1
|
Salimi Asl A, Davari M, Ghorbani A, Seddighi N, Arabi K, Saburi E. Neoadjuvant immunotherapy and oncolytic virotherapy in HPV positive and HPV negative skin cancer: A comprehensive review. Int Immunopharmacol 2025; 146:113790. [PMID: 39673996 DOI: 10.1016/j.intimp.2024.113790] [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: 10/07/2024] [Revised: 12/03/2024] [Accepted: 12/03/2024] [Indexed: 12/16/2024]
Abstract
Skin cancer is the most common new cancer among Caucasians. This cancer has different types, of which non-melanoma skin cancer is the most common type. Various factors affect this disease, one of which is viral infections, including HPV. This virus plays an important role in skin cancer, especially cSCCs. There are various options for the treatment of skin cancer, and today special attention has been paid to treatments based on therapeutic goals, immunotherapy and combination therapy. In this study, we have investigated treatments based on immunotherapy and virotherapy and the effect of HPV virus on the effectiveness of these treatments in skin cancer. Treatments based on virotherapy are performed for a long time in combination with other common treatments such as radiotherapy and chemotherapy in order to have a greater effect and lower its side effects, which include: shortness of breath, tachycardia, lowering blood pressure in the patient. Also, the most important axis of immunotherapy is to focus on PD1-PDL1, despite abundant evidence on the importance of immunotherapy, many studies investigate the use of immunotherapy inhibitors in the adjuvant and neoadjuvant setting in various cancers. Also, previous findings show conflicting evidence of the effect of HPV status on the response to immunotherapy.
Collapse
Affiliation(s)
- Ali Salimi Asl
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Mohsen Davari
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Atousa Ghorbani
- Department of Biology, North Tehran Branch, Islamic Azad University, Tehran, Iran; Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran.
| | - Narjes Seddighi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Kimia Arabi
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran.
| | - Ehsan Saburi
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Genetics and Molecular Medicine Department, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Rong Y, Ning Y, Zhu J, Feng P, Zhu W, Zhao X, Xiong Z, Ruan C, Jin J, Wang H, Cai T, Zhang S, Yang Y. Oncolytic adenovirus encoding decorin and CD40 ligand inhibits tumor growth and liver metastasis via immune activation in murine colorectal tumor model. MOLECULAR BIOMEDICINE 2024; 5:39. [PMID: 39306655 PMCID: PMC11416448 DOI: 10.1186/s43556-024-00202-1] [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: 01/22/2024] [Accepted: 08/20/2024] [Indexed: 09/25/2024] Open
Abstract
Colorectal cancer (CRC) is the second common cause of cancer mortality worldwide, and it still lacks effective approaches for relapsed and metastatic CRC. Recently, oncolytic virus has been emerged as a promising immune therapeutic strategy. In this study, we develop a novel oncolytic adenovirus, rAd.mDCN.mCD40L, which drive oncolytic activity by telomerase reverse transcriptase promoter (TERTp). rAd.mDCN.mCD40L expressed both mouse genes of decorin (mDCN) and CD40 ligand (mCD40L), and produced effective cytotoxicity in both human and mouse CRC cells. Moreover, oncolytic adenovirus mediated mDCN over-expression inhibited Met expression in vitro. In CT26 subcutaneous tumor model, intratumorally delivery of oncolytic adenoviruses could inhibit tumor growth and liver metastasis, while mDCN and/or mCD40L armed oncolytic adenoviruses produced much more impressive responses. No obvious toxicity was detected in lung, liver and spleen. Moreover, mDCN and/or mCD40L armed oncolytic adenoviruses altered the immune state to activate anti-tumor responses, including increasing CD8+ T effector cells and CD4+ memory T cells, reducing MDSCs and Tregs in peripheral blood. Furthermore, mDCN and/or mCD40L armed oncolytic adenoviruses mediated mDCN and/or mCD40L expression in tumors, and up-regulated Th1 cytokines and reduced Th2 cytokines in tumors, which will be benefit for remodeling tumor microenvironment. Importantly, rAd.mDCN.mCD40L and rAd.mCD40L prevented tumor liver metastasis much more effectively than rAd.Null and rAd.mDCN. Therefore, rAd.mDCN.mCD40L and rAd.mCD40L are promising approaches for CRC therapy.
Collapse
Affiliation(s)
- Yejing Rong
- Department of Experimental Medical Science, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Yingjun Ning
- Department of Experimental Medical Science, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Jianping Zhu
- Department of Pharmacy, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Pei Feng
- Ningbo Qianyang Talent Service Co., Ltd, Ningbo, 315020, China
| | - Weixin Zhu
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, 315032, China
| | - Xin Zhao
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, 315032, China
| | - Zi Xiong
- Department of Experimental Medical Science, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Chunyan Ruan
- Department of Experimental Medical Science, Ningbo No.2 Hospital, Ningbo, 315010, China
| | - Jiachang Jin
- Jiangbei Center For Disease Control and Prevention Ningbo, Ningbo, 315020, China
| | - Hua Wang
- Department of Experimental Haematology, Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing, 100850, China
| | - Ting Cai
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, 315032, China.
| | - Shun Zhang
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo, 315032, China.
| | - Yuefeng Yang
- Department of Experimental Medical Science, Ningbo No.2 Hospital, Ningbo, 315010, China.
| |
Collapse
|
3
|
Kato Y, Rice N, Pokrass M, Jeong J, Rodriguez R, Field JJ, Nowyhed H. Nonclinical characterization of ICVB-1042 as a selective oncolytic adenovirus for solid tumor treatment. Commun Biol 2024; 7:1132. [PMID: 39271928 PMCID: PMC11399272 DOI: 10.1038/s42003-024-06839-6] [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: 01/08/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
ICVB-1042 is an oncolytic adenovirus containing modifications to enhance replication, lysis, and viral spreading in tumor cells. The anti-tumor activity, immune activation, tropism, selectivity, and mechanism of action were evaluated in preparation for a first-in-human study. ICVB-1042 was at least 100-fold more cytotoxic in A549 cells than in normal primary cells tested, demonstrating its high tumor selectivity and a low likelihood of targeting primary tissues. ICVB-1042 administered to mice intravenously or intratumorally was effective in reducing tumor burden. Its intravenous administration also inhibited tumor growth in orthotopic models. ICVB-1042 was well tolerated in mice compared to HAdV-C5 (Wt Ad5), with reduced liver sequestration, supporting safety of the drug for systemic delivery. These preclinical data demonstrating the safety and potency of ICVB-1042 for treatment of various solid tumors support the ongoing clinical investigation (NCT05904236).
Collapse
Affiliation(s)
- Yu Kato
- IconOVir Bio, New York, NY, USA
| | | | | | | | | | | | | |
Collapse
|
4
|
Gujar S, Pol JG, Kumar V, Lizarralde-Guerrero M, Konda P, Kroemer G, Bell JC. Tutorial: design, production and testing of oncolytic viruses for cancer immunotherapy. Nat Protoc 2024; 19:2540-2570. [PMID: 38769145 DOI: 10.1038/s41596-024-00985-1] [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: 05/10/2023] [Accepted: 02/12/2024] [Indexed: 05/22/2024]
Abstract
Oncolytic viruses (OVs) represent a novel class of cancer immunotherapy agents that preferentially infect and kill cancer cells and promote protective antitumor immunity. Furthermore, OVs can be used in combination with established or upcoming immunotherapeutic agents, especially immune checkpoint inhibitors, to efficiently target a wide range of malignancies. The development of OV-based therapy involves three major steps before clinical evaluation: design, production and preclinical testing. OVs can be designed as natural or engineered strains and subsequently selected for their ability to kill a broad spectrum of cancer cells rather than normal, healthy cells. OV selection is further influenced by multiple factors, such as the availability of a specific viral platform, cancer cell permissivity, the need for genetic engineering to render the virus non-pathogenic and/or more effective and logistical considerations around the use of OVs within the laboratory or clinical setting. Selected OVs are then produced and tested for their anticancer potential by using syngeneic, xenograft or humanized preclinical models wherein immunocompromised and immunocompetent setups are used to elucidate their direct oncolytic ability as well as indirect immunotherapeutic potential in vivo. Finally, OVs demonstrating the desired anticancer potential progress toward translation in patients with cancer. This tutorial provides guidelines for the design, production and preclinical testing of OVs, emphasizing considerations specific to OV technology that determine their clinical utility as cancer immunotherapy agents.
Collapse
Affiliation(s)
- Shashi Gujar
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Biology, Dalhousie University, Halifax, Nova Scotia, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada
| | - Jonathan G Pol
- INSERM, U1138, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Cité, Paris, France
- Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, UMS AMICCa, Gustave Roussy, Villejuif, France
| | - Vishnupriyan Kumar
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada
| | - Manuela Lizarralde-Guerrero
- INSERM, U1138, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Cité, Paris, France
- Sorbonne Université, Paris, France
- Metabolomics and Cell Biology Platforms, UMS AMICCa, Gustave Roussy, Villejuif, France
- Ecole Normale Supérieure de Lyon, Lyon, France
| | - Prathyusha Konda
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Harvard University, Boston, MA, USA
| | - Guido Kroemer
- INSERM, U1138, Paris, France.
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.
- Université Paris Cité, Paris, France.
- Sorbonne Université, Paris, France.
- Metabolomics and Cell Biology Platforms, UMS AMICCa, Gustave Roussy, Villejuif, France.
- Institut Universitaire de France, Paris, France.
- Institut du Cancer Paris CARPEM, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| | - John C Bell
- Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
- Department of Biochemistry, Microbiology & Immunology, University of Ottawa, Ottawa, Ontario, Canada.
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.
| |
Collapse
|
5
|
Baabdulla AA, Hillen T. Oscillations in a Spatial Oncolytic Virus Model. Bull Math Biol 2024; 86:93. [PMID: 38896363 DOI: 10.1007/s11538-024-01322-z] [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: 12/19/2023] [Accepted: 05/31/2024] [Indexed: 06/21/2024]
Abstract
Virotherapy treatment is a new and promising target therapy that selectively attacks cancer cells without harming normal cells. Mathematical models of oncolytic viruses have shown predator-prey like oscillatory patterns as result of an underlying Hopf bifurcation. In a spatial context, these oscillations can lead to different spatio-temporal phenomena such as hollow-ring patterns, target patterns, and dispersed patterns. In this paper we continue the systematic analysis of these spatial oscillations and discuss their relevance in the clinical context. We consider a bifurcation analysis of a spatially explicit reaction-diffusion model to find the above mentioned spatio-temporal virus infection patterns. The desired pattern for tumor eradication is the hollow ring pattern and we find exact conditions for its occurrence. Moreover, we derive the minimal speed of travelling invasion waves for the cancer and for the oncolytic virus. Our numerical simulations in 2-D reveal complex spatial interactions of the virus infection and a new phenomenon of a periodic peak splitting. An effect that we cannot explain with our current methods.
Collapse
Affiliation(s)
- Arwa Abdulla Baabdulla
- Department of Mathematical and Statistical Sciences, University of Alberta, Edmonton, Canada.
| | - Thomas Hillen
- Department of Mathematical and Statistical Sciences, University of Alberta, Edmonton, Canada
| |
Collapse
|
6
|
Shirazi MMA, Saedi TA, Moghaddam ZS, Nemati M, Shiri R, Negahdari B, Goradel NH. Nanotechnology and nano-sized tools: Newer approaches to circumvent oncolytic adenovirus limitations. Pharmacol Ther 2024; 256:108611. [PMID: 38387653 DOI: 10.1016/j.pharmthera.2024.108611] [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: 10/19/2023] [Revised: 01/03/2024] [Accepted: 02/14/2024] [Indexed: 02/24/2024]
Abstract
Oncolytic adenoviruses (OAds), engineered Ads preferentially infect and lyse tumor cells, have attracted remarkable attention as immunotherapy weapons for the treatment of various malignancies. Despite hopeful successes in preclinical investigations and translation into clinical phases, they face some challenges that thwart their therapeutic effectiveness, including low infectivity of cancer cells, liver sequestration, pre-existing neutralizing antibodies, physical barriers to the spread of Ads, and immunosuppressive TME. Nanotechnology and nano-sized tools provide several advantages to overcome these limitations of OAds. Nano-sized tools could improve the therapeutic efficacy of OAds by enhancing infectivity and cellular uptake, targeting and protecting from pre-existing immune responses, masking and preventing liver tropism, and co-delivery with other therapeutic agents. Herein, we reviewed the constructs of various OAds and their application in clinical trials, as well as the limitations they have faced. Furthermore, we emphasized the potential applications of nanotechnology to solve the constraints of OAds to improve their anti-tumor activities.
Collapse
Affiliation(s)
| | - Tayebeh Azam Saedi
- Department of Genetics, Faculty of Science, Islamic Azad University, Tonekabon Branch, Tonekabon, Iran
| | - Zahra Samadi Moghaddam
- Department of Medical Biotechnology, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Mahnaz Nemati
- Amir Oncology Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Shiri
- Department of Basic Sciences, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Babak Negahdari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nasser Hashemi Goradel
- Department of Medical Biotechnology, Maragheh University of Medical Sciences, Maragheh, Iran; Arthropod-Borne Diseases Research Centre, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
7
|
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
|
8
|
Zeng M, Zhang W, Li Y, Yu L. Harnessing adenovirus in cancer immunotherapy: evoking cellular immunity and targeting delivery in cell-specific manner. Biomark Res 2024; 12:36. [PMID: 38528632 DOI: 10.1186/s40364-024-00581-1] [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: 12/07/2023] [Accepted: 03/09/2024] [Indexed: 03/27/2024] Open
Abstract
Recombinant adenovirus (rAd) regimens, including replication-competent oncolytic adenovirus (OAV) and replication-deficient adenovirus, have been identified as potential cancer therapeutics. OAV presents advantages such as selective replication, oncolytic efficacy, and tumor microenvironment (TME) remodeling. In this perspective, the principles and advancements in developing OAV toolkits are reviewed. The burgeoning rAd may dictate efficacy of conventional cancer therapies as well as cancer immunotherapies, including cancer vaccines, synergy with adoptive cell therapy (ACT), and TME reshaping. Concurrently, we explored the potential of rAd hitchhiking to adoptive immune cells or stem cells, highlighting how this approach facilitates synergistic interactions between rAd and cellular therapeutics at tumor sites. Results from preclinical and clinical trials in which immune and stem cells were infected with rAd have been used to address significant oncological challenges, such as postsurgical residual tumor tissue and metastatic tissue. Briefly, rAd can eradicate tumors through various mechanisms, resulting from tumor immunogenicity, reprogramming of the TME, enhancement of cellular immunity, and effective tumor targeting. In this context, we argue that rAd holds immense potential for enhancing cellular immunity and synergistically improving antitumor effects in combination with novel cancer immunotherapies.
Collapse
Affiliation(s)
- Miao Zeng
- Department of Hematology and Oncology, Shenzhen University General Hospital, International Cancer Center, Hematology Institution of Shenzhen University, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518000, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Wei Zhang
- Department of Hematology and Oncology, Shenzhen University General Hospital, International Cancer Center, Hematology Institution of Shenzhen University, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518000, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Yisheng Li
- Shenzhen Haoshi Biotechnology Co., Ltd. No, 155 Hongtian Road, Xinqiao Street, Bao'an District, Shenzhen, Guangdong, 518125, China.
| | - Li Yu
- Department of Hematology and Oncology, Shenzhen University General Hospital, International Cancer Center, Hematology Institution of Shenzhen University, Shenzhen University Medical School, Shenzhen University, Shenzhen, Guangdong, 518000, China.
| |
Collapse
|
9
|
Wang C, Lu N, Yan L, Li Y. The efficacy and safety assessment of oncolytic virotherapies in the treatment of advanced melanoma: a systematic review and meta-analysis. Virol J 2023; 20:252. [PMID: 37919738 PMCID: PMC10623758 DOI: 10.1186/s12985-023-02220-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: 01/15/2023] [Accepted: 10/28/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND The efficacy and safety of oncolytic virotherapies in the treatment of advanced melanoma still remains controversal. It is necessary to conduct quantitative evaluation on the basis of preclinical trial reports. METHODS Publicly available databases (PubMed, Embase, Medline, Web of Science and Cochrane Library.) and register (Clinicaltrials.gov) were searched to collect treatment outcomes of oncolytic virotherapies (including herpes simplex virus type 1 (HSV), coxsackievirus A21 (CVA21), adenovirus, poxvirus and reovirus) for advanced/unresectable melanoma. Comparisons of treatment response, adverse events (AEs) and survival analyses for different virotherapies were performed by R software based on the extracted data from eligible studies. RESULTS Finally, thirty-four eligible studies were analysed and HSV virotherapy had the highest average complete response (CR, 24.8%) and HSV had a slightly higher average overall response rate (ORR) than CVA21 (43.8% vs 42.6%). In the pooled results of comparing talimogene laherparepve (T-VEC) with or without GM-CSF/ICIs (immune checkpoint inhibitors) to GM-CSF/ICIs monotherapy suggested virotherapy was more efficient in subgroups CR (RR = 1.80, 95% CI [1.30; 2.51], P < 0.01), ORR (RR = 1.17, 95% CI [1.02; 1.34], P < 0.05), and DCR (RR = 1.27, 95% CI [1.15; 1.40], P < 0.01). In patients treated with T-VEC+ICIs, 2-year overall survival (12.1 ± 6.9 months) and progression-free survival (9.9 ± 6.9) were significantly longer than those treated with T-VEC alone. Furthermore, we found that AEs occurred frequently in virotherapy but decreased in a large cohort of enrolled patients, some of which, such as abdominal distension/pain, injection site pain and pruritus, were found to be positively associated with disease progression in patients treated with T-VEC monotherapy. CONCLUSION Given the relative safety and tolerability of oncolytic viruses, and the lack of reports of dose-limiting-dependent toxicities, more patients treated with T-VEC with or without ICIs should be added to future assessment analyses. There is still a long way to go before it can be used as a first-line therapy for patients with advanced or unresectable melanoma.
Collapse
Affiliation(s)
- Changyuan Wang
- Department of Dermatology, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), NO.1 Jiaozhou Road, Qingdao, 266000, Shandong Province, China
| | - Nanxiao Lu
- Department of Dermatology, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), NO.1 Jiaozhou Road, Qingdao, 266000, Shandong Province, China
| | - Lin Yan
- Department of Dermatology, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), NO.1 Jiaozhou Road, Qingdao, 266000, Shandong Province, China
| | - Yang Li
- Department of Dermatology, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), NO.1 Jiaozhou Road, Qingdao, 266000, Shandong Province, China.
| |
Collapse
|
10
|
Hu M, Liao X, Tao Y, Chen Y. Advances in oncolytic herpes simplex virus and adenovirus therapy for recurrent glioma. Front Immunol 2023; 14:1285113. [PMID: 38022620 PMCID: PMC10652401 DOI: 10.3389/fimmu.2023.1285113] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
Recurrent glioma treatment is challenging due to molecular heterogeneity and treatment resistance commonly observed in these tumors. Researchers are actively pursuing new therapeutic strategies. Oncolytic viruses have emerged as a promising option. Oncolytic viruses selectively replicate within tumor cells, destroying them and stimulating the immune system for an enhanced anticancer response. Among Oncolytic viruses investigated for recurrent gliomas, oncolytic herpes simplex virus and oncolytic adenovirus show notable potential. Genetic modifications play a crucial role in optimizing their therapeutic efficacy. Different generations of replicative conditioned oncolytic human adenovirus and oncolytic HSV have been developed, incorporating specific modifications to enhance tumor selectivity, replication efficiency, and immune activation. This review article summarizes these genetic modifications, offering insights into the underlying mechanisms of Oncolytic viruses' therapy. It also aims to identify strategies for further enhancing the therapeutic benefits of Oncolytic viruses. However, it is important to acknowledge that additional research and clinical trials are necessary to establish the safety, efficacy, and optimal utilization of Oncolytic viruses in treating recurrent glioblastoma.
Collapse
Affiliation(s)
- Mingming Hu
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - XuLiang Liao
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yi Tao
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Yaohui Chen
- Institute of Thoracic Oncology, West China Hospital, Sichuan University, Chengdu, China
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
Kudling TV, Clubb JH, Pakola S, Quixabeira DC, Lähdeniemi IA, Heiniö C, Arias V, Havunen R, Cervera-Carrascon V, Santos JM, Sutinen E, Räsänen J, Borenius K, Mäyränpää MI, Aaltonen E, Sorsa S, Hemminki O, Kanerva A, Verschuren EW, Ilonen I, Hemminki A. Effective intravenous delivery of adenovirus armed with TNFα and IL-2 improves anti-PD-1 checkpoint blockade in non-small cell lung cancer. Oncoimmunology 2023; 12:2241710. [PMID: 37546696 PMCID: PMC10399490 DOI: 10.1080/2162402x.2023.2241710] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/24/2023] [Accepted: 07/24/2023] [Indexed: 08/08/2023] Open
Abstract
Lung cancer remains among the most difficult-to-treat malignancies and is the leading cause of cancer-related deaths worldwide. The introduction of targeted therapies and checkpoint inhibitors has improved treatment outcomes; however, most patients with advanced-stage non-small cell lung cancer (NSCLC) eventually fail these therapies. Therefore, there is a major unmet clinical need for checkpoint refractory/resistant NSCLC. Here, we tested the combination of aPD-1 and adenovirus armed with TNFα and IL-2 (Ad5-CMV-mTNFα/mIL-2) in an immunocompetent murine NSCLC model. Moreover, although local delivery has been standard for virotherapy, treatment was administered intravenously to facilitate clinical translation and putative routine use. We showed that treatment of tumor-bearing animals with aPD-1 in combination with intravenously injected armed adenovirus significantly decreased cancer growth, even in the presence of neutralizing antibodies. We observed an increased frequency of cytotoxic tumor-infiltrating lymphocytes, including tumor-specific cells. Combination treatment led to a decreased percentage of immunosuppressive tumor-associated macrophages and an improvement in dendritic cell maturation. Moreover, we observed expansion of the tumor-specific memory T cell compartment in secondary lymphoid organs in the group that received aPD-1 with the virus. However, although the non-replicative Ad5-CMV-mTNFα/mIL-2 virus allows high transgene expression in the murine model, it does not fully reflect the clinical outcome in humans. Thus, we complemented our findings using NSCLC ex vivo models fully permissive for the TNFα and IL-2- armed oncolytic adenovirus TILT-123. Overall, our data demonstrate the ability of systemically administered adenovirus armed with TNFα and IL-2 to potentiate the anti-tumor efficacy of aPD-1 and warrant further investigation in clinical trials.
Collapse
Affiliation(s)
- Tatiana V. Kudling
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - James H.A. Clubb
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Santeri Pakola
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Dafne C.A. Quixabeira
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Iris A.K. Lähdeniemi
- Translational Lung Cancer Research Group, Institute for Molecular Medicine Finland (FIMM), HiLife, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Camilla Heiniö
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Victor Arias
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Riikka Havunen
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Victor Cervera-Carrascon
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Joao M. Santos
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Eva Sutinen
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Pulmonary Medicine, Heart and Lung Center, Helsinki University Hospital, Helsinki, Finland
| | - Jari Räsänen
- General Thoracic and Esophageal Surgery, Heart and Lung Center, Helsinki University Hospital and Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Kristian Borenius
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- General Thoracic and Esophageal Surgery, Heart and Lung Center, Helsinki University Hospital and Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mikko I. Mäyränpää
- Pathology, University of Helsinki and Helsinki University Hospital (HUSLAB), Helsinki, Finland
| | - Eero Aaltonen
- Faculty of Medicine, Medicum, University of Helsinki, Helsinki, Finland
| | - Suvi Sorsa
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Otto Hemminki
- Comprehensive Cancer Center, Helsinki University Hospital (HUS), Helsinki, Finland
- Department of Urology, Helsinki University Hospital, Helsinki, Finland
| | - Anna Kanerva
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Comprehensive Cancer Center, Helsinki University Hospital (HUS), Helsinki, Finland
- Department of Gynecology and Obstetrics, Helsinki University Hospital, Helsinki, Finland
| | - Emmy W. Verschuren
- Translational Lung Cancer Research Group, Institute for Molecular Medicine Finland (FIMM), HiLife, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Ilkka Ilonen
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- General Thoracic and Esophageal Surgery, Heart and Lung Center, Helsinki University Hospital and Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
- Comprehensive Cancer Center, Helsinki University Hospital (HUS), Helsinki, Finland
| |
Collapse
|
12
|
Brănişteanu DE, Porumb-Andrese E, Porumb V, Stărică A, Moraru AD, Nicolescu AC, Zemba M, Brănişteanu CI, Brănişteanu G, Brănişteanu DC. New Treatment Horizons in Uveal and Cutaneous Melanoma. Life (Basel) 2023; 13:1666. [PMID: 37629523 PMCID: PMC10455832 DOI: 10.3390/life13081666] [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: 06/18/2023] [Revised: 07/22/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
Melanoma is a complex and heterogeneous malignant tumor with distinct genetic characteristics and therapeutic challenges in both cutaneous melanoma (CM) and uveal melanoma (UM). This review explores the underlying molecular features and genetic alterations in these melanoma subtypes, highlighting the importance of employing specific model systems tailored to their unique profiles for the development of targeted therapies. Over the past decade, significant progress has been made in unraveling the molecular and genetic characteristics of CM and UM, leading to notable advancements in treatment options. Genetic mutations in the mitogen-activated protein kinase (MAPK) pathway drive CM, while UM is characterized by mutations in genes like GNAQ, GNA11, BAP1, EIF1AX, and SF3B1. Chromosomal aberrations, including monosomy 3 in UM and monosomy 10 in CM, play significant roles in tumorigenesis. Immune cell infiltration differs between CM and UM, impacting prognosis. Therapeutic advancements targeting these genetic alterations, including oncolytic viruses and immunotherapies, have shown promise in preclinical and clinical studies. Oncolytic viruses selectively infect malignant cells, inducing oncolysis and activating antitumor immune responses. Talimogene laherparepvec (T-VEC) is an FDA-approved oncolytic virus for CM treatment, and other oncolytic viruses, such as coxsackieviruses and HF-10, are being investigated. Furthermore, combining oncolytic viruses with immunotherapies, such as CAR-T cell therapy, holds great potential. Understanding the intrinsic molecular features of melanoma and their role in shaping novel therapeutic approaches provides insights into targeted interventions and paves the way for more effective treatments for CM and UM.
Collapse
Affiliation(s)
- Daciana Elena Brănişteanu
- Department of Medical Specialties (III)-Dermatology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Railway Clinical Hospital, 700506 Iasi, Romania;
| | - Elena Porumb-Andrese
- Department of Medical Specialties (III)-Dermatology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Railway Clinical Hospital, 700506 Iasi, Romania;
| | - Vlad Porumb
- Department of Surgery, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
- Military Emergency Clinical Hospital “Dr. Iacob Czihac”, 700506 Iasi, Romania
| | | | - Andreea Dana Moraru
- Department of Ophthalmology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | | | - Mihail Zemba
- Department of Ophthalmology, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania;
| | | | - George Brănişteanu
- “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.I.B.); (G.B.)
| | - Daniel Constantin Brănişteanu
- Railway Clinical Hospital, 700506 Iasi, Romania;
- Department of Ophthalmology, Faculty of Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| |
Collapse
|
13
|
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: 1.5] [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
|
14
|
Zhu X, Fan C, Xiong Z, Chen M, Li Z, Tao T, Liu X. Development and application of oncolytic viruses as the nemesis of tumor cells. Front Microbiol 2023; 14:1188526. [PMID: 37440883 PMCID: PMC10335770 DOI: 10.3389/fmicb.2023.1188526] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/18/2023] [Indexed: 07/15/2023] Open
Abstract
Viruses and tumors are two pathologies that negatively impact human health, but what occurs when a virus encounters a tumor? A global consensus among cancer patients suggests that surgical resection, chemotherapy, radiotherapy, and other methods are the primary means to combat cancer. However, with the innovation and development of biomedical technology, tumor biotherapy (immunotherapy, molecular targeted therapy, gene therapy, oncolytic virus therapy, etc.) has emerged as an alternative treatment for malignant tumors. Oncolytic viruses possess numerous anti-tumor properties, such as directly lysing tumor cells, activating anti-tumor immune responses, and improving the tumor microenvironment. Compared to traditional immunotherapy, oncolytic virus therapy offers advantages including high killing efficiency, precise targeting, and minimal side effects. Although oncolytic virus (OV) therapy was introduced as a novel approach to tumor treatment in the 19th century, its efficacy was suboptimal, limiting its widespread application. However, since the U.S. Food and Drug Administration (FDA) approved the first OV therapy drug, T-VEC, in 2015, interest in OV has grown significantly. In recent years, oncolytic virus therapy has shown increasingly promising application prospects and has become a major research focus in the field of cancer treatment. This article reviews the development, classification, and research progress of oncolytic viruses, as well as their mechanisms of action, therapeutic methods, and routes of administration.
Collapse
Affiliation(s)
- Xiao Zhu
- Zhejiang Provincial People's Hospital Affiliated to Hangzhou Medical College, Hangzhou Medical College, Hangzhou, China
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
- Department of Biological and Chemical Sciences, New York Institute of Technology—Manhattan Campus, New York, NY, United States
| | - Chenyang Fan
- Department of Clinical Medicine, Medicine and Technology, School of Zunyi Medical University, Zunyi, China
| | - Zhuolong Xiong
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Mingwei Chen
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Zesong Li
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital(Shenzhen Institute of Translational Medicine), Shenzhen, China
| | - Tao Tao
- Department of Gastroenterology, Zibo Central Hospital, Zibo, China
| | - Xiuqing Liu
- Department of Clinical Laboratory, Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, China
| |
Collapse
|
15
|
Zolaly MA, Mahallawi W, Khawaji ZY, Alahmadi MA. The Clinical Advances of Oncolytic Viruses in Cancer Immunotherapy. Cureus 2023; 15:e40742. [PMID: 37485097 PMCID: PMC10361339 DOI: 10.7759/cureus.40742] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2023] [Indexed: 07/25/2023] Open
Abstract
A promising future for oncology treatment has been brought about by the emergence of a novel approach utilizing oncolytic viruses in cancer immunotherapy. Oncolytic viruses are viruses that have been exploited genetically to assault malignant cells and activate a robust immune response. Several techniques have been developed to endow viruses with an oncolytic activity through genetic engineering. For instance, redirection capsid modification, stimulation of anti-neoplastic immune response, and genetically arming viruses with cytokines such as IL-12. Oncolytic viral clinical outcomes are sought after, particularly in more advanced cancers. The effectiveness and safety profile of the oncolytic virus in clinical studies with or without the combination of standard treatment (chemotherapy, radiotherapy, or primary excision) has been assessed using response evaluation criteria in solid tumors (RECIST). This review will comprehensively outline the most recent clinical applications and provide the results from various phases of clinical trials in a variety of cancers in the latest published literature.
Collapse
Affiliation(s)
- Mohammed A Zolaly
- Pediatric Hematology Oncology, Taibah University, Al-Madinah al-Munawwarah, SAU
| | - Waleed Mahallawi
- Medical Laboratory Technology Department, College of Applied Medical Sciences, Taibah University, Al-Madinah al-Munawwarah, SAU
| | - Zakaria Y Khawaji
- Medicine and Surgery, Taibah University, Al-Madinah al-Munawwarah, SAU
| | | |
Collapse
|
16
|
Thoidingjam S, Sriramulu S, Freytag S, Brown SL, Kim JH, Chetty IJ, Siddiqui F, Movsas B, Nyati S. Oncolytic virus-based suicide gene therapy for cancer treatment: a perspective of the clinical trials conducted at Henry Ford Health. TRANSLATIONAL MEDICINE COMMUNICATIONS 2023; 8:11. [PMID: 37065938 PMCID: PMC10088621 DOI: 10.1186/s41231-023-00144-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 04/04/2023] [Indexed: 06/19/2023]
Abstract
Gene therapy manipulates or modifies a gene that provides a new cellular function to treat or correct a pathological condition, such as cancer. The approach of using gene manipulation to modify patient's cells to improve cancer therapy and potentially find a cure is gaining popularity. Currently, there are 12 gene therapy products approved by US-FDA, EMA and CFDA for cancer management, these include Rexin-G, Gendicine, Oncorine, Provange among other. The Radiation Biology Research group at Henry Ford Health has been actively developing gene therapy approaches for improving clinical outcome in cancer patients. The team was the first to test a replication-competent oncolytic virus armed with a therapeutic gene in humans, to combine this approach with radiation in humans, and to image replication-competent adenoviral gene expression/activity in humans. The adenoviral gene therapy products developed at Henry Ford Health have been evaluated in more than 6 preclinical studies and evaluated in 9 investigator initiated clinical trials treating more than100 patients. Two phase I clinical trials are currently following patients long term and a phase I trial for recurrent glioma was initiated in November 2022. This systematic review provides an overview of gene therapy approaches and products employed for treating cancer patients including the products developed at Henry Ford Health.
Collapse
Affiliation(s)
- Shivani Thoidingjam
- Department of Radiation Oncology, Henry Ford Health, 1 Ford Place, 5D-42, Detroit, MI 48202 USA
| | - Sushmitha Sriramulu
- Department of Radiation Oncology, Henry Ford Health, 1 Ford Place, 5D-42, Detroit, MI 48202 USA
| | - Svend Freytag
- Department of Radiation Oncology, Henry Ford Health, 1 Ford Place, 5D-42, Detroit, MI 48202 USA
| | - Stephen L. Brown
- Department of Radiation Oncology, Henry Ford Health, 1 Ford Place, 5D-42, Detroit, MI 48202 USA
- College of Human Medicine, Michigan State University, East Lansing, MI 48824 USA
| | - Jae Ho Kim
- Department of Radiation Oncology, Henry Ford Health, 1 Ford Place, 5D-42, Detroit, MI 48202 USA
| | - Indrin J. Chetty
- Department of Radiation Oncology, Henry Ford Health, 1 Ford Place, 5D-42, Detroit, MI 48202 USA
| | - Farzan Siddiqui
- Department of Radiation Oncology, Henry Ford Health, 1 Ford Place, 5D-42, Detroit, MI 48202 USA
| | - Benjamin Movsas
- Department of Radiation Oncology, Henry Ford Health, 1 Ford Place, 5D-42, Detroit, MI 48202 USA
- College of Human Medicine, Michigan State University, East Lansing, MI 48824 USA
| | - Shyam Nyati
- Department of Radiation Oncology, Henry Ford Health, 1 Ford Place, 5D-42, Detroit, MI 48202 USA
- College of Human Medicine, Michigan State University, East Lansing, MI 48824 USA
| |
Collapse
|
17
|
Gryciuk A, Rogalska M, Baran J, Kuryk L, Staniszewska M. Oncolytic Adenoviruses Armed with Co-Stimulatory Molecules for Cancer Treatment. Cancers (Basel) 2023; 15:cancers15071947. [PMID: 37046608 PMCID: PMC10093006 DOI: 10.3390/cancers15071947] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/19/2023] [Accepted: 03/22/2023] [Indexed: 04/14/2023] Open
Abstract
In clinical trials, adenovirus vectors (AdVs) are commonly used platforms for human gene delivery therapy. High genome capacity and flexibility in gene organization make HAdVs suitable for cloning. Recent advancements in molecular techniques have influenced the development of genetically engineered adenovirus vectors showing therapeutic potential. Increased molecular understanding of the benefits and limitations of HAdVs in preclinical research and clinical studies is a crucial point in the engineering of refined oncolytic vectors. This review presents HAdV species (A-G) used in oncotherapy. We describe the adenovirus genome organizations and modifications, the possibilities oncolytic viruses offer, and their current limitations. Ongoing and ended clinical trials based on oncolytic adenoviruses are presented. This review provides a broad overview of the current knowledge of oncolytic therapy. HAdV-based strategies targeting tumors by employing variable immune modifiers or delivering immune stimulatory factors are of great promise in the field of immune oncologyy This approach can change the face of the fight against cancer, supplying the medical tools to defeat tumors more selectively and safely.
Collapse
Affiliation(s)
- Aleksander Gryciuk
- Department of Microbiology, Molecular Genetics and Genomics, Centre of Advanced Materials and Technology CEZAMAT, Warsaw University of Technology, 02-822 Warsaw, Poland
| | - Marta Rogalska
- Department of Microbiology, Molecular Genetics and Genomics, Centre of Advanced Materials and Technology CEZAMAT, Warsaw University of Technology, 02-822 Warsaw, Poland
| | - Joanna Baran
- Department of Microbiology, Molecular Genetics and Genomics, Centre of Advanced Materials and Technology CEZAMAT, Warsaw University of Technology, 02-822 Warsaw, Poland
| | - Lukasz Kuryk
- Department of Virology, National Institute of Public Health NIH-NRI, 00-791 Warsaw, Poland
- Valo Therapeutics, 00790 Helsinki, Finland
| | - Monika Staniszewska
- Department of Microbiology, Molecular Genetics and Genomics, Centre of Advanced Materials and Technology CEZAMAT, Warsaw University of Technology, 02-822 Warsaw, Poland
| |
Collapse
|
18
|
Liu S, Li M, Sun F, Zhang J, Liu F. Enhancing the immune effect of oHSV-1 therapy through TLR3 signaling in uveal melanoma. J Cancer Res Clin Oncol 2023; 149:901-912. [PMID: 36030435 DOI: 10.1007/s00432-022-04272-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 08/07/2022] [Indexed: 11/26/2022]
Abstract
PURPOSE Uveal melanoma (UM) is the most common primary intraocular malignant tumor in adults, with patients having a low overall survival rate. Oncolytic viruses (OVs) have been shown effective as monotherapy or combined with immunotherapy in the treatment of UM. Oncolytic herpes simplex type I virus (oHSV-1) was found to alter gene expression and immune function in UMs. We investigated whether a combination treatment would be more effective in treating UM and reactive immune cells. METHODS RNA sequencing analysis were used to identify the effect of oHSV-1 infection in UM cells and protein changes were validated by western blot. Cell viability assays were performed through UM cell lines (MUM2B, 92.1, and MP41) and retinal pigment epithelial cell line (ARPE-19) to identify the efficacy and safety of the combination treatment. Western blot, qRT-PCR, cell viability assay and immunocytochemistry were performed to discover the reactivation of immune cells (U937 and HMC3). RESULTS Through RNA sequencing analysis and in vitro molecular biology assays, this study tested the ability of oHSV-1 combined with the TLR3 agonist poly(I:C) to re-activate the TLR3 meditated NF-ƙB signaling pathway and further increase the anti-tumor activity of UM cells and macrophages, including the stimulation of macrophage polarization and proliferation. CONCLUSIONS These findings indicate that the treatment of UM with a combination of oHSV-1 and poly(I:C) generates immune responses and enhances anti-tumoral activity, suggesting the need for further investigations and clinical trials of this combination.
Collapse
Affiliation(s)
- Sisi Liu
- Beijing Tongren Eye Center, Beijing Tongren Hospital, Capital Medical University, Dongjiao Minxiang 1, Dongcheng District, Beijing, 100730, China
| | - Mingxin Li
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Beijing Laboratory of Biomedical Materials, Department of Neurosurgery, Beijing Tiantan Hospital affiliated to Capital Medical University, No. 119 Nansihuan West Road, Fengtai District, Beijing, 100070, China
| | - Fengqiao Sun
- Department of Neurosurgery, Peking University International Hospital, Peking University Health Science Center, Peking University, Shengming Kexueyuan 1, Changping District, Beijing, 102206, China
| | - Junwen Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Beijing Laboratory of Biomedical Materials, Department of Neurosurgery, Beijing Tiantan Hospital affiliated to Capital Medical University, No. 119 Nansihuan West Road, Fengtai District, Beijing, 100070, China.
| | - Fusheng Liu
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Beijing Laboratory of Biomedical Materials, Department of Neurosurgery, Beijing Tiantan Hospital affiliated to Capital Medical University, No. 119 Nansihuan West Road, Fengtai District, Beijing, 100070, China.
| |
Collapse
|
19
|
Kuryk L, Møller ASW. Next generation oncolytic viruses expressing PADI1 and TIMP2 exhibit anti-tumor activity against melanoma in nude and humanized mouse models. Mol Ther Oncolytics 2023; 28:158-170. [PMID: 36816748 PMCID: PMC9922816 DOI: 10.1016/j.omto.2023.01.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
Immunotherapy of metastatic melanoma (MM) has vastly improved the longevity of only a minority of patients. To broaden the repertoire of agents against MM, we investigated the effectiveness of locally interrupting tumor blood endothelial cell proliferation and angiogenesis, arginine deprivation, or both on the growth of melanoma by constructing and characterizing the effectiveness of four oncolytic adenoviruses. ONCOS-207 (which expressed tissue inhibitor of metalloprotease type 2 [TIMP2]), ONCOS-209 (which expressed peptidyl arginine deiminase [PADI1]), and ONCOS-210 and ONCOS-212 (which expressed both TIMP2 and PADI1) exhibited oncolytic activity against four melanoma cell lines in vitro. ONCOS-212 treatments significantly inhibited tumor growth in an A2058 tumor model in nude mice compared with vehicle control. The inhibitory effects of the two transgenes of ONCOS-212 on tumor growth appeared to be synergistic. These viruses also significantly inhibited tumor growth in a humanized NOG model of melanoma (A2058 xenograft). All viruses significantly increased the percentage of activated CD8+ T cells in the tumor-infiltrating lymphocytes. The abscopal effect of ONCOS-212 treatments in the A2058 tumor challenge model in hNOG mice supports the hypothesis that the human immune response contributes to the anti-tumor activity of ONCOS-212. These results support the further development of ONCOS-212 for cancer treatment.
Collapse
Affiliation(s)
- Lukasz Kuryk
- Targovax ASA, Clinical Science, Vollsveien 19, NO-1366 Lysaker Oslo, Norway,National Institute of Public Health NIH – National Research Institute, Department of Virology, Chocimska 24, 00-791 Warsaw, Poland,Corresponding author: Lukasz Kuryk, National Institute of Public Health NIH – National Research Institute, Department of Virology, Chocimska 24, 00-791 Warsaw, Poland.
| | | |
Collapse
|
20
|
Effects of pre-existing anti-adenovirus antibodies on transgene expression levels and therapeutic efficacies of arming oncolytic adenovirus. Sci Rep 2022; 12:21560. [PMID: 36513733 PMCID: PMC9747716 DOI: 10.1038/s41598-022-26030-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 12/08/2022] [Indexed: 12/15/2022] Open
Abstract
Oncolytic adenoviruses (OAds), most of which are based on species C human adenovirus serotype 5 (Ad5) (OAd5), have recently received much attention as potential anticancer agents. High seroprevalence of anti-Ad5 neutralizing antibodies is a major hurdle for Ad5-based gene therapy. However, the impacts of anti-Ad5 neutralizing antibodies on OAd5-mediated transgene expression in the tumor and antitumor effects remain to be fully elucidated. In this study, we examined the impact of anti-Ad5 neutralizing antibodies on the OAd5-mediated antitumor effects and OAd5-mediated transgene expression. The luciferase expression of OAd-tAIB-Luc, which contains the cytomegalovirus promoter-driven luciferase gene, was inhibited in human cultured cells in the presence of human serum. Although the inhibitory effects of human serum possessing the low anti-Ad5 neutralizing antibody titers were overcome by long-term infection, the in vitro tumor cell lysis activities of OAd-tAIB-Luc were entirely attenuated by human serum containing the high titers of anti-Ad5 neutralizing antibodies. OAd-tAIB-Luc-mediated luciferase expression in the subcutaneous tumors 3 days after administration and tumor growth suppression levels following intratumoral administration were significantly lower in mice possessing the high titers of anti-Ad5 neutralizing antibodies, compared to those in control mice. These results suggested that pre-existing anti-Ad5 antibodies attenuated both transgene expression and potential antitumor effects of OAd5 following intratumoral administration.
Collapse
|
21
|
Li SJ, Sun ZJ. Fueling immune checkpoint blockade with oncolytic viruses: Current paradigms and challenges ahead. Cancer Lett 2022; 550:215937. [DOI: 10.1016/j.canlet.2022.215937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/20/2022] [Accepted: 09/29/2022] [Indexed: 11/29/2022]
|
22
|
Vorobjeva IV, Zhirnov OP. Modern approaches to treating cancer with oncolytic viruses. MICROBIOLOGY INDEPENDENT RESEARCH JOURNAL 2022. [DOI: 10.18527/2500-2236-2022-9-1-91-112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023] Open
Abstract
According to the World Health Organization, cancer is the second leading cause of death in the world. This serves as a powerful incentive to search for new effective cancer treatments. Development of new oncolytic viruses capable of selectively destroying cancer cells is one of the modern approaches to cancer treatment. The advantage of this method – the selective lysis of tumor cells with the help of viruses – leads to an increase in the antitumor immune response of the body, that in turn promotes the destruction of the primary tumor and its metastases. Significant progress in development of this method has been achieved in the last decade. In this review we analyze the literature data on families of oncolytic viruses that have demonstrated a positive therapeutic effect against malignant neoplasms in various localizations. We discuss the main mechanisms of the oncolytic action of viruses and assess their advantages over other methods of cancer therapy as well as the prospects for their use in clinical practice.
Collapse
Affiliation(s)
- I. V. Vorobjeva
- N. F. Gamaleya National Research Center for Epidemiology and Microbiology, D. I. Ivanovsky Institute of Virology
| | - O. P. Zhirnov
- N. F. Gamaleya National Research Center for Epidemiology and Microbiology, D. I. Ivanovsky Institute of Virology; The Russian-German Academy of Medical and Biotechnological Sciences
| |
Collapse
|
23
|
Oronsky B, Gastman B, Conley AP, Reid C, Caroen S, Reid T. Oncolytic Adenoviruses: The Cold War against Cancer Finally Turns Hot. Cancers (Basel) 2022; 14:4701. [PMID: 36230621 PMCID: PMC9562194 DOI: 10.3390/cancers14194701] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/21/2022] [Accepted: 09/25/2022] [Indexed: 11/27/2022] Open
Abstract
Oncolytic viruses, colloquially referred to as "living drugs", amplify themselves and the therapeutic transgenes that they carry to stimulate an immune response both locally and systemically. Remarkable exceptions aside, such as the recent 14-patient trial with the PD-1 inhibitor, dostarlimab, in mismatch repair (MMR) deficient rectal cancer, where the complete response rate was 100%, checkpoint inhibitors are not cure-alls, which suggests the need for a combination partner like oncolytic viruses to prime and augment their activity. This review focuses on adenoviruses, the most clinically investigated of all the oncolytic viruses. It covers specific design features of clinical adenoviral candidates and highlights their potential both alone and in combination with checkpoint inhibitors in clinical trials to turn immunologically "cold" and unresponsive tumors into "hotter" and more responsive ones through a domino effect. Finally, a "mix-and-match" combination of therapies based on the paradigm of the cancer-immunity cycle is proposed to augment the immune responses of oncolytic adenoviruses.
Collapse
Affiliation(s)
| | | | - Anthony P. Conley
- University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Scott Caroen
- EpicentRx, Torrey Pines, La Jolla, CA 92037, USA
| | - Tony Reid
- EpicentRx, Torrey Pines, La Jolla, CA 92037, USA
| |
Collapse
|
24
|
Yun CO, Hong J, Yoon AR. Current clinical landscape of oncolytic viruses as novel cancer immunotherapeutic and recent preclinical advancements. Front Immunol 2022; 13:953410. [PMID: 36091031 PMCID: PMC9458317 DOI: 10.3389/fimmu.2022.953410] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/03/2022] [Indexed: 12/12/2022] Open
Abstract
Oncolytic viruses (OVs) have been gaining attention in the pharmaceutical industry as a novel immunotherapeutic and therapeutic adjuvant due to their ability to induce and boost antitumor immunity through multiple mechanisms. First, intrinsic mechanisms of OVs that enable exploitation of the host immune system (e.g., evading immune detection) can nullify the immune escape mechanism of tumors. Second, many types of OVs have been shown to cause direct lysis of tumor cells, resulting in an induction of tumor-specific T cell response mediated by release of tumor-associated antigens and danger signal molecules. Third, armed OV-expressing immune stimulatory therapeutic genes could be highly expressed in tumor tissues to further improve antitumor immunity. Last, these OVs can inflame cold tumors and their microenvironment to be more immunologically favorable for other immunotherapeutics. Due to these unique characteristics, OVs have been tested as an adjuvant of choice in a variety of therapeutics. In light of these promising attributes of OVs in the immune-oncology field, the present review will examine OVs in clinical development and discuss various strategies that are being explored in preclinical stages for the next generation of OVs that are optimized for immunotherapy applications.
Collapse
Affiliation(s)
- Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, South Korea
- Institute of Nano Science and Technology (INST), Hanyang University, Seoul, South Korea
- Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul, South Korea
- GeneMedicine CO., Ltd., Seoul, South Korea
| | | | - A-Rum Yoon
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, South Korea
- Institute of Nano Science and Technology (INST), Hanyang University, Seoul, South Korea
- Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul, South Korea
| |
Collapse
|
25
|
Recent Advances and Challenges in Uveal Melanoma Immunotherapy. Cancers (Basel) 2022; 14:cancers14133094. [PMID: 35804863 PMCID: PMC9264803 DOI: 10.3390/cancers14133094] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 06/19/2022] [Accepted: 06/21/2022] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Uveal melanoma is the most common primary intraocular malignancy in adults. Although it can be controlled locally, half of the patients still develop metastases. To date, there have been no standard therapeutic strategies for the prevention or treatment of metastases. Existing therapies, such as chemotherapy and targeted therapies, induce only minimal responses. This review focuses on newly published research on immunotherapy. We highlight expanding treatments and their clinical outcomes, as well as propose promising new treatments and feasible checkpoints. Based on these findings, we provide innovative insights into feasible strategies for the treatment of patients with uveal melanoma. Abstract Uveal melanoma (UM) is the most common primary intraocular malignancy in adults. Compared to cutaneous melanoma (CM), which mainly harbors BRAF or NRAS mutations, UM predominantly harbors GNAQ or GNA11 mutations. Although primary UM can be controlled locally, approximately 50% of patients still develop metastases. To date, there have been no standard therapeutic strategies for the prevention or treatment of metastases. Unfortunately, chemotherapy and targeted therapies only induce minimal responses in patients with metastatic UM, with a median survival time of only 4–5 months after metastasis detection. Immunotherapy agents, such as immune checkpoint inhibitors, have achieved pioneering outcomes in CM but have shown limited effects in UM. Researchers have explored several feasible checkpoints to identify options for future therapies. Cancer vaccines have shown little in the way of therapeutic benefit in patients with UM, and there are few ongoing trials providing favorable evidence, but adoptive cell transfer-related therapies seem promising and deserve further investigation. More recently, the immune-mobilizing monoclonal T-cell receptor against the cancer molecule tebentafusp showed impressive antitumor effects. Meanwhile, oncolytic viruses and small molecule inhibitors have also gained ground. This review highlights recent progress in burgeoning treatments and provides innovative insights on feasible strategies for the treatment of UM.
Collapse
|
26
|
Ziogas DC, Martinos A, Petsiou DP, Anastasopoulou A, Gogas H. Beyond Immunotherapy: Seizing the Momentum of Oncolytic Viruses in the Ideal Platform of Skin Cancers. Cancers (Basel) 2022; 14:2873. [PMID: 35740539 PMCID: PMC9221332 DOI: 10.3390/cancers14122873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/30/2022] [Accepted: 06/03/2022] [Indexed: 11/24/2022] Open
Abstract
Despite the durable remissions induced by ICIs and targeted therapies in advanced melanoma and non-melanoma skin cancers, both subtypes usually relapse. Many systematic therapies have been tested to increase efficacy and delay relapse in ICIs, but their success has been limited. Due the feasibility of this approach, skin cancers have become the ideal platform for intralesional infusions of many novel agents, including oncolytic viruses (OVs). Talimogene laherparepvec (T-VEC) was the first FDA-approved OV for the treatment of unresectable melanoma and this virus opened up further potential for the use of this class of agents, especially in combination with ICIs, in order to achieve deeper and longer immune-mediated responses. However, the recently announced phase III MASTERKEY-265 trial was not able to confirm that the addition of T-VEC to pembrolizumab treatment improves progression-free or overall survival over the use of pembrolizumab alone. Despite these results, numerous studies are currently active, evaluating T-VEC and several other OVs as monotherapies or in regimens with ICIs in different subtypes of skin cancer. This overview provides a comprehensive update on the evolution status of all available OVs in melanoma and non-melanoma skin cancers and summarizes the more interesting preclinical findings, the latest clinical evidence, and the future insights in relation to the expected selective incorporation of some of these OVs into oncological practice.
Collapse
Affiliation(s)
| | | | | | | | - Helen Gogas
- First Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, 115 27 Athens, Greece; (D.C.Z.); (A.M.); (D.-P.P.); (A.A.)
| |
Collapse
|
27
|
Tumor-Homing of Mesenchymal Stem Cells Infected with Oncolytic Virus in a Canine Patient. Vet Sci 2022; 9:vetsci9060285. [PMID: 35737337 PMCID: PMC9228126 DOI: 10.3390/vetsci9060285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 06/04/2022] [Accepted: 06/07/2022] [Indexed: 11/30/2022] Open
Abstract
Intravenous administration of oncolytic adenovirus (OAds) can be challenging, although various vehicles for the delivery of the virus to the tumor have been described. The efficacy of mesenchymal stem cells (MSCs) as a virus vehicle has been reported in mouse models and canine and human patients, but the actual action mechanism has never been described in patients. It is of importance to determine whether MSCs infected with OAds can reach the tumor and release the virus in a clinical setting. For this purpose, GFP-labeled MSCs were infected with an OAd and inoculated into a companion dog diagnosed with spontaneous lung carcinoma. Forty-eight hours later, the tumor was excised and analyzed microscopically by flow cytometry for GFP fluorescence detection, and a cellular culture was established. Peripheral blood samples were taken to quantify the oncolytic adenovirus by qRT-PCR. Green fluorescence cells detected in the cellular culture by microscopy and flow cytometry revealed 0.69% GFP-positive cells in the tumor. OAd in peripheral blood was confirmed by qRT-PCR during follow-up. For the first time, the tumoral-homing capacity of OAds infected-MSC has been confirmed in a clinical setting, helping to explain the clinical response mechanism, whose efficacy was previously reported in canine and human patients.
Collapse
|
28
|
Coughlan L, Kremer EJ, Shayakhmetov DM. Adenovirus-based vaccines-a platform for pandemic preparedness against emerging viral pathogens. Mol Ther 2022; 30:1822-1849. [PMID: 35092844 PMCID: PMC8801892 DOI: 10.1016/j.ymthe.2022.01.034] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 11/24/2022] Open
Abstract
Zoonotic viruses continually pose a pandemic threat. Infection of humans with viruses for which we typically have little or no prior immunity can result in epidemics with high morbidity and mortality. These epidemics can have public health and economic impact and can exacerbate civil unrest or political instability. Changes in human behavior in the past few decades-increased global travel, farming intensification, the exotic animal trade, and the impact of global warming on animal migratory patterns, habitats, and ecosystems-contribute to the increased frequency of cross-species transmission events. Investing in the pre-clinical advancement of vaccine candidates against diverse emerging viral threats is crucial for pandemic preparedness. Replication-defective adenoviral (Ad) vectors have demonstrated their utility as an outbreak-responsive vaccine platform during the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic. Ad vectors are easy to engineer; are amenable to rapid, inexpensive manufacturing; are relatively safe and immunogenic in humans; and, importantly, do not require specialized cold-chain storage, making them an ideal platform for equitable global distribution or stockpiling. In this review, we discuss the progress in applying Ad-based vaccines against emerging viruses and summarize their global safety profile, as reflected by their widespread geographic use during the SARS-CoV-2 pandemic.
Collapse
Affiliation(s)
- Lynda Coughlan
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Center for Vaccine Development and Global Health (CVD), University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Eric J Kremer
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, CNRS 5535, Montpellier, France.
| | - Dmitry M Shayakhmetov
- Lowance Center for Human Immunology, Emory University School of Medicine, Atlanta, GA 30322, USA; Emory Vaccine Center, Departments of Pediatrics and Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA; Discovery and Developmental Therapeutics Program, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA.
| |
Collapse
|
29
|
Cytokine Responses to Adenovirus and Adenovirus Vectors. Viruses 2022; 14:v14050888. [PMID: 35632630 PMCID: PMC9145601 DOI: 10.3390/v14050888] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/18/2022] [Accepted: 04/20/2022] [Indexed: 12/15/2022] Open
Abstract
The expression of cytokines and chemokines in response to adenovirus infection is tightly regulated by the innate immune system. Cytokine-mediated toxicity and cytokine storm are known clinical phenomena observed following naturally disseminated adenovirus infection in immunocompromised hosts as well as when extremely high doses of adenovirus vectors are injected intravenously. This dose-dependent, cytokine-mediated toxicity compromises the safety of adenovirus-based vectors and represents a critical problem, limiting their utility for gene therapy applications and the therapy of disseminated cancer, where intravenous injection of adenovirus vectors may provide therapeutic benefits. The mechanisms triggering severe cytokine response are not sufficiently understood, prompting efforts to further investigate this phenomenon, especially in clinically relevant settings. In this review, we summarize the current knowledge on cytokine and chemokine activation in response to adenovirus- and adenovirus-based vectors and discuss the underlying mechanisms that may trigger acute cytokine storm syndrome. First, we review profiles of cytokines and chemokines that are activated in response to adenovirus infection initiated via different routes. Second, we discuss the molecular mechanisms that lead to cytokine and chemokine transcriptional activation. We further highlight how immune cell types in different organs contribute to synthesis and systemic release of cytokines and chemokines in response to adenovirus sensing. Finally, we review host factors that can limit cytokine and chemokine expression and discuss currently available and potential future interventional approaches that allow for the mitigation of the severity of the cytokine storm syndrome. Effective cytokine-targeted interventional approaches may improve the safety of systemic adenovirus delivery and thus broaden the potential clinical utility of adenovirus-based therapeutic vectors.
Collapse
|
30
|
Cerqueira OLD, Antunes F, Assis NG, Cardoso EC, Clavijo-Salomón MA, Domingues AC, Tessarollo NG, Strauss BE. Perspectives for Combining Viral Oncolysis With Additional Immunotherapies for the Treatment of Melanoma. Front Mol Biosci 2022; 9:777775. [PMID: 35495634 PMCID: PMC9048901 DOI: 10.3389/fmolb.2022.777775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 03/22/2022] [Indexed: 12/19/2022] Open
Abstract
Melanoma is the deadliest type of skin cancer with steadily increasing incidence worldwide during the last few decades. In addition to its tumor associated antigens (TAAs), melanoma has a high mutation rate compared to other tumors, which promotes the appearance of tumor specific antigens (TSAs) as well as increased lymphocytic infiltration, inviting the use of therapeutic tools that evoke new or restore pre-existing immune responses. Innovative therapeutic proposals, such as immune checkpoint inhibitors (ICIs), have emerged as effective options for melanoma. However, a significant portion of these patients relapse and become refractory to treatment. Likewise, strategies using viral vectors, replicative or not, have garnered confidence and approval by different regulatory agencies around the world. It is possible that further success of immune therapies against melanoma will come from synergistic combinations of different approaches. In this review we outline molecular features inherent to melanoma and how this supports the use of viral oncolysis and immunotherapies when used as monotherapies or in combination.
Collapse
Affiliation(s)
- Otto Luiz Dutra Cerqueira
- Centro de Investigação Translacional em Oncologia (CTO)/LIM, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Fernanda Antunes
- Centro de Investigação Translacional em Oncologia (CTO)/LIM, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Nadine G Assis
- Centro de Investigação Translacional em Oncologia (CTO)/LIM, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Elaine C Cardoso
- Department of Pediatrics, Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Maria A Clavijo-Salomón
- Centro de Investigação Translacional em Oncologia (CTO)/LIM, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Ana C Domingues
- Centro de Investigação Translacional em Oncologia (CTO)/LIM, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Nayara G Tessarollo
- Centro de Investigação Translacional em Oncologia (CTO)/LIM, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
| | - Bryan E Strauss
- Centro de Investigação Translacional em Oncologia (CTO)/LIM, Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo (FMUSP), São Paulo, Brazil
- *Correspondence: Bryan E Strauss,
| |
Collapse
|
31
|
Revisiting the melanomagenic pathways and current therapeutic approaches. Mol Biol Rep 2022; 49:9651-9671. [DOI: 10.1007/s11033-022-07412-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 03/22/2022] [Indexed: 01/10/2023]
|
32
|
Ernst M, Giubellino A. The Current State of Treatment and Future Directions in Cutaneous Malignant Melanoma. Biomedicines 2022; 10:822. [PMID: 35453572 PMCID: PMC9029866 DOI: 10.3390/biomedicines10040822] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/25/2022] [Accepted: 03/29/2022] [Indexed: 02/01/2023] Open
Abstract
Malignant melanoma is the leading cause of death among cutaneous malignancies. While its incidence is increasing, the most recent cancer statistics show a small but clear decrease in mortality rate. This trend reflects the introduction of novel and more effective therapeutic regimens, including the two cornerstones of melanoma therapy: immunotherapies and targeted therapies. Immunotherapies exploit the highly immunogenic nature of melanoma by modulating and priming the patient's own immune system to attack the tumor. Treatments combining immunotherapies with targeted therapies, which disable the carcinogenic products of mutated cancer cells, have further increased treatment efficacy and durability. Toxicity and resistance, however, remain critical challenges to the field. The present review summarizes past treatments and novel therapeutic interventions and discusses current clinical trials and future directions.
Collapse
Affiliation(s)
| | - Alessio Giubellino
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA;
| |
Collapse
|
33
|
Garcia-Carbonero R, Bazan-Peregrino M, Gil-Martín M, Álvarez R, Macarulla T, Riesco-Martinez MC, Verdaguer H, Guillén-Ponce C, Farrera-Sal M, Moreno R, Mato-Berciano A, Maliandi MV, Torres-Manjon S, Costa M, Del Pozo N, Martínez de Villarreal J, Real FX, Vidal N, Capella G, Alemany R, Blasi E, Blasco C, Cascalló M, Salazar R. Phase I, multicenter, open-label study of intravenous VCN-01 oncolytic adenovirus with or without nab-paclitaxel plus gemcitabine in patients with advanced solid tumors. J Immunother Cancer 2022; 10:e003255. [PMID: 35338084 PMCID: PMC8961117 DOI: 10.1136/jitc-2021-003255] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2022] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND VCN-01 is an oncolytic adenovirus (Ad5 based) designed to replicate in cancer cells with dysfunctional RB1 pathway, express hyaluronidase to enhance virus intratumoral spread and facilitate chemotherapy and immune cells extravasation into the tumor. This phase I clinical trial was aimed to find the maximum tolerated dose/recommended phase II dose (RP2D) and dose-limiting toxicity (DLT) of the intravenous delivery of the replication-competent VCN-01 adenovirus in patients with advanced cancer. METHODS Part I: patients with advanced refractory solid tumors received one single dose of VCN-01. Parts II and III: patients with pancreatic adenocarcinoma received VCN-01 (only in cycle 1) and nab-paclitaxel plus gemcitabine (VCN-concurrent on day 1 in Part II, and 7 days before chemotherapy in Part III). Patients were required to have anti-Ad5 neutralizing antibody (NAbs) titers lower than 1/350 dilution. Pharmacokinetic and pharmacodynamic analyses were performed. RESULTS 26% of the patients initially screened were excluded based on high NAbs levels. Sixteen and 12 patients were enrolled in Part I and II, respectively: RP2D were 1×1013 viral particles (vp)/patient (Part I), and 3.3×1012 vp/patient (Part II). Fourteen patients were included in Part III: there were no DLTs and the RP2D was 1×1013 vp/patient. Observed DLTs were grade 4 aspartate aminotransferase increase in one patient (Part I, 1×1013 vp), grade 4 febrile neutropenia in one patient and grade 5 thrombocytopenia plus enterocolitis in another patient (Part II, 1×1013 vp). In patients with pancreatic adenocarcinoma overall response rate were 50% (Part II) and 50% (Part III). VCN-01 viral genomes were detected in tumor tissue in five out of six biopsies (day 8). A second viral plasmatic peak and increased hyaluronidase serum levels suggested replication after intravenous injection in all patients. Increased levels of immune biomarkers (interferon-γ, soluble lymphocyte activation gene-3, interleukin (IL)-6, IL-10) were found after VCN-01 administration. CONCLUSIONS Treatment with VCN-01 is feasible and has an acceptable safety. Encouraging biological and clinical activity was observed when administered in combination with nab-paclitaxel plus gemcitabine to patients with pancreatic adenocarcinoma. TRIAL REGISTRATION NUMBER NCT02045602.
Collapse
Affiliation(s)
- Rocio Garcia-Carbonero
- Oncology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), UCM, CNIO, CIBERONC, Madrid, Spain
| | | | - Marta Gil-Martín
- Medical Oncology Department, Institut Catala d'Oncologia, L'Hospitalet de Llobregat, Barcelona, Spain
- Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Rafael Álvarez
- Centro Integral Oncológico Clara Campal (CIOCC), Madrid, Spain
| | - Teresa Macarulla
- Vall d'Hebron University Hospital & Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Maria C Riesco-Martinez
- Oncology Department, Hospital Universitario 12 de Octubre, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), UCM, CNIO, CIBERONC, Madrid, Spain
| | - Helena Verdaguer
- Vall d'Hebron University Hospital & Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | | | - Martí Farrera-Sal
- VCN Biosciences, Sant Cugat del Vallès, Barcelona, Spain
- Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
- ProCure Program, Institut Catala d'Oncologia, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Rafael Moreno
- Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
- ProCure Program, Institut Catala d'Oncologia, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | | | | | - Silvia Torres-Manjon
- Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
- ProCure Program, Institut Catala d'Oncologia, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Marcel Costa
- Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
- ProCure Program, Institut Catala d'Oncologia, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Natalia Del Pozo
- Epithelial Carcinogenesis Group, Molecular Oncology Programme, Spanish National Cancer Research Centre-CNIO, Madrid, Spain
| | - Jaime Martínez de Villarreal
- Epithelial Carcinogenesis Group, Molecular Oncology Programme, Spanish National Cancer Research Centre-CNIO, Madrid, Spain
| | - Francisco X Real
- Epithelial Carcinogenesis Group, Molecular Oncology Programme, Spanish National Cancer Research Centre-CNIO, Madrid, Spain
- Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, Barcelona, Spain
| | - Noemí Vidal
- Department of Pathology, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Spain
| | - Gabriel Capella
- Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
- Hereditary Cancer Program, Catalan Institute of Oncology, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain, Spain
| | - Ramon Alemany
- Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
- ProCure Program, Institut Catala d'Oncologia, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Emma Blasi
- VCN Biosciences, Sant Cugat del Vallès, Barcelona, Spain
| | - Carmen Blasco
- VCN Biosciences, Sant Cugat del Vallès, Barcelona, Spain
| | - Manel Cascalló
- VCN Biosciences, Sant Cugat del Vallès, Barcelona, Spain
| | - Ramon Salazar
- Medical Oncology Department, Institut Catala d'Oncologia, L'Hospitalet de Llobregat, Barcelona, Spain
- Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain, Spain
- University of Barcelona, Barcelona, Spain
| |
Collapse
|
34
|
Naumenko VA, Stepanenko AA, Lipatova AV, Vishnevskiy DA, Chekhonin VP. Infection of non-cancer cells: A barrier or support for oncolytic virotherapy? MOLECULAR THERAPY - ONCOLYTICS 2022; 24:663-682. [PMID: 35284629 PMCID: PMC8898763 DOI: 10.1016/j.omto.2022.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Oncolytic viruses are designed to specifically target cancer cells, sparing normal cells. Although numerous studies demonstrate the ability of oncolytic viruses to infect a wide range of non-tumor cells, the significance of this phenomenon for cancer virotherapy is poorly understood. To fill the gap, we summarize the data on infection of non-cancer targets by oncolytic viruses with a special focus on tumor microenvironment and secondary lymphoid tissues. The review aims to address two major questions: how do attenuated viruses manage to infect normal cells, and whether it is of importance for oncolytic virotherapy.
Collapse
Affiliation(s)
- Victor A. Naumenko
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow 119034, Russia
- Corresponding author Victor A. Naumenko, PhD, V. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow 119034, Russia.
| | - Aleksei A. Stepanenko
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow 119034, Russia
- Department of Medical Nanobiotechnology, N.I Pirogov Russian National Research Medical University, Moscow 117997, Russia
| | - Anastasiia V. Lipatova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia
| | - Daniil A. Vishnevskiy
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow 119034, Russia
| | - Vladimir P. Chekhonin
- V. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow 119034, Russia
- Department of Medical Nanobiotechnology, N.I Pirogov Russian National Research Medical University, Moscow 117997, Russia
| |
Collapse
|
35
|
Ban W, Guan J, Huang H, He Z, Sun M, Liu F, Sun J. Emerging systemic delivery strategies of oncolytic viruses: A key step toward cancer immunotherapy. NANO RESEARCH 2022; 15:4137-4153. [PMID: 35194488 PMCID: PMC8852960 DOI: 10.1007/s12274-021-4031-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/24/2021] [Accepted: 11/28/2021] [Indexed: 05/16/2023]
Abstract
Oncolytic virotherapy (OVT) is a novel type of immunotherapy that induces anti-tumor responses through selective self-replication within cancer cells and oncolytic virus (OV)-mediated immunostimulation. Notably, talimogene laherparepvec (T-Vec) developed by the Amgen company in 2015, is the first FDA-approved OV product to be administered via intratumoral injection and has been the most successful OVT treatment. However, the systemic administration of OVs still faces huge challenges, including in vivo pre-existing neutralizing antibodies and poor targeting delivery efficacy. Recently, state-of-the-art progress has been made in the development of systemic delivery of OVs, which demonstrates a promising step toward broadening the scope of cancer immunotherapy and improving the clinical efficacy of OV delivery. Herein, this review describes the general characteristics of OVs, focusing on the action mechanisms of OVs as well as the advantages and disadvantages of OVT. The emerging multiple systemic administration approaches of OVs are summarized in the past five years. In addition, the combination treatments between OVT and traditional therapies (chemotherapy, thermotherapy, immunotherapy, and radiotherapy, etc.) are highlighted. Last but not least, the future prospects and challenges of OVT are also discussed, with the aim of facilitating medical researchers to extensively apply the OVT in the cancer therapy.
Collapse
Affiliation(s)
- Weiyue Ban
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016 China
| | - Jianhuan Guan
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016 China
| | - Hanwei Huang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang, 110016 China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016 China
| | - Mengchi Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016 China
| | - Funan Liu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, China Medical University, Ministry of Education, Shenyang, 110016 China
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016 China
| |
Collapse
|
36
|
Biegert GWG, Rosewell Shaw A, Suzuki M. Current development in adenoviral vectors for cancer immunotherapy. Mol Ther Oncolytics 2021; 23:571-581. [PMID: 34938857 DOI: 10.1016/j.omto.2021.11.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Adenoviruses are well characterized and thus easily modified to generate oncolytic vectors that directly lyse tumor cells and can be "armed" with transgenes to promote lysis, antigen presentation, and immunostimulation. Oncolytic adenoviruses (OAds) are safe, versatile, and potent immunostimulants in patients. Since transgene expression is restricted to the tumor, adenoviral transgenes overcome the toxicities and short half-life of systemically administered cytokines, immune checkpoint blockade molecules, and bispecific T cell engagers. While OAds expressing immunostimulatory molecules ("armed" OAds) have demonstrated anti-tumor potential in preclinical solid tumor models, the efficacy has not translated into significant clinical outcomes as a monotherapy. However, OAds synergize with established standards of care and novel immunotherapeutic agents, providing a multifaceted means to address complexities associated with solid tumors. Critically, armed OAds revitalize endogenous and adoptively transferred immune cells while simultaneously enhancing their anti-tumor function. To properly evaluate these novel vectors and reduce the gap in the cycle between bench-to-bedside and back, improving model systems must be a priority. The future of OAds will involve a multidimensional approach that provides immunostimulatory molecules, immune checkpoint blockade, and/or immune engagers in concert with endogenous and exogenous immune cells to initiate durable and comprehensive anti-tumor responses.
Collapse
Affiliation(s)
- Greyson Willis Grossman Biegert
- Department of Medicine, Section of Hematology/Oncology, Baylor College of Medicine, Houston, TX, USA.,Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX, USA
| | - Amanda Rosewell Shaw
- Department of Medicine, Section of Hematology/Oncology, Baylor College of Medicine, Houston, TX, USA.,Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX, USA
| | - Masataka Suzuki
- Department of Medicine, Section of Hematology/Oncology, Baylor College of Medicine, Houston, TX, USA.,Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX, USA
| |
Collapse
|
37
|
Rossi E, Croce M, Reggiani F, Schinzari G, Ambrosio M, Gangemi R, Tortora G, Pfeffer U, Amaro A. Uveal Melanoma Metastasis. Cancers (Basel) 2021; 13:5684. [PMID: 34830841 PMCID: PMC8616038 DOI: 10.3390/cancers13225684] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 02/07/2023] Open
Abstract
Uveal melanoma (UM) is characterized by relatively few, highly incident molecular alterations and their association with metastatic risk is deeply understood. Nevertheless, this knowledge has so far not led to innovative therapies for the successful treatment of UM metastases or for adjuvant therapy, leaving survival after diagnosis of metastatic UM almost unaltered in decades. The driver mutations of UM, mainly in the G-protein genes GNAQ and GNA11, activate the MAP-kinase pathway as well as the YAP/TAZ pathway. At present, there are no drugs that target the latter and this likely explains the failure of mitogen activated kinase kinase inhibitors. Immune checkpoint blockers, despite the game changing effect in cutaneous melanoma (CM), show only limited effects in UM probably because of the low mutational burden of 0.5 per megabase and the unavailability of antibodies targeting the main immune checkpoint active in UM. The highly pro-tumorigenic microenvironment of UM also contributes to therapy resistance. However, T-cell redirection by a soluble T-cell receptor that is fused to an anti-CD3 single-chain variable fragment, local, liver specific therapy, new immune checkpoint blockers, and YAP/TAZ specific drugs give new hope to repeating the success of innovative therapy obtained for CM.
Collapse
Affiliation(s)
- Ernesto Rossi
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (E.R.); (G.S.); (G.T.)
| | - Michela Croce
- Laboratory of Biotherapies, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.C.); (R.G.)
| | - Francesco Reggiani
- Laboratory of Epigenetics, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (F.R.); (M.A.); (A.A.)
| | - Giovanni Schinzari
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (E.R.); (G.S.); (G.T.)
- Medical Oncology, Università Cattolica del S. Cuore, 00168 Rome, Italy
| | - Marianna Ambrosio
- Laboratory of Epigenetics, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (F.R.); (M.A.); (A.A.)
| | - Rosaria Gangemi
- Laboratory of Biotherapies, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (M.C.); (R.G.)
| | - Giampaolo Tortora
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (E.R.); (G.S.); (G.T.)
- Medical Oncology, Università Cattolica del S. Cuore, 00168 Rome, Italy
| | - Ulrich Pfeffer
- Laboratory of Epigenetics, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (F.R.); (M.A.); (A.A.)
| | - Adriana Amaro
- Laboratory of Epigenetics, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (F.R.); (M.A.); (A.A.)
| |
Collapse
|
38
|
Concepts in Oncolytic Adenovirus Therapy. Int J Mol Sci 2021; 22:ijms221910522. [PMID: 34638863 PMCID: PMC8508870 DOI: 10.3390/ijms221910522] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 02/07/2023] Open
Abstract
Oncolytic adenovirus therapy is gaining importance as a novel treatment option for the management of various cancers. Different concepts of modification within the adenovirus vector have been identified that define the mode of action against and the interaction with the tumour. Adenoviral vectors allow for genetic manipulations that restrict tumour specificity and also the expression of specific transgenes in order to support the anti-tumour effect. Additionally, replication of the virus and reinfection of neighbouring tumour cells amplify the therapeutic effect. Another important aspect in oncolytic adenovirus therapy is the virus induced cell death which is a process that activates the immune system against the tumour. This review describes which elements in adenovirus vectors have been identified for modification not only to utilize oncolytic adenovirus vectors into conditionally replicating adenoviruses (CRAds) that allow replication specifically in tumour cells but also to confer specific characteristics to these viruses. These advances in development resulted in clinical trials that are summarized based on the conceptual design.
Collapse
|
39
|
Santos Apolonio J, Lima de Souza Gonçalves V, Cordeiro Santos ML, Silva Luz M, Silva Souza JV, Rocha Pinheiro SL, de Souza WR, Sande Loureiro M, de Melo FF. Oncolytic virus therapy in cancer: A current review. World J Virol 2021; 10:229-255. [PMID: 34631474 PMCID: PMC8474975 DOI: 10.5501/wjv.v10.i5.229] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/19/2021] [Accepted: 08/09/2021] [Indexed: 02/06/2023] Open
Abstract
In view of the advancement in the understanding about the most diverse types of cancer and consequently a relentless search for a cure and increased survival rates of cancer patients, finding a therapy that is able to combat the mechanism of aggression of this disease is extremely important. Thus, oncolytic viruses (OVs) have demonstrated great benefits in the treatment of cancer because it mediates antitumor effects in several ways. Viruses can be used to infect cancer cells, especially over normal cells, to present tumor-associated antigens, to activate "danger signals" that generate a less immune-tolerant tumor microenvironment, and to serve transduction vehicles for expression of inflammatory and immunomodulatory cytokines. The success of therapies using OVs was initially demonstrated by the use of the genetically modified herpes virus, talimogene laherparepvec, for the treatment of melanoma. At this time, several OVs are being studied as a potential treatment for cancer in clinical trials. However, it is necessary to be aware of the safety and possible adverse effects of this therapy; after all, an effective treatment for cancer should promote regression, attack the tumor, and in the meantime induce minimal systemic repercussions. In this manuscript, we will present a current review of the mechanism of action of OVs, main clinical uses, updates, and future perspectives on this treatment.
Collapse
Affiliation(s)
- Jonathan Santos Apolonio
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, Vitória da Conquista 45029-094, Bahia, Brazil
| | | | - Maria Luísa Cordeiro Santos
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Marcel Silva Luz
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, Vitória da Conquista 45029-094, Bahia, Brazil
| | - João Victor Silva Souza
- Universidade Estadual do Sudoeste da Bahia, Campus Vitória da Conquista, Vitória da Conquista 45083-900, Bahia, Brazil
| | - Samuel Luca Rocha Pinheiro
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Wedja Rafaela de Souza
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Matheus Sande Loureiro
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, Vitória da Conquista 45029-094, Bahia, Brazil
| | - Fabrício Freire de Melo
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde, Vitória da Conquista 45029-094, Bahia, Brazil
| |
Collapse
|
40
|
Toro MD, Gozzo L, Tracia L, Cicciù M, Drago F, Bucolo C, Avitabile T, Rejdak R, Nowomiejska K, Zweifel S, Yousef YA, Nazzal R, Romano GL. New Therapeutic Perspectives in the Treatment of Uveal Melanoma: A Systematic Review. Biomedicines 2021; 9:biomedicines9101311. [PMID: 34680428 PMCID: PMC8533164 DOI: 10.3390/biomedicines9101311] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/15/2021] [Accepted: 09/21/2021] [Indexed: 12/13/2022] Open
Abstract
Uveal melanoma (UM) is a rare disease, but the most common primary intraocular cancer, mostly localized in the choroid. Currently, the first-line treatment options for UM are radiation therapy, resection, and enucleation. However, although these treatments could potentially be curative, half of all patients will develop metastatic disease, whose prognosis is still poor. Indeed, effective therapeutic options for patients with advanced or metastatic disease are still lacking. Recently, the development of new treatment modalities with a lower incidence of adverse events, a better disease control rate, and new therapeutic approaches, have merged as new potential and promising therapeutic strategies. Additionally, several clinical trials are ongoing to find new therapeutic options, mainly for those with metastatic disease. Many interventions are still in the preliminary phases of clinical development, being investigated in phase I trial or phase I/II. The success of these trials could be crucial for changing the prognosis of patients with advanced/metastatic UM. In this systematic review, we analyzed all emerging and available literature on the new perspectives in the treatment of UM and patient outcomes; furthermore, their current limitations and more common adverse events are summarized.
Collapse
Affiliation(s)
- Mario Damiano Toro
- Department of Ophthalmology, University of Zurich, 8091 Zurich, Switzerland; (M.D.T.); (S.Z.)
- Department of General and Pediatric Ophthalmology, Medical University of Lublin, 20079 Lublin, Poland; (R.R.); (K.N.)
| | - Lucia Gozzo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (F.D.); (C.B.); (G.L.R.)
- Clinical Pharmacology Unit, Regional Pharmacovigilance Centre, University Hospital of Catania, 95123 Catania, Italy
- Correspondence: ; Tel.: +39-095-3781757
| | - Luciano Tracia
- Plastic and Reconstructive Surgery Department, American Hospital Dubai, Dubai, United Arab Emirates;
| | - Marco Cicciù
- Department of Biomedical and Dental Sciences, Morphological and Functional Images, University of Messina, AOU ‘G. Martino’, 98124 Messina, Italy;
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (F.D.); (C.B.); (G.L.R.)
- Clinical Pharmacology Unit, Regional Pharmacovigilance Centre, University Hospital of Catania, 95123 Catania, Italy
- Centre for Research and Consultancy in HTA and Drug Regulatory Affairs (CERD), University of Catania, 95123 Catania, Italy
| | - Claudio Bucolo
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (F.D.); (C.B.); (G.L.R.)
- Center of Research in Ocular Pharmacology—CERFO, University of Catania, 95123 Catania, Italy
| | - Teresio Avitabile
- Department of Ophthalmology, University of Catania, 95123 Catania, Italy;
| | - Robert Rejdak
- Department of General and Pediatric Ophthalmology, Medical University of Lublin, 20079 Lublin, Poland; (R.R.); (K.N.)
| | - Katarzyna Nowomiejska
- Department of General and Pediatric Ophthalmology, Medical University of Lublin, 20079 Lublin, Poland; (R.R.); (K.N.)
| | - Sandrine Zweifel
- Department of Ophthalmology, University of Zurich, 8091 Zurich, Switzerland; (M.D.T.); (S.Z.)
| | - Yacoub A. Yousef
- Department of Surgery/Ophthalmology, King Hussein Cancer Center, Amman 11941, Jordan;
| | | | - Giovanni Luca Romano
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy; (F.D.); (C.B.); (G.L.R.)
- Center of Research in Ocular Pharmacology—CERFO, University of Catania, 95123 Catania, Italy
| |
Collapse
|
41
|
Comito F, Marchese PV, Ricci AD, Tober N, Peterle C, Sperandi F, Melotti B. Systemic and liver-directed therapies in metastatic uveal melanoma: state-of-the-art and novel perspectives. Future Oncol 2021; 17:4583-4606. [PMID: 34431316 DOI: 10.2217/fon-2021-0318] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Metastatic uveal melanoma (MUM) is the most common form of noncutaneous melanoma. It is different from its cutaneous counterpart and is characterized by a very poor prognosis. Despite groundbreaking improvements in the treatment of cutaneous melanoma, there have been few advances in the treatment of MUM, and standard treatments for MUM have not been defined. We performed a systematic review focusing our attention on all interventional studies, ongoing or already published, concerning the treatment of MUM. We present results from studies of chemotherapy, targeted therapy, immunotherapy and liver-directed therapies. Although the results in this setting have been disappointing until now, trials investigating novel immunotherapeutic strategies alone and in combination with targeted agents and liver-directed therapies are ongoing.
Collapse
Affiliation(s)
- Francesca Comito
- Division of Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna.,Department of Experimental, Diagnostic & Specialty Medicine, S. Orsola-Malpighi University Hospital of Bologna 40138, Italy
| | - Paola Valeria Marchese
- Department of Experimental, Diagnostic & Specialty Medicine, S. Orsola-Malpighi University Hospital of Bologna 40138, Italy
| | - Angela Dalia Ricci
- Department of Experimental, Diagnostic & Specialty Medicine, S. Orsola-Malpighi University Hospital of Bologna 40138, Italy
| | - Nastassja Tober
- Department of Experimental, Diagnostic & Specialty Medicine, S. Orsola-Malpighi University Hospital of Bologna 40138, Italy
| | - Chiara Peterle
- Department of Experimental, Diagnostic & Specialty Medicine, S. Orsola-Malpighi University Hospital of Bologna 40138, Italy
| | - Francesca Sperandi
- Division of Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna
| | - Barbara Melotti
- Division of Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna
| |
Collapse
|
42
|
Mercer-Smith AR, Findlay IA, Bomba HN, Hingtgen SD. Intravenously Infused Stem Cells for Cancer Treatment. Stem Cell Rev Rep 2021; 17:2025-2041. [PMID: 34138421 DOI: 10.1007/s12015-021-10192-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2021] [Indexed: 01/14/2023]
Abstract
Despite the recent influx of immunotherapies and small molecule drugs to treat tumors, cancer remains a leading cause of death in the United States, in large part due to the difficulties of treating metastatic cancer. Stem cells, which are inherently tumoritropic, provide a useful drug delivery vehicle to target both primary and metastatic tumors. Intravenous infusions of stem cells carrying or secreting therapeutic payloads show significant promise in the treatment of cancer. Stem cells may be engineered to secrete cytotoxic products, loaded with oncolytic viruses or nanoparticles containing small molecule drugs, or conjugated with immunotherapies. Herein we describe these preclinical and clinical studies, discuss the distribution and migration of stem cells following intravenous infusion, and examine both the limitations of and the methods to improve the migration and therapeutic efficacy of tumoritropic, therapeutic stem cells.
Collapse
Affiliation(s)
- Alison R Mercer-Smith
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, North Carolina, Chapel Hill, 27599, USA
| | - Ingrid A Findlay
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, North Carolina, Chapel Hill, 27599, USA
| | - Hunter N Bomba
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, North Carolina, Chapel Hill, 27599, USA
| | - Shawn D Hingtgen
- Division of Pharmacoengineering and Molecular Pharmaceutics, UNC Eshelman School of Pharmacy, The University of North Carolina at Chapel Hill, North Carolina, Chapel Hill, 27599, USA. .,Department of Neurosurgery, The University of North Carolina at Chapel Hill, North Carolina, Chapel Hill, 27599, USA.
| |
Collapse
|
43
|
Farrera-Sal M, Moya-Borrego L, Bazan-Peregrino M, Alemany R. Evolving Status of Clinical Immunotherapy with Oncolytic Adenovirus. Clin Cancer Res 2021; 27:2979-2988. [PMID: 33526422 DOI: 10.1158/1078-0432.ccr-20-1565] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 12/02/2020] [Accepted: 01/15/2021] [Indexed: 11/16/2022]
Abstract
Cancer immunotherapy targeting immune checkpoint inhibitors shows efficacy in several human cancers, but "cold tumors" that lack immune cells are typically unresponsive. Among the potential therapeutic approaches that could "heat" or promote lymphocyte infiltration of cold tumors, oncolytic viruses have attracted interest for their lytic and immunogenic mechanisms of action. In this article, we review the use of oncolytic adenoviruses in cancer immunotherapy, with a particular focus on preclinical and clinical data of oncolytic adenovirus-triggered immune responses against tumor antigens. We also discuss parameters to consider in clinical trial design and the combination of oncolytic adenoviruses with conventional treatments or other immunotherapies.
Collapse
Affiliation(s)
- Martí Farrera-Sal
- ProCure Program, IDIBELL-Institut Català d'Oncologia, Barcelona, Spain.,VCN Biosciences SL, Barcelona, Spain
| | | | | | - Ramon Alemany
- ProCure Program, IDIBELL-Institut Català d'Oncologia, Barcelona, Spain.
| |
Collapse
|
44
|
Lin W, Zhao Y, Zhong L. Current strategies of virotherapy in clinical trials for cancer treatment. J Med Virol 2021; 93:4668-4692. [PMID: 33738818 DOI: 10.1002/jmv.26947] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 02/12/2021] [Accepted: 03/15/2021] [Indexed: 12/19/2022]
Abstract
As a novel immune-active agent for cancer treatment, viruses have the ability of infecting and replicating in tumor cells. The safety and efficacy of viruses has been tested and confirmed in preclinical and clinical trials. In the last decade, virotherapy has been adopted as a monotherapy or combined therapy with immunotherapy, chemotherapy, or radiotherapy, showing promising outcomes against cancer. In this review, the current strategies of viruses used in clinical trials are classified and described. Besides this, the challenge and future prospects of virotherapy in the management for cancer patients are discussed in this review.
Collapse
Affiliation(s)
- Weijian Lin
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, China
| | - Yongxiang Zhao
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, China
| | - Liping Zhong
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, China
| |
Collapse
|
45
|
Villani A, Scalvenzi M, Fabbrocini G, Ocampo-Candiani J, Ocampo-Garza SS. Looking into a Better Future: Novel Therapies for Metastatic Melanoma. Dermatol Ther (Heidelb) 2021; 11:751-767. [PMID: 33866515 PMCID: PMC8163929 DOI: 10.1007/s13555-021-00525-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Indexed: 02/06/2023] Open
Abstract
Even though melanoma represents a small percentage of all cutaneous cancers, it is responsible for most deaths from skin neoplasms. In early stages it can be successfully treated with surgery, but as the disease expands the survival rate drops significantly. For many years the mainstay of treatment for metastatic melanoma was chemotherapeutic agents, even though they failed to prove survival prolongation. After the advent of ipilimumab, a survival benefit and better overall response rate could be offered to the patients. Other new therapies, such as immunotherapies, targeted therapies, vaccines, and small molecules, are currently being studied. Also, combination regimens have demonstrated superiority to some monotherapies. Nowadays, ipilimumab should no longer be considered the first-line therapy given its severe toxicity and lower efficacy, while nivolumab remains efficacious and has a good safety profile. T-VEC as monotherapy has been shown to be an elegant alternative even for the elderly or cases of head and neck melanomas. If the BRAF mutation status is positive, the combination of dabrafenib and trametinib could be an option to consider. Despite the success of the novel treatments, their effectiveness is still limited. New studies have opened up new avenues for future research in melanoma treatment, which is expected to lead to better therapeutic outcomes for our patients. The objective of this review is to discuss the novel therapies for metastatic melanoma that have been tested in humans during the last 3 years to obtain a sharper perspective of the available treatment options for specific patient characteristics.
Collapse
Affiliation(s)
- Alessia Villani
- Dermatology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy.
| | - Massimiliano Scalvenzi
- Dermatology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Gabriella Fabbrocini
- Dermatology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Jorge Ocampo-Candiani
- Dermatology Department, Universidad Autónoma de Nuevo León, University Hospital "Dr. José Eleuterio González", Monterrey, NL, Mexico
| | - Sonia Sofía Ocampo-Garza
- Dermatology Unit, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy.,Dermatology Department, Universidad Autónoma de Nuevo León, University Hospital "Dr. José Eleuterio González", Monterrey, NL, Mexico
| |
Collapse
|
46
|
Briolay T, Petithomme T, Fouet M, Nguyen-Pham N, Blanquart C, Boisgerault N. Delivery of cancer therapies by synthetic and bio-inspired nanovectors. Mol Cancer 2021; 20:55. [PMID: 33761944 PMCID: PMC7987750 DOI: 10.1186/s12943-021-01346-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 03/05/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND As a complement to the clinical development of new anticancer molecules, innovations in therapeutic vectorization aim at solving issues related to tumor specificity and associated toxicities. Nanomedicine is a rapidly evolving field that offers various solutions to increase clinical efficacy and safety. MAIN: Here are presented the recent advances for different types of nanovectors of chemical and biological nature, to identify the best suited for translational research projects. These nanovectors include different types of chemically engineered nanoparticles that now come in many different flavors of 'smart' drug delivery systems. Alternatives with enhanced biocompatibility and a better adaptability to new types of therapeutic molecules are the cell-derived extracellular vesicles and micro-organism-derived oncolytic viruses, virus-like particles and bacterial minicells. In the first part of the review, we describe their main physical, chemical and biological properties and their potential for personalized modifications. The second part focuses on presenting the recent literature on the use of the different families of nanovectors to deliver anticancer molecules for chemotherapy, radiotherapy, nucleic acid-based therapy, modulation of the tumor microenvironment and immunotherapy. CONCLUSION This review will help the readers to better appreciate the complexity of available nanovectors and to identify the most fitting "type" for efficient and specific delivery of diverse anticancer therapies.
Collapse
Affiliation(s)
- Tina Briolay
- Université de Nantes, Inserm, CRCINA, F-44000, Nantes, France
| | | | - Morgane Fouet
- Université de Nantes, Inserm, CRCINA, F-44000, Nantes, France
| | | | | | | |
Collapse
|
47
|
Masaoutis C, Kokkali S, Theocharis S. Immunotherapy in uveal melanoma: novel strategies and opportunities for personalized treatment. Expert Opin Investig Drugs 2021; 30:555-569. [PMID: 33650931 DOI: 10.1080/13543784.2021.1898587] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Uveal melanoma (UM) is the most common intraocular cancer and represents a discrete subtype of melanoma. Metastatic disease, which occurs in half of patients, has a dismal prognosis. Immunotherapy with immune checkpoint inhibitors has produced promising results in cutaneous melanoma but has failed to show analogous efficacy in metastatic UM. This is attributable to UM's distinct genetics and its complex interaction with the immune system. Hence, more efficacious immunotherapeutic approaches are under investigation. AREAS COVERED We discuss those novel immunotherapeutic strategies in clinical and preclinical studies for advanced disease and which are thought to overcome the hurdles set by UM in terms of immune recognition. We also highlight the need to determine predictive markers in relation to these strategies to improve clinical outcomes. We used a simple narrative analysis to summarize the data. The search methodology is located in the Introduction. EXPERT OPINION Novel immunotherapeutic strategies focus on transforming immune excluded tumor microenvironment in metastatic UM to T cell inflamed. Preliminary results of approaches such as vaccines, adoptive cell transfer and other novel molecules are encouraging. Factors such as HLA compatibility and expression level of targeted antigens should be considered to optimize personalized management.
Collapse
Affiliation(s)
- Christos Masaoutis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Stefania Kokkali
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, Athens, Greece.,First Medical Oncology Clinic, Saint-Savvas Anticancer Hospital, Athens, Greece
| | - Stamatios Theocharis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
48
|
Liu S, Zhang J, Fang S, Zhang Q, Zhu G, Tian Y, Zhao M, Liu F. Macrophage polarization contributes to the efficacy of an oncolytic HSV-1 targeting human uveal melanoma in a murine xenograft model. Exp Eye Res 2021; 202:108285. [PMID: 33039456 DOI: 10.1016/j.exer.2020.108285] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 09/04/2020] [Accepted: 09/29/2020] [Indexed: 12/28/2022]
Abstract
Uveal melanoma (UM), the most common primary malignant tumor of the eye in adults, is difficult-to-treat. UM has a relatively high mortality secondary to distant metastasis and poor overall survival with existing therapies. The oncolytic virus herpes simplex virus type-1 (HSV-1) has been approved for clinical use in melanoma. This double-stranded DNA virus was suspected to directly activate lysis specifically in neoplastic cells. We tested the antitumor efficacy of recombinant oncolytic HSV-1-EGFP (oHSV-EGFP) in UM and characterized the local and systemic antitumor innate immune response in a murine xenograft model. We first determined the efficacy of the oncolytic virus in 92.1, MUM2B and MP41 cell lines. In murine xenograft models, oHSV-EGFP reduced intraocular tumors as well as systemic subcutaneous tumors. A significant change in cytokines was observed in viral infected cells, especially a rise in IFNγ. In vivo analyses showed increased anti-tumorigenic M1 macrophages and decreased pro-tumorigenic M2 macrophages in peripheral blood, and intraocular and distant tumors after intravitreal viral treatment. Increased infiltration of natural killer cells and mature dendritic cells was also detected after viral treatment. In addition, no virus was detected in major organs after the treatment. Our data support that oHSV-EGFP is effective, neoplasm specific, immune active and safe, providing possible clinical translatable options to treat ocular and metastatic UM.
Collapse
Affiliation(s)
- Sisi Liu
- Department of Ophthalmology, Peking University People's Hospital, Eye Diseases and Optometry Institute, Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center, Beijing, China
| | - Junwen Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Sheng Fang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Qing Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Guidong Zhu
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Yifu Tian
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Mingwei Zhao
- Department of Ophthalmology, Peking University People's Hospital, Eye Diseases and Optometry Institute, Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center, Beijing, China.
| | - Fusheng Liu
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing Laboratory of Biomedical Materials, Beijing, China.
| |
Collapse
|
49
|
Ortega MA, Fraile-Martínez O, García-Honduvilla N, Coca S, Álvarez-Mon M, Buján J, Teus MA. Update on uveal melanoma: Translational research from biology to clinical practice (Review). Int J Oncol 2020; 57:1262-1279. [PMID: 33173970 PMCID: PMC7646582 DOI: 10.3892/ijo.2020.5140] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 09/24/2020] [Indexed: 02/06/2023] Open
Abstract
Uveal melanoma is the most common type of intraocular cancer with a low mean annual incidence of 5‑10 cases per million. Tumours are located in the choroid (90%), ciliary body (6%) or iris (4%) and of 85% are primary tumours. As in cutaneous melanoma, tumours arise in melanocytes; however, the characteristics of uveal melanoma differ, accounting for 3‑5% of melanocytic cancers. Among the numerous risk factors are age, sex, genetic and phenotypic predisposition, the work environment and dermatological conditions. Management is usually multidisciplinary, including several specialists such as ophthalmologists, oncologists and maxillofacial surgeons, who participate in the diagnosis, treatment and complex follow‑up of these patients, without excluding the management of the immense emotional burden. Clinically, uveal melanoma generates symptoms that depend as much on the affected ocular globe site as on the tumour size. The anatomopathological study of uveal melanoma has recently benefited from developments in molecular biology. In effect, disease classification or staging according to molecular profile is proving useful for the assessment of this type of tumour. Further, the improved knowledge of tumour biology is giving rise to a more targeted approach to diagnosis, prognosis and treatment development; for example, epigenetics driven by microRNAs as a target for disease control. In the present study, the main epidemiological, clinical, physiopathological and molecular features of this disease are reviewed, and the associations among all these factors are discussed.
Collapse
Affiliation(s)
- Miguel A. Ortega
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, 28871 Madrid
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid
- University Center for The Defense of Madrid (CUD-ACD), 28047 Madrid
| | - Oscar Fraile-Martínez
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, 28871 Madrid
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, 28871 Madrid
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid
- University Center for The Defense of Madrid (CUD-ACD), 28047 Madrid
| | - Santiago Coca
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, 28871 Madrid
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid
- University Center for The Defense of Madrid (CUD-ACD), 28047 Madrid
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, 28871 Madrid
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid
- University Center for The Defense of Madrid (CUD-ACD), 28047 Madrid
- Internal and Oncology Service (CIBER-EHD), University Hospital Príncipe de Asturias, Alcalá de Henares, 28805 Madrid
| | - Julia Buján
- Department of Medicine and Medical Specialties, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, 28871 Madrid
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid
- University Center for The Defense of Madrid (CUD-ACD), 28047 Madrid
| | - Miguel A. Teus
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, 28871 Madrid
- Ophthalmology Service, University Hospital Príncipe de Asturias, Alcalá de Henares, 28805 Madrid, Spain
| |
Collapse
|
50
|
Liu S, Liu F, Zhao M, Zhang J. Antitumor Efficacy of Oncolytic Herpes Virus Type 1 Armed with GM-CSF in Murine Uveal Melanoma Xenografts. Cancer Manag Res 2020; 12:11803-11812. [PMID: 33239914 PMCID: PMC7680789 DOI: 10.2147/cmar.s274605] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/29/2020] [Indexed: 12/17/2022] Open
Abstract
Background Uveal melanoma (UM) is the most common primary intraocular tumor in adults with a high incidence of metastasis. Standard care therapies for UM include enucleation and radiation, which are minimally effective in prolonging patient survival. Oncolytic virus treatment has become a new trend in cancer field. Of which, oncolytic herpes simplex virus type 1 (HSV-1) therapy is one of the most effective antitumor treatments. Here, we established an oncolytic HSV-1 encoding granulocyte-macrophage colony-stimulating factor (GM-CSF), tested its efficacy in UM therapy, and investigated the innate immune response induced by this virus. Methods Oncolytic HSV-1 expressing GM-CSF (HSV-GM-CSF) was constructed, then verified using qPCR and Western blot assays. Cell viability assays and transmission electron microscopy were conducted on three UM cell lines, MUM2B, 92.1, and MP41, to assess the cell-killing ability and virus infection of this virus. For in vivo experiments, BALB/c-nude mice in situ UM xenografts were established to testify the efficacy of the oncolytic virus, oncolytic HSV-1, and HSV-GM-CSF groups, respectively. IVIS images, ocular volumes, mice weights, and survivals were tracked to see the efficacy of the virus. Hematoxylin and eosin staining, immunohistochemistry, and flow cytometry analyses were conducted to demonstrate the immune activity after virus treatment. Results All three tested UM cell lines were sensitive to infection by HSV-GM-CSF. In vivo xenograft experiments revealed that oncolytic virus HSV-1 reduced UM tumor volume and that oncolytic virus HSV-1 armed with GM-CSF enhanced the antitumor effect compared with unmodified HSV-1. The bodyweights of untreated control group mice were significantly lower than those of mice in either virus-treated group (HSV-1 or HSV-GM-CSF). Follow-up survivals were prolonged in the virus-treated groups compared with the control group and were prolonged to a greater extent in the HSV-GM-CSF group than in the HSV-1 group. Macrophage stimulation was observed following HSV-GM-CSF treatment. Conclusion Our results indicate that the recombinant oncolytic virus HSV-GM-CSF is a potential therapeutic treatment for UM.
Collapse
Affiliation(s)
- Sisi Liu
- Department of Ophthalmology, Peking University People's Hospital, Eye Diseases and Optometry Institute, Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center, Beijing, People's Republic of China
| | - Fusheng Liu
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Beijing Laboratory of Biomedical Materials, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing 100070, People's Republic of China
| | - Mingwei Zhao
- Department of Ophthalmology, Peking University People's Hospital, Eye Diseases and Optometry Institute, Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center, Beijing, People's Republic of China
| | - Junwen Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Beijing Laboratory of Biomedical Materials, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing 100070, People's Republic of China
| |
Collapse
|