang-In Kim
- Department of Surgery, City of Hope National Medical Center, Duarte, California, USA
| | - Saul Priceman
- Cancer Immunotherapeutics Program, Beckman Research Institute, City of Hope National Medical Center, Duarte, California, USA
| | - Yuman Fong
- Department of Surgery, City of Hope National Medical Center, Duarte, California, USA
| | - Yanghee Woo
- Department of Surgery, City of Hope National Medical Center, Duarte, California, USA
| |
Collapse
|
35
|
Huang S, Hu H, Tang G, Liu K, Luo Z, Zeng W. An oncolytic herpes simplex virus type 1 strain expressing a single-chain variable region antibody fragment against PD-1 and a PI3K inhibitor synergize to elicit antitumor immunity in ovarian cancer. Arch Virol 2023; 168:128. [PMID: 37002434 DOI: 10.1007/s00705-023-05754-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 02/21/2023] [Indexed: 04/03/2023]
Abstract
Due to recurrence and resistance to chemotherapy, the current standard therapeutics are not fully effective against ovarian cancer. Therefore, we aimed to find an effective approach to improve the prognosis and therapy of ovarian cancer. NG34ScFvPD-1 is a modified oncolytic herpes simplex virus NG34 strain that expresses a single-chain antibody against programmed cell death protein 1 (PD-1) (ScFvPD-1). We assessed its efficacy and its regulatory mechanism in a mouse model of ovarian cancer. Enzyme-linked immunosorbent assay and western blot techniques were used to measure protein expression. Oncolysis caused by NG34ScFvPD-1 was examined using cytotoxicity and replication assays. The mechanism by which NG34ScFvPD-1 regulates apoptosis of ovarian cancer cells in vitro was also evaluated. We assessed the antitumor immunity and therapeutic potency of NG34ScFvPD-1 in combination with a phosphoinositide 3-kinase (PI3K) inhibitor. We found that NG34ScFvPD-1-infected ovarian cancer cells expressed and secreted ScFvPD-1, which bound mouse PD-1. The insertion of the ScFvPD-1 sequence did not inhibit the oncolytic activity of NG34ScFvPD-1, which induced apoptosis of ovarian cancer cells via the caspase-dependent pathway in vitro and activated the PI3K/AKT signaling pathway. Synergy was observed between NG34ScFvPD-1 and a PI3K inhibitor, and the combination was able to suppress tumor development, to prolong survival, and to elicit potent antitumor immunity. Thus, inhibition of PI3K enhanced the potent antitumor immunity induced by NG34ScFvPD-1 against ovarian cancer.
Collapse
Affiliation(s)
- Shanying Huang
- Inpatient Department of Gynaecology, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, No.2004 Hongli Road, Futian District, Shenzhen, Guangdong, 518028, China.
| | - Haiyan Hu
- Inpatient Department of Gynaecology, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, No.2004 Hongli Road, Futian District, Shenzhen, Guangdong, 518028, China
| | - Guoling Tang
- Inpatient Department of Gynaecology, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, No.2004 Hongli Road, Futian District, Shenzhen, Guangdong, 518028, China
| | - Kai Liu
- Inpatient Department of Gynaecology, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, No.2004 Hongli Road, Futian District, Shenzhen, Guangdong, 518028, China
| | - Zhihua Luo
- Inpatient Department of Gynaecology, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, No.2004 Hongli Road, Futian District, Shenzhen, Guangdong, 518028, China
| | - Weiwei Zeng
- Inpatient Department of Gynaecology, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, No.2004 Hongli Road, Futian District, Shenzhen, Guangdong, 518028, China
| |
Collapse
|
36
|
Garate-Soraluze E, Serrano-Mendioroz I, Rodriguez-Ruiz M. Methods to assess radiation induced abscopal responses in mice. Methods Cell Biol 2023; 180:81-92. [PMID: 37890934 DOI: 10.1016/bs.mcb.2023.02.011] [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] [Indexed: 03/30/2023]
Abstract
Radiotherapy (RT) can work together with the immune system to eliminate cancer. It can cause immunogenic cell death and facilitate tumor neoantigen presentation and thereby the cross-priming of tumor-specific T-lymphocytes, turning irradiated tumors into in-situ vaccines. Accumulating preclinical and clinical evidence indicates that RT in conjunction with ICB leads to systemic anti-tumor immune responses, thus stimulating interest in using ICB to overcome primary and acquired cancer resistance to radiotherapy. However, the systemic effects (abscopal effects) obtained to date are far from being acceptable for clinical translation. In this context, multiple preclinical mouse models have demonstrated that a variety of immunotherapy agents can be delivered locally to enhance antitumor immunity both in a local and systemic fashion. Using two slightly asynchronous and anatomically distant subcutaneous B16OVA tumors in syngeneic immunocompetent hosts (C57BL/6), we describe the feasibility of a local immunotherapy treatment given in combination with external beam irradiation, which exerts immune-mediated antitumor effects in mice and humans upon intratumoral delivery. With minor variations, the same technique can be easily applied to a variety of mouse transplantable tumors.
Collapse
Affiliation(s)
- Eneko Garate-Soraluze
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - Irantzu Serrano-Mendioroz
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain
| | - María Rodriguez-Ruiz
- Program of Immunology and Immunotherapy, Center for Applied Medical Research (CIMA), Pamplona, Spain; Department of Radiation Oncology, University of Navarra Clinic, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
| |
Collapse
|
37
|
Ghasemi Darestani N, Gilmanova AI, Al-Gazally ME, Zekiy AO, Ansari MJ, Zabibah RS, Jawad MA, Al-Shalah SAJ, Rizaev JA, Alnassar YS, Mohammed NM, Mustafa YF, Darvishi M, Akhavan-Sigari R. Mesenchymal stem cell-released oncolytic virus: an innovative strategy for cancer treatment. Cell Commun Signal 2023; 21:43. [PMID: 36829187 PMCID: PMC9960453 DOI: 10.1186/s12964-022-01012-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/10/2022] [Indexed: 02/26/2023] Open
Abstract
Oncolytic viruses (OVs) infect, multiply, and finally remove tumor cells selectively, causing no damage to normal cells in the process. Because of their specific features, such as, the ability to induce immunogenic cell death and to contain curative transgenes in their genomes, OVs have attracted attention as candidates to be utilized in cooperation with immunotherapies for cancer treatment. This treatment takes advantage of most tumor cells' inherent tendency to be infected by certain OVs and both innate and adaptive immune responses are elicited by OV infection and oncolysis. OVs can also modulate tumor microenvironment and boost anti-tumor immune responses. Mesenchymal stem cells (MSC) are gathering interest as promising anti-cancer treatments with the ability to address a wide range of cancers. MSCs exhibit tumor-trophic migration characteristics, allowing them to be used as delivery vehicles for successful, targeted treatment of isolated tumors and metastatic malignancies. Preclinical and clinical research were reviewed in this study to discuss using MSC-released OVs as a novel method for the treatment of cancer. Video Abstract.
Collapse
Affiliation(s)
| | - Anna I Gilmanova
- Department of Prosthetic Dentistry of the I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation
| | | | - Angelina O Zekiy
- Department of Prosthetic Dentistry of the I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russian Federation
| | - Mohammad Javed Ansari
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Rahman S Zabibah
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | | | - Saif A J Al-Shalah
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Iraq
| | - Jasur Alimdjanovich Rizaev
- Department of Public Health and Healthcare Management, Rector, Samarkand State Medical University, Samarkand, Uzbekistan
| | | | | | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | - Mohammad Darvishi
- Department of Aerospace and Subaquatic Medicine, Infectious Diseases and Tropical Medicine Research Center (IDTMRC), AJA University of Medical Sciences, Tehran, Iran.
| | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center, Tuebingen, Germany.,Department of Health Care Management and Clinical Research, Collegium Humanum Warsaw Management University, Warsaw, Poland
| |
Collapse
|
38
|
Improved Targeting of Therapeutics by Nanocarrier-Based Delivery in Cancer Immunotherapy and Their Future Perspectives. BIONANOSCIENCE 2023. [DOI: 10.1007/s12668-023-01065-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
39
|
Xue C, Chu Q, Zheng Q, Yuan X, Su Y, Bao Z, Lu J, Li L. Current understanding of the intratumoral microbiome in various tumors. Cell Rep Med 2023; 4:100884. [PMID: 36652905 PMCID: PMC9873978 DOI: 10.1016/j.xcrm.2022.100884] [Citation(s) in RCA: 20] [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: 06/25/2022] [Revised: 09/18/2022] [Accepted: 12/13/2022] [Indexed: 01/19/2023]
Abstract
It is estimated that in the future, the number of new cancer cases worldwide will exceed the 19.3 million recorded in 2020, and the number of deaths will exceed 10 million. Cancer remains the leading cause of human mortality and lagging socioeconomic development. Intratumoral microbes have been revealed to exist in many cancer types, including pancreatic, colorectal, liver, esophageal, breast, and lung cancers. Intratumoral microorganisms affect not only the host immune system, but also the effectiveness of tumor chemotherapy. This review concentrates on the characteristics and roles of intratumoral microbes in various tumors. In addition, the potential of therapies targeting intratumoral microbes, as well as the main challenges currently delaying these therapies, are explored. Furthermore, we briefly summarize existing technical methods used to characterize intratumoral microbes. We hope to provide ideas for exploring intratumoral microbes as potential biomarkers and targets for tumor diagnosis, treatment, and prognostication.
Collapse
Affiliation(s)
- Chen Xue
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Qingfei Chu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Qiuxian Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Xin Yuan
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yuanshuai Su
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Zhengyi Bao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
40
|
Intratumoral microbiota: roles in cancer initiation, development and therapeutic efficacy. Signal Transduct Target Ther 2023; 8:35. [PMID: 36646684 PMCID: PMC9842669 DOI: 10.1038/s41392-022-01304-4] [Citation(s) in RCA: 82] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/31/2022] [Accepted: 12/26/2022] [Indexed: 01/18/2023] Open
Abstract
Microorganisms, including bacteria, viruses, fungi, and other eukaryotes, play critical roles in human health. An altered microbiome can be associated with complex diseases. Intratumoral microbial components are found in multiple tumor tissues and are closely correlated with cancer initiation and development and therapy efficacy. The intratumoral microbiota may contribute to promotion of the initiation and progression of cancers by DNA mutations, activating carcinogenic pathways, promoting chronic inflammation, complement system, and initiating metastasis. Moreover, the intratumoral microbiota may not only enhance antitumor immunity via mechanisms including STING signaling activation, T and NK cell activation, TLS production, and intratumoral microbiota-derived antigen presenting, but also decrease antitumor immune responses and promote cancer progression through pathways including upregulation of ROS, promoting an anti-inflammatory environment, T cell inactivation, and immunosuppression. The effect of intratumoral microbiota on antitumor immunity is dependent on microbiota composition, crosstalk between microbiota and the cancer, and status of cancers. The intratumoral microbiota may regulate cancer cell physiology and the immune response by different signaling pathways, including ROS, β-catenin, TLR, ERK, NF-κB, and STING, among others. These viewpoints may help identify the microbiota as diagnosis or prognosis evaluation of cancers, and as new therapeutic strategy and potential therapeutic targets for cancer therapy.
Collapse
|
41
|
Wang G, Zhang Z, Zhong K, Wang Z, Yang N, Tang X, Li H, Lu Q, Wu Z, Yuan B, Zheng M, Cheng P, Tong A, Zhou L. CXCL11-armed oncolytic adenoviruses enhance CAR-T cell therapeutic efficacy and reprogram tumor microenvironment in glioblastoma. Mol Ther 2023; 31:134-153. [PMID: 36056553 PMCID: PMC9840126 DOI: 10.1016/j.ymthe.2022.08.021] [Citation(s) in RCA: 81] [Impact Index Per Article: 81.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/15/2022] [Accepted: 08/26/2022] [Indexed: 01/28/2023] Open
Abstract
Glioblastoma (GBM) is the most aggressive primary malignant brain cancer and urgently requires effective treatments. Chimeric antigen receptor T (CAR-T) cell therapy offers a potential treatment method, but it is often hindered by poor infiltration of CAR-T cells in tumors and highly immunosuppressive tumor microenvironment (TME). Here, we armed an oncolytic adenovirus (oAds) with a chemokine CXCL11 to increase the infiltration of CAR-T cells and reprogram the immunosuppressive TME, thus improving its therapeutic efficacy. In both immunodeficient and immunocompetent orthotopic GBM mice models, we showed that B7H3-targeted CAR-T cells alone failed to inhibit GBM growth but, when combined with the intratumoral administration of CXCL11-armed oAd, it achieved a durable antitumor response. Besides, oAd-CXCL11 had a potent antitumor effect and reprogramed the immunosuppressive TME in GL261 GBM models, in which increased infiltration of CD8+ T lymphocytes, natural killer (NK) cells, and M1-polarized macrophages, while decreased proportions of myeloid-derived suppressor cells (MDSCs), regulatory T cells (Tregs) and M2-polarized macrophages were observed. Furthermore, the antitumor effect of the oAd-CXCL11 was CD8+ T cell dependent. Our findings thus revealed that CXCL11-armed oAd can improve immune-virotherapy and can be a promising adjuvant of CAR-T therapy for GBM.
Collapse
Affiliation(s)
- Guoqing Wang
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, PR China
| | - Zongliang Zhang
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Kunhong Zhong
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Zeng Wang
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Nian Yang
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Xin Tang
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, PR China
| | - Hexian Li
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Qizhong Lu
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Zhiguo Wu
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Boyang Yuan
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, PR China
| | - Meijun Zheng
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, PR China
| | - Ping Cheng
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Aiping Tong
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| | - Liangxue Zhou
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
42
|
Mani I, Singh V. Receptor biology: Challenges and opportunities. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 196:337-349. [PMID: 36813364 DOI: 10.1016/bs.pmbts.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Receptor biology provides a great opportunity to understand the ligand-receptor signaling involved in health and disease processes. Receptor endocytosis and signaling play a vital role in health conditions. Receptor-based signaling is the main form of communication between cells and cells with the environment. However, if any irregularities happen during these events, the consequences of pathophysiological conditions occur. Various methods are utilized to know structure, function, and regulation of receptor proteins. Further, live-cell imaging and genetic manipulations have aided in the understanding of receptor internalization, subcellular trafficking, signaling, metabolic degradation, etc. Understanding the genetics, biochemistry, and physiology of receptors and ligands is very helpful to explore various aspects such as prognosis, diagnosis, and treatment of disease. However, there are enormous challenges that exist to explore receptor biology further. This chapter briefly discusses the current challenges and emerging opportunities of receptor biology.
Collapse
Affiliation(s)
- Indra Mani
- Department of Microbiology, Gargi College, University of Delhi, New Delhi, India.
| | - Vijai Singh
- Department of Biosciences, School of Science, Indrashil University, Rajpur, Mehsana, Gujarat, India
| |
Collapse
|
43
|
Del Baldo G, Del Bufalo F, Pinacchio C, Carai A, Quintarelli C, De Angelis B, Merli P, Cacchione A, Locatelli F, Mastronuzzi A. The peculiar challenge of bringing CAR-T cells into the brain: Perspectives in the clinical application to the treatment of pediatric central nervous system tumors. Front Immunol 2023; 14:1142597. [PMID: 37025994 PMCID: PMC10072260 DOI: 10.3389/fimmu.2023.1142597] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/09/2023] [Indexed: 04/08/2023] Open
Abstract
Childhood malignant brain tumors remain a significant cause of death in the pediatric population, despite the use of aggressive multimodal treatments. New therapeutic approaches are urgently needed for these patients in order to improve prognosis, while reducing side effects and long-term sequelae of the treatment. Immunotherapy is an attractive option and, in particular, the use of gene-modified T cells expressing a chimeric antigen receptor (CAR-T cells) represents a promising approach. Major hurdles in the clinical application of this approach in neuro-oncology, however, exist. The peculiar location of brain tumors leads to both a difficulty of access to the tumor mass, shielded by the blood-brain barrier (BBB), and to an increased risk of potentially life-threatening neurotoxicity, due to the primary location of the disease in the CNS and the low intracranial volume reserve. There are no unequivocal data on the best way of CAR-T cell administration. Multiple trials exploring the use of CD19 CAR-T cells for hematologic malignancies proved that genetically engineered T cells can cross the BBB, suggesting that systemically administered CAR-T cell can be used in the neuro-oncology setting. Intrathecal and intra-tumoral delivery can be easily managed with local implantable devices, suitable also for a more precise neuro-monitoring. The identification of specific approaches of neuro-monitoring is of utmost importance in these patients. In the present review, we highlight the most relevant potential challenges associated with the application of CAR-T cell therapy in pediatric brain cancers, focusing on the evaluation of the best route of delivery, the peculiar risk of neurotoxicity and the related neuro-monitoring.
Collapse
Affiliation(s)
- Giada Del Baldo
- Department of Pediatric Haematology and Oncology, and Cell and Gene Therapy Bambino Gesù Children’s Hospital, Scientific Institute for Reasearch, Hospitalization and Healthcare (IRCCS), Rome, Italy
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Francesca Del Bufalo
- Department of Pediatric Haematology and Oncology, and Cell and Gene Therapy Bambino Gesù Children’s Hospital, Scientific Institute for Reasearch, Hospitalization and Healthcare (IRCCS), Rome, Italy
| | - Claudia Pinacchio
- Department of Pediatric Haematology and Oncology, and Cell and Gene Therapy Bambino Gesù Children’s Hospital, Scientific Institute for Reasearch, Hospitalization and Healthcare (IRCCS), Rome, Italy
| | - Andrea Carai
- Department of Neurosciences, Neurosurgery Unit, Bambino Gesù Children’s Hospital, Scientific Institute for Reasearch, Hospitalization and Healthcare (IRCCS), Rome, Italy
| | - Concetta Quintarelli
- Department of Pediatric Haematology and Oncology, and Cell and Gene Therapy Bambino Gesù Children’s Hospital, Scientific Institute for Reasearch, Hospitalization and Healthcare (IRCCS), Rome, Italy
| | - Biagio De Angelis
- Department of Pediatric Haematology and Oncology, and Cell and Gene Therapy Bambino Gesù Children’s Hospital, Scientific Institute for Reasearch, Hospitalization and Healthcare (IRCCS), Rome, Italy
| | - Pietro Merli
- Department of Pediatric Haematology and Oncology, and Cell and Gene Therapy Bambino Gesù Children’s Hospital, Scientific Institute for Reasearch, Hospitalization and Healthcare (IRCCS), Rome, Italy
| | - Antonella Cacchione
- Department of Pediatric Haematology and Oncology, and Cell and Gene Therapy Bambino Gesù Children’s Hospital, Scientific Institute for Reasearch, Hospitalization and Healthcare (IRCCS), Rome, Italy
| | - Franco Locatelli
- Department of Pediatric Haematology and Oncology, and Cell and Gene Therapy Bambino Gesù Children’s Hospital, Scientific Institute for Reasearch, Hospitalization and Healthcare (IRCCS), Rome, Italy
- Department of Life Sciences and Public Health, Catholic University of the Sacred Heart, Rome, Italy
| | - Angela Mastronuzzi
- Department of Pediatric Haematology and Oncology, and Cell and Gene Therapy Bambino Gesù Children’s Hospital, Scientific Institute for Reasearch, Hospitalization and Healthcare (IRCCS), Rome, Italy
- *Correspondence: Angela Mastronuzzi,
| |
Collapse
|
44
|
Muscolini M, Hiscott J, Tassone E. A Genome-Wide CRISPR-Cas9 Loss-of-Function Screening to Identify Host Restriction Factors Modulating Oncolytic Virotherapy. Methods Mol Biol 2023; 2589:379-399. [PMID: 36255638 DOI: 10.1007/978-1-0716-2788-4_25] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Oncolytic virotherapy represents an efficient immunotherapeutic approach for cancer treatment. Oncolytic viruses (OVs) promote antitumor responses through tumor-selective cell lysis and immune system activation. However, some tumor cell lines and primary tumors display resistance to therapy. Here we describe a protocol to identify novel host factors responsible for tumor resistance to oncolysis using an unbiased genome-wide CRISPR-Cas9 loss-of-function screening. Cas9-expressing tumor cells are transduced with a library of pooled single-guide RNA (sgRNA)-expressing lentiviruses that target all human genes to obtain a cell population where each cell is knocked out for a single gene. Upon OV infection, resistant cells survive, while sensitive cells die. The relative abundance of each genome-integrated sgRNA is measured by next-generation sequencing (NGS) in resistant and control cells. This protocol is amenable to uncover host factors involved in the resistance to different OVs in multiple tumor models.
Collapse
Affiliation(s)
| | - John Hiscott
- Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Evelyne Tassone
- Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy.
| |
Collapse
|
45
|
Fang C, Xiao G, Wang T, Song L, Peng B, Xu B, Zhang K. Emerging Nano-/Biotechnology Drives Oncolytic Virus-Activated and Combined Cancer Immunotherapy. RESEARCH 2023; 6:0108. [PMID: 37040283 PMCID: PMC10079287 DOI: 10.34133/research.0108] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 03/15/2023] [Indexed: 04/05/2023]
Abstract
Oncolytic viruses (OVs) as one promising antitumor methods have made important contributions to tumor immunotherapy, which arouse increasing attention. They provide the dual mechanisms including direct killing effect toward tumor cells and immune activation for elevating antitumor responses, which have been proved in many preclinical studies. Especially, natural or genetically modified viruses as clinical immune preparations have emerged as a new promising approach objective to oncology treatment. The approval of talimogene laherparepvec (T-VEC) by the U.S. Food and Drug Administration (FDA) for the therapy of advanced melanoma could be considered as a milestone achievement in the clinical translation of OV. In this review, we first discussed the antitumor mechanisms of OVs with an emphasis on targeting, replication, and propagation. We further outlined the state of the art of current OVs in tumor and underlined the activated biological effects especially including immunity. More significantly, the enhanced immune responses based on OVs were systematically discussed from different perspectives such as combination with immunotherapy, genetic engineering of OVs, integration with nanobiotechnology or nanoparticles, and antiviral response counteraction, where their principles were shed light on. The development of OVs in the clinics was also highlighted to analyze the actuality and concerns of different OV applications in clinical trials. At last, the future perspectives and challenges of OVs as an already widely accepted treatment means were discussed. This review will provide a systematic review and deep insight into OV development and also offer new opportunities and guidance pathways to drive the further clinical translation.
Collapse
Affiliation(s)
- Chao Fang
- Central Laboratory and Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine,
Tongji University, No. 301 Yan-chang-zhong Road, Shanghai 200072, China
| | - Gaozhe Xiao
- National Center for International Research of Bio-targeting Theranostics,
Guangxi Medical University, No. 22 Shuangyong Road 22, Nanning, Guangxi 530021, China
| | - Taixia Wang
- Central Laboratory and Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine,
Tongji University, No. 301 Yan-chang-zhong Road, Shanghai 200072, China
| | - Li Song
- Central Laboratory and Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine,
Tongji University, No. 301 Yan-chang-zhong Road, Shanghai 200072, China
| | - Bo Peng
- Central Laboratory and Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine,
Tongji University, No. 301 Yan-chang-zhong Road, Shanghai 200072, China
| | - Bin Xu
- Department of Urology, Shanghai Ninth People’s Hospital,
Shanghai Jiaotong University School of Medicine, No. 639 Zhizaoju Road, Huangpu, Shanghai 200011, China
| | - Kun Zhang
- Central Laboratory and Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine,
Tongji University, No. 301 Yan-chang-zhong Road, Shanghai 200072, China
- National Center for International Research of Bio-targeting Theranostics,
Guangxi Medical University, No. 22 Shuangyong Road 22, Nanning, Guangxi 530021, China
| |
Collapse
|
46
|
Li S, Li Q, Ren Y, Yi J, Guo J, Kong X. HSV: The scout and assault for digestive system tumors. Front Mol Biosci 2023; 10:1142498. [PMID: 36926680 PMCID: PMC10011716 DOI: 10.3389/fmolb.2023.1142498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 02/15/2023] [Indexed: 03/08/2023] Open
Abstract
More than 25% of all malignant tumors are digestive system tumors (DSTs), which mostly include esophageal cancer, gastric cancer, pancreatic cancer, liver cancer, gallbladder cancer and cholangiocarcinoma, and colorectal cancer. DSTs have emerged as one of the prominent reasons of morbidity and death in many nations and areas around the world, posing a serious threat to human life and health. General treatments such as radiotherapy, chemotherapy, and surgical resection can poorly cure the patients and have a bad prognosis. A type of immunotherapy known as oncolytic virus therapy, have recently shown extraordinary anti-tumor effectiveness. One of the viruses that has been the subject of the greatest research in this field, the herpes simplex virus (HSV), has shown excellent potential in DSTs. With a discussion of HSV-1 based on recent studies, we outline the therapeutic effects of HSV on a number of DSTs in this review. Additionally, the critical function of HSV in the detection of cancers is discussed, and some HSV future possibilities are shown.
Collapse
Affiliation(s)
- Sheng Li
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qingbo Li
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yi Ren
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jia Yi
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jinhe Guo
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xianbin Kong
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
47
|
Omole RK, Oluwatola O, Akere MT, Eniafe J, Agboluaje EO, Daramola OB, Ayantunji YJ, Omotade TI, Torimiro N, Ayilara MS, Adeyemi OI, Salinsile OS. Comprehensive assessment on the applications of oncolytic viruses for cancer immunotherapy. Front Pharmacol 2022; 13:1082797. [PMID: 36569326 PMCID: PMC9772532 DOI: 10.3389/fphar.2022.1082797] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 11/24/2022] [Indexed: 12/13/2022] Open
Abstract
The worldwide burden of cancers is increasing at a very high rate, including the aggressive and resistant forms of cancers. Certain levels of breakthrough have been achieved with the conventional treatment methods being used to treat different forms of cancers, but with some limitations. These limitations include hazardous side effects, destruction of non-tumor healthy cells that are rapidly dividing and developing, tumor resistance to anti-cancer drugs, damage to tissues and organs, and so on. However, oncolytic viruses have emerged as a worthwhile immunotherapeutic option for the treatment of different types of cancers. In this treatment approach, oncolytic viruses are being modeled to target cancer cells with optimum cytotoxicity and spare normal cells with optimal safety, without the oncolytic viruses themselves being killed by the host immune defense system. Oncolytic viral infection of the cancer cells are also being genetically manipulated (either by removal or addition of certain genes into the oncolytic virus genome) to make the tumor more visible and available for attack by the host immune cells. Hence, different variants of these viruses are being developed to optimize their antitumor effects. In this review, we examined how grave the burden of cancer is on a global level, particularly in sub-Saharan Africa, major conventional therapeutic approaches to the treatment of cancer and their individual drawbacks. We discussed the mechanisms of action employed by these oncolytic viruses and different viruses that have found their relevance in the fight against various forms of cancers. Some pre-clinical and clinical trials that involve oncolytic viruses in cancer management were reported. This review also examined the toxicity and safety concerns surrounding the adoption of oncolytic viro-immunotherapy for the treatment of cancers and the likely future directions for researchers and general audience who wants updated information.
Collapse
Affiliation(s)
- Richard Kolade Omole
- Department of Microbiology, Obafemi Awolowo University, Ile-Ife, Nigeria,Microbiology Unit, Department of Applied Sciences, Osun State College of Technology, Esa-Oke, Nigeria,*Correspondence: Richard Kolade Omole,
| | - Oluwaseyi Oluwatola
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL, United States,Department of Immunology, Moffit Cancer Center, Tampa, FL, United States
| | - Millicent Tambari Akere
- Department of Medicinal and Biological Chemistry, University of Toledo, Toledo, OH, United States
| | - Joseph Eniafe
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | | | | | - Yemisi Juliet Ayantunji
- Department of Microbiology, Obafemi Awolowo University, Ile-Ife, Nigeria,Advanced Space Technology Applications Laboratory, Cooperative Information Network, National Space Research and Development Agency, Ile-Ife, Nigeria
| | | | - Nkem Torimiro
- Department of Microbiology, Obafemi Awolowo University, Ile-Ife, Nigeria
| | - Modupe Stella Ayilara
- Food Security and Safety Focus Area, Faculty of Natural and Agricultural Sciences, North-West University, Mmabatho, South Africa
| | - Oluwole Isaac Adeyemi
- Department of Pharmacology, Faculty of Pharmacy, Obafemi Awolowo University, Ile-Ife, Nigeria
| | | |
Collapse
|
48
|
Liu Y, Xu C, Xiao X, Chen Y, Wang X, Liu W, Tan Y, Zhu W, Hu J, Liang J, Yan G, Lin Y, Cai J. Overcoming resistance to oncolytic virus M1 by targeting PI3K-γ in tumor-associated myeloid cells. Mol Ther 2022; 30:3677-3693. [PMID: 35552024 PMCID: PMC9734023 DOI: 10.1016/j.ymthe.2022.05.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 04/21/2022] [Accepted: 05/07/2022] [Indexed: 02/07/2023] Open
Abstract
Oncolytic viruses (OVs) have become a category of promising anticancer immunotherapeutic agents over the last decade. However, the fact that many individuals fail to respond to OVs highlights the importance of defining the barely known immunosuppressive mechanisms that lead to treatment resistance. Here we found that the immunosuppression mediated by tumor-associated myeloid cells (TAMCs) directly quenches the antitumor effect of oncolytic virus M1 (OVM). OVM induces myeloid cells to migrate into tumors and strengthens their immunosuppressive phenotypes. Mechanically, tumor cells treated with OVM secrete interleukin-6 (IL-6) to activate the phosphatidylinositol 3-kinase (PI3K)-γ/Akt axis in TAMCs, promoting infiltration of TAMCs and aggravating their inhibition on cytotoxic CD8+ T lymphocytes. Pharmacologically targeting PI3K-γ relieves TAMC-mediated immunosuppression and enhances the efficacy of OVM. Additional treatment with immune checkpoint antibodies eradicates multiple refractory solid tumors and induces potent long-term antitumor immune memory. Our findings indicate that OVM functions as a double-edged sword in antitumor immunity and provide insights into the rationale for liberating T cell-mediated antitumor activity by abolishing TAMC-mediated immunosuppression.
Collapse
Affiliation(s)
- Yang Liu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Cuiying Xu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiaoting Xiao
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Yinting Chen
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiaobo Wang
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Wenfeng Liu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Yaqian Tan
- The Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou 510080, China
| | - Wenbo Zhu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Jun Hu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Jiankai Liang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Guangmei Yan
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Yuan Lin
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.
| | - Jing Cai
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
49
|
Muhi Rasoul L, Ali Marhoon A, Fadhel Abbas Albaayit S, Waleed Ali R, Hadi Saleh T, Abdullah Laftah Al-Rubaii B. Cytotoxic effect of cloned EGFP gene on NCI-H727 cell line via genetically engineered gene transfer system. Biomedicine (Taipei) 2022. [DOI: 10.51248/.v42i5.1934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Introduction and Aim: Cancers are a complex group of genetic illnesses that develop through multistep, mutagenic processes which can invade or spread throughout the body. Recent advances in cancer treatment involve oncolytic viruses to infect and destroy cancer cells. The Newcastle disease virus (NDV), an oncolytic virus has shown to have anti-cancer effects either directly by lysing cancer cells or indirectly by activating the immune system. The green fluorescent protein (GFP) has been widely used in studying the anti-tumor activity of oncolytic viruses. This study aimed to study the anticancer effect of a recombinant rNDV-GFP clone on NCI-H727 lung carcinoma cell line in vitro.
Materials and Methods: The GFP gene was inserted to a NDV strain to create a recombinant NDV (rNDV- GFP) using reverse genetics technology. The MTT assay was used in evaluating the oncolytic effect of rNDV- GFP on the lung carcinoma NCI-H727 cells. Light and fluorescent microscopy was used to study the cytopathic effects of rNDV-GFP.
Results: MTT assay showed that rNDV-GPF inhibited the NCI-H727 tumor cell death in a time-dependent manner. A significant inhibitory effect (78.3%) for rNDV-GPF on cancer cells was observed at 96h in comparison to rNDV (22.7%) and the cytotoxicity rate was directly proportional to the MOI used. Microscopic studies showed rNDV-GPF to induce cytopathic effect post 24 h of infection.
Conclusion: The GFP-expressing recombinant NDV strains exhibited encouraging results in terms of tumor growth inhibition. Our research set the groundwork for employing recombinant NDV as an anticancer viral vector.
Collapse
|
50
|
Adenovirus-Inspired Virus-Like-Particles Displaying Melanoma Tumor Antigen Specifically Target Human DC Subsets and Trigger Antigen-Specific Immune Responses. Biomedicines 2022; 10:biomedicines10112881. [DOI: 10.3390/biomedicines10112881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/24/2022] [Accepted: 11/02/2022] [Indexed: 11/12/2022] Open
Abstract
Virus-like particles constitute versatile vectors that can be used as vaccine platforms in many fields from infectiology and more recently to oncology. We previously designed non-infectious adenovirus-inspired 60-mer dodecahedric virus-like particles named ADDomers displaying on their surface either a short epitope or a large tumor/viral antigen. In this work, we explored for the first time the immunogenicity of ADDomers exhibiting melanoma-derived tumor antigen/epitope and their impact on the features of human dendritic cell (DC) subsets. We first demonstrated that ADDomers displaying tumor epitope/antigen elicit a strong immune-stimulating potential of human DC subsets (cDC2s, cDC1s, pDCs), which were able to internalize and cross-present tumor antigen, and subsequently cross-prime antigen-specific T-cell responses. To further limit off-target effects and enhance DC targeting, we engineered specific motifs to de-target epithelial cells and improve DCs’ addressing. The improved engineered platform making it possible to display large antigen represents a tool to overcome the barrier of immune allele restriction, broadening the immune response, and paving the way to its potential utilization in humans as an off-the-shelf vaccine.
Collapse
|