1
|
Zinovieva M, Ryapolova A, Karabelsky A, Minskaia E. Oncolytic Vesicular Stomatitis Virus: Optimisation Strategies for Anti-Cancer Therapies. FRONT BIOSCI-LANDMRK 2024; 29:374. [PMID: 39614430 DOI: 10.31083/j.fbl2911374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 07/22/2024] [Accepted: 07/31/2024] [Indexed: 12/01/2024]
Abstract
Oncolytic viruses (OVs) represent a targeted anti-cancer therapy approach due to their ability not only to selectively infect and destroy malignant cells but also to induce an immune response. Vesicular stomatitis virus (VSV) offers a promising platform due to its low prevalence and pathogenicity in humans, lack of pre-existing immunity, easily manipulated genome, rapid growth to high titers in a broad range of cell lines, and inability to integrate into the host genome. However, despite its many advantages, many unresolved problems remain: problematic production based on the reverse genetics system, oncological selectivity, and the overall effectiveness of VSV monotherapy. This review will discuss various attempts at viral genome modifications aimed at improving the oncolytic properties of VSV. These strategies include inhibition of viral genes, modification of genes responsible for targeting cancer cells over healthy ones, insertion of foreign genes for boosting immune response, and changing the order of viral and inserted foreign genes. In addition, possible ways to improve VSV-based anti-tumor therapy and achieve higher efficiency will be considered by evaluating the effectiveness of various delivery methods as well as discussing treatment options by combining VSV with other groups of anticancer drugs.
Collapse
Affiliation(s)
- Margarita Zinovieva
- Department of Gene Therapy, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Anastasia Ryapolova
- Department of Gene Therapy, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Alexander Karabelsky
- Department of Gene Therapy, Sirius University of Science and Technology, 354340 Sochi, Russia
| | - Ekaterina Minskaia
- Department of Gene Therapy, Sirius University of Science and Technology, 354340 Sochi, Russia
| |
Collapse
|
2
|
Roser LA, Sommer C, Ortega Iannazzo S, Sakellariou C, Waibler Z, Gogesch P. Revival of recombinant IL-2 therapy - approaches from the past until today. J Immunotoxicol 2024; 21:S38-S47. [PMID: 39655498 DOI: 10.1080/1547691x.2024.2335219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 03/04/2024] [Accepted: 03/21/2024] [Indexed: 12/18/2024] Open
Abstract
Interleukin-2 (IL-2) was one of the first cytokines discovered and its central role in T cell function soon led to the notion that the cytokine could specifically activate immune cells to combat cancer cells. Recombinant human IL-2 (recIL-2) belonged to the first anti-cancer immunotherapeutics that received marketing authorization and while it mediated anti-tumor effects in some cancer entities, treatment was associated with severe and systemic side effects. RecIL-2 holds an exceptional therapeutic potential, which can either lead to stimulation of the immune system - favorable during cancer treatment - or immunosuppression - used for treatment of inflammatory diseases such as autoimmunity. Due to these pleiotropic immune effects, recIL-2 therapy is still a hot topic in research and modified recIL-2 drug candidates show ameliorated efficacy and safety in pre-clinical and clinical studies. The Immune Safety Avatar (imSAVAR) consortium aims to systemically assess mechanisms leading to adverse events provoked by recIL-2 immunotherapy as a use case in order to aid safety evaluation of future recIL-2-based therapies. Here, we summarize the historical use of recIL-2 therapy, associated side effects, and describe the molecular basis of the dual role of IL-2. Finally, an overview of new recIL-2 compounds and delivery systems, which are currently being developed, will be given, highlighting a possible comeback of recIL-2 therapy.
Collapse
Affiliation(s)
- Luise A Roser
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
| | - Charline Sommer
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, Preclinical Pharmacology and In-Vitro Toxicology, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
- Member of the Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Hannover, Germany
| | | | | | - Zoe Waibler
- Paul-Ehrlich-Institut, Division of Immunology, Langen, Germany
| | | |
Collapse
|
3
|
Wu M, Wang Y, Wu C, Huang H, Zhou X, Wang J, Xiong S, Dong C. A novel vesicular stomatitis virus armed with IL-2 mimic for oncolytic therapy. Virol Sin 2024; 39:821-832. [PMID: 39299564 DOI: 10.1016/j.virs.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024] Open
Abstract
Oncolytic virus (OV) is increasingly being recognized as a novel vector in cancer immunotherapy. Increasing evidence suggests that OV has the ability to change the immune status of tumor microenvironment, so called transformation of 'cold' tumors into 'hot' tumors. The improved anti-tumor immunity can be induced by OV and further enhanced through the combination of various immunomodulators. The Neo-2/15 is a newly de novo synthesized cytokine that functions as both IL-2 and IL-15. However, it specifically lacks the binding site of IL-2 receptor α subunit (CD25), therefore unable to induce the Treg proliferation. In present study, a recombinant vesicular stomatitis virus expressing the Neo-2/15 (VSVM51R-Neo-2/15) was generated. Intratumoral delivery of VSVM51R-Neo-2/15 efficiently inhibited tumor growth in mice without causing the IL-2-related toxicity previously observed in clinic. Moreover, treatment with VSVM51R-Neo-2/15 increased the number of activated CD8+ T cells but not Treg cells in tumors. More tumor-bearing mice were survival with VSVM51R-Neo-2/15 treatment, and the surviving mice displayed enhanced protection against tumor cell rechallenge due to the induced anti-tumor immunity. In addition, combination therapy of OV and anti-PD-L1 immune checkpoint inhibitors further enhanced the anti-tumor immune response. These findings suggest that our novel VSVM51R-Neo-2/15 can effectively inhibit the tumor growth and enhance the sensitivity to immune checkpoint inhibitors, providing promising attempts for further clinical trials.
Collapse
Affiliation(s)
- Manman Wu
- The Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, China
| | - Yiwei Wang
- The Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, China
| | - Chuanjian Wu
- The Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, China
| | - Huang Huang
- Department of Cardiology, No. 981 Hospital, PLA (People's Liberation Army of China), Chengde 067000, China
| | - Xinyuan Zhou
- Institute of Immunology, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China.
| | - Jun Wang
- The Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, China.
| | - Sidong Xiong
- The Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, China.
| | - Chunsheng Dong
- The Institutes of Biology and Medical Sciences, MOE Key Laboratory of Geriatric Diseases and Immunology, Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215123, China.
| |
Collapse
|
4
|
Guan X, Guo H, Guo Y, Han Q, Li Z, Zhang C. Perforin 1 in Cancer: Mechanisms, Therapy, and Outlook. Biomolecules 2024; 14:910. [PMID: 39199299 PMCID: PMC11352983 DOI: 10.3390/biom14080910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 09/01/2024] Open
Abstract
PRF1 (perforin 1) is a key cytotoxic molecule that plays a crucial role in the killing function of natural killer (NK) cells and cytotoxic T lymphocytes (CTLs). Recent studies have focused on PRF1's role in cancer development, progression, and prognosis. Studies have shown that aberrant PRF1 expression has a significant role to play in cancer development and progression. In some cancers, high expression of the PRF1 gene is associated with a better prognosis for patients, possibly because it helps enhance the body's immune response to tumors. However, some studies have also shown that the absence of PRF1 may make it easier for tumors to evade the body's immune surveillance, thus affecting patient survival. Furthermore, recent studies have explored therapeutic strategies based on PRF1, such as enhancing the ability of immune cells to kill cancer cells by boosting PRF1 activity. In addition, they have improved the efficacy of immunotherapy by modulating its expression to enhance the effectiveness of the treatment. Based on these findings, PRF1 may be a valuable biomarker both for the treatment of cancer and for its prognosis in the future. To conclude, PRF1 has an important biological function and has clinical potential for the treatment of cancer, which indicates that it deserves more research and development in the future.
Collapse
Affiliation(s)
- Xiaoya Guan
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, China; (H.G.); (Y.G.); (Q.H.); (Z.L.)
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Huina Guo
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, China; (H.G.); (Y.G.); (Q.H.); (Z.L.)
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Yujia Guo
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, China; (H.G.); (Y.G.); (Q.H.); (Z.L.)
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Qi Han
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, China; (H.G.); (Y.G.); (Q.H.); (Z.L.)
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Zhongxun Li
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, China; (H.G.); (Y.G.); (Q.H.); (Z.L.)
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Chunming Zhang
- Shanxi Key Laboratory of Otorhinolaryngology Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, China; (H.G.); (Y.G.); (Q.H.); (Z.L.)
- Shanxi Province Clinical Medical Research Center for Precision Medicine of Head and Neck Cancer, First Hospital of Shanxi Medical University, Taiyuan 030001, China
- Department of Otolaryngology Head & Neck Surgery, First Hospital of Shanxi Medical University, Taiyuan 030001, China
| |
Collapse
|
5
|
Lin C, Teng W, Tian Y, Li S, Xia N, Huang C. Immune landscape and response to oncolytic virus-based immunotherapy. Front Med 2024; 18:411-429. [PMID: 38453818 DOI: 10.1007/s11684-023-1048-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 11/15/2023] [Indexed: 03/09/2024]
Abstract
Oncolytic virus (OV)-based immunotherapy has emerged as a promising strategy for cancer treatment, offering a unique potential to selectively target malignant cells while sparing normal tissues. However, the immunosuppressive nature of tumor microenvironment (TME) poses a substantial hurdle to the development of OVs as effective immunotherapeutic agents, as it restricts the activation and recruitment of immune cells. This review elucidates the potential of OV-based immunotherapy in modulating the immune landscape within the TME to overcome immune resistance and enhance antitumor immune responses. We examine the role of OVs in targeting specific immune cell populations, including dendritic cells, T cells, natural killer cells, and macrophages, and their ability to alter the TME by inhibiting angiogenesis and reducing tumor fibrosis. Additionally, we explore strategies to optimize OV-based drug delivery and improve the efficiency of OV-mediated immunotherapy. In conclusion, this review offers a concise and comprehensive synopsis of the current status and future prospects of OV-based immunotherapy, underscoring its remarkable potential as an effective immunotherapeutic agent for cancer treatment.
Collapse
Affiliation(s)
- Chaolong Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, 361102, China
| | - Wenzhong Teng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, 361102, China
| | - Yang Tian
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, 361102, China
| | - Shaopeng Li
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, 361102, China
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China.
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, 361102, China.
| | - Chenghao Huang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China.
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, 361102, China.
| |
Collapse
|
6
|
Quixabeira DCA, Pakola S, Jirovec E, Havunen R, Basnet S, Santos JM, Kudling TV, Clubb JHA, Haybout L, Arias V, Grönberg-Vähä-Koskela S, Cervera-Carrascon V, Kerkelä E, Pasanen A, Anttila M, Tapper J, Kanerva A, Hemminki A. Boosting cytotoxicity of adoptive allogeneic NK cell therapy with an oncolytic adenovirus encoding a human vIL-2 cytokine for the treatment of human ovarian cancer. Cancer Gene Ther 2023; 30:1679-1690. [PMID: 37949944 PMCID: PMC10721546 DOI: 10.1038/s41417-023-00674-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/31/2023] [Accepted: 09/29/2023] [Indexed: 11/12/2023]
Abstract
Despite good results in the treatment of hematological malignancies, Natural killer (NK) cells have shown limited effectiveness in solid tumors, such as ovarian cancer (OvCa). Here, we assessed the potential of an oncolytic adenovirus expressing a variant interleukin-2 (vIL-2) cytokine, Ad5/3-E2F-d24-vIL2 (vIL-2 virus), also known as TILT-452, to enhance NK cell therapy efficacy in human OvCa ex vivo. Human OvCa surgical specimens were processed into single-cell suspensions and NK cells were expanded from healthy blood donors. OvCa sample digests were co-cultured ex vivo with NK cells and vIL-2 virus and cancer cell killing potential assessed in real time through cell impedance measurement. Proposed therapeutic combination was evaluated in vivo with an OvCa patient-derived xenograft (PDX) in mice. Addition of vIL-2 virus significantly enhanced NK cell therapy killing potential in treated OvCa co-cultures. Similarly, vIL-2 virus in combination with NK cell therapy promoted the best in vivo OvCa tumor control. Mechanistically, vIL-2 virus induced higher percentages of granzyme B in NK cells, and CD8+ T cells, while T regulatory cell proportions remained comparable to NK cell monotherapy in vivo. Ad5/3-E2F-d24-vIL2 virus treatment represents a promising strategy to boost adoptive NK cell therapeutic effect in human OvCa.
Collapse
Affiliation(s)
- D C A Quixabeira
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - S Pakola
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - E Jirovec
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - R Havunen
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - S Basnet
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - J M Santos
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - T V Kudling
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - J H A Clubb
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - L Haybout
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - V Arias
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - S Grönberg-Vähä-Koskela
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - V Cervera-Carrascon
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - E Kerkelä
- Finnish Red Cross Blood Service, Helsinki, Finland
| | - A Pasanen
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - M Anttila
- Pathology, Finnish Food Authority, Helsinki, Finland
| | - J Tapper
- Department of Obstetrics and Gynecology, Helsinki University Central Hospital, Helsinki, Finland
| | - A Kanerva
- Department of Obstetrics and Gynecology, Helsinki University Central Hospital, Helsinki, Finland
| | - A Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- TILT Biotherapeutics Ltd, Helsinki, Finland.
- Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.
| |
Collapse
|
7
|
Yoon AR, Hong J, Jung BK, Ahn HM, Zhang S, Yun CO. Oncolytic adenovirus as pancreatic cancer-targeted therapy: Where do we go from here? Cancer Lett 2023; 579:216456. [PMID: 37940067 DOI: 10.1016/j.canlet.2023.216456] [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: 08/18/2023] [Revised: 10/10/2023] [Accepted: 10/17/2023] [Indexed: 11/10/2023]
Abstract
Pancreatic cancer remains one of the deadliest cancers with extremely high mortality rate, and the number of cases is expected to steadily increase with time. Pancreatic cancer is refractory to conventional cancer treatment options, like chemotherapy and radiotherapy, and commercialized immunotherapeutics, owing to its immunosuppressive and desmoplastic phenotype. Due to these reasons, development of an innovative treatment option that can overcome these challenges posed by the pancreatic tumor microenvironment (TME) is in an urgent need. The present review aims to summarize the evolution of oncolytic adenovirus (oAd) engineering and usage as therapeutics (either monotherapy or combination therapy) over the last decade to overcome these hurdles to instigate a potent antitumor effect against desmoplastic and immunosuppressive pancreatic cancer.
Collapse
Affiliation(s)
- A-Rum Yoon
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea; Institute of Nano Science and Technology (INST), Hanyang University, Seoul, Republic of Korea; Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul, Republic of Korea
| | - JinWoo Hong
- GeneMedicine Co., Ltd., 222 Wangsimni-ro, Seongdong-gu, Seoul, Republic of Korea
| | - Bo-Kyeong Jung
- GeneMedicine Co., Ltd., 222 Wangsimni-ro, Seongdong-gu, Seoul, Republic of Korea
| | - Hyo Min Ahn
- GeneMedicine Co., Ltd., 222 Wangsimni-ro, Seongdong-gu, Seoul, Republic of Korea
| | - Songnam Zhang
- Department of Medical Oncology, Yanbian University Hospital, Jilin, China
| | - Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, Republic of Korea; Institute of Nano Science and Technology (INST), Hanyang University, Seoul, Republic of Korea; Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul, Republic of Korea; GeneMedicine Co., Ltd., 222 Wangsimni-ro, Seongdong-gu, Seoul, Republic of Korea.
| |
Collapse
|
8
|
Roser LA, Luckhardt S, Ziegler N, Thomas D, Wagner PV, Damm G, Scheffschick A, Hewitt P, Parnham MJ, Schiffmann S. Immuno-inflammatory in vitro hepatotoxicity models to assess side effects of biologicals exemplified by aldesleukin. Front Immunol 2023; 14:1275368. [PMID: 38045689 PMCID: PMC10693457 DOI: 10.3389/fimmu.2023.1275368] [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: 08/09/2023] [Accepted: 10/27/2023] [Indexed: 12/05/2023] Open
Abstract
Introduction Hepatotoxicity induced by immunotherapeutics is an appearing cause for immune-mediated drug-induced liver injury. Such immuno-toxic mechanisms are difficult to assess using current preclinical models and the incidence is too low to detect in clinical trials. As hepatotoxicity is a frequent reason for post-authorisation drug withdrawal, there is an urgent need for immuno-inflammatory in vitro models to assess the hepatotoxic potential of immuno-modulatory drug candidates. We developed several immuno-inflammatory hepatotoxicity test systems based on recombinant human interleukin-2 (aldesleukin). Methods Co-culture models of primary human CD8+ T cells or NK cells with the hepatocyte cell line HepaRG were established and validated with primary human hepatocytes (PHHs). Subsequently, the HepaRG model was refined by increasing complexity by inclusion of monocyte-derived macrophages (MdMs). The main readouts were cytotoxicity, inflammatory mediator release, surface marker expression and specific hepatocyte functions. Results We identified CD8+ T cells as possible mediators of aldesleukin-mediated hepatotoxicity, with MdMs being implicated in increased aldesleukin-induced inflammatory effects. In co-cultures of CD8+ T cells with MdMs and HepaRG cells, cytotoxicity was induced at intermediate/high aldesleukin concentrations and perforin was upregulated. A pro-inflammatory milieu was created measured by interleukin-6 (IL-6), c-reactive protein (CRP), interferon gamma (IFN-γ), and monocyte chemoattractant protein-1 (MCP-1) increase. NK cells responded to aldesleukin, however, only minor aldesleukin-induced cytotoxic effects were measured in co-cultures. Results obtained with HepaRG cells and with PHHs were comparable, especially regarding cytotoxicity, but high inter-donor variations limited meaningfulness of the PHH model. Discussion The in vitro test systems developed contribute to the understanding of potential key mechanisms in aldesleukin-mediated hepatotoxicity. In addition, they may aid assessment of immune-mediated hepatotoxicity during the development of novel immunotherapeutics.
Collapse
Affiliation(s)
- Luise A. Roser
- Department of Preclinical Research, Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt am Main, Germany
| | - Sonja Luckhardt
- Department of Preclinical Research, Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt am Main, Germany
| | - Nicole Ziegler
- Department of Preclinical Research, Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt am Main, Germany
| | - Dominique Thomas
- Department of Preclinical Research, Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt am Main, Germany
- pharmazentrum frankfurt/ZAFES, Department of Clinical Pharmacology, Goethe-University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Pia Viktoria Wagner
- Department of Preclinical Research, Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt am Main, Germany
| | - Georg Damm
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, Leipzig, Germany
| | - Andrea Scheffschick
- Department of Hepatobiliary Surgery and Visceral Transplantation, University Hospital, Leipzig University, Leipzig, Germany
| | - Philip Hewitt
- Chemical and Preclinical Safety, Merck Healthcare KGaA, Darmstadt, Germany
| | - Michael J. Parnham
- Department of Preclinical Research, Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt am Main, Germany
| | - Susanne Schiffmann
- Department of Preclinical Research, Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), Frankfurt am Main, Germany
- pharmazentrum frankfurt/ZAFES, Department of Clinical Pharmacology, Goethe-University Hospital Frankfurt, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases (CIMD), Frankfurt am Main, Germany
| |
Collapse
|
9
|
Quixabeira DCA, Jirovec E, Pakola S, Havunen R, Basnet S, Santos JM, Kudling TV, Clubb JHA, Haybout L, Arias V, Grönberg-Vähä-Koskela S, Cervera-Carrascon V, Pasanen A, Anttila M, Tapper J, Kanerva A, Hemminki A. Improving the cytotoxic response of tumor-infiltrating lymphocytes towards advanced stage ovarian cancer with an oncolytic adenovirus expressing a human vIL-2 cytokine. Cancer Gene Ther 2023; 30:1543-1553. [PMID: 37666898 PMCID: PMC10645590 DOI: 10.1038/s41417-023-00658-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/19/2023] [Accepted: 08/02/2023] [Indexed: 09/06/2023]
Abstract
While the presence of tumor-infiltrating lymphocytes (TILs) associates with improved survival prognosis in ovarian cancer (OvCa) patients, TIL therapy benefit is limited. Here, we evaluated an oncolytic adenovirus coding for a human variant IL-2 (vIL-2) cytokine, Ad5/3-E2F-d24-vIL2 (vIL-2 virus), also known as TILT-452, as an immunotherapeutic strategy to enhance TIL responsiveness towards advanced stage OvCa tumors. Fragments of resected human OvCa tumors were processed into single-cell suspensions, and autologous TILs were expanded from said samples. OvCa tumor specimens were co-cultured with TILs plus vIL-2 virus, and cell killing was assessed in real time through cell impedance measurement. Combination therapy was further evaluated in vivo through a patient-derived xenograft (PDX) ovarian cancer murine model. The combination of vIL-2 virus plus TILs had best cancer cell killing ex vivo compared to TILs monotherapy. These results were supported by an in vivo experiment, where the best OvCa tumor control was obtained when vIL-2 virus was added to TIL therapy. Furthermore, the proposed therapy induced a highly cytotoxic phenotype demonstrated by increased granzyme B intensity in NK cells, CD4+ T, and CD8+ T cells in treated tumors. Our results demonstrate that Ad5/3-E2F-d24-vIL2 therapy consistently improved TILs therapy cytotoxicity in treated human OvCa tumors.
Collapse
Affiliation(s)
- D C A Quixabeira
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - E Jirovec
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - S Pakola
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - R Havunen
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - S Basnet
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - J M Santos
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - T V Kudling
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - J H A Clubb
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - L Haybout
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - V Arias
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - S Grönberg-Vähä-Koskela
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - V Cervera-Carrascon
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - A Pasanen
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - M Anttila
- Pathology, Finnish Food Authority, Helsinki, Finland
| | - J Tapper
- Department of Obstetrics and Gynecology, Helsinki University Central Hospital, Helsinki, Finland
| | - A Kanerva
- Department of Obstetrics and Gynecology, Helsinki University Central Hospital, Helsinki, Finland
| | - A Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- TILT Biotherapeutics Ltd, Helsinki, Finland.
- Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.
| |
Collapse
|
10
|
Wang H, Borlongan M, Hemminki A, Basnet S, Sah N, Kaufman HL, Rabkin SD, Saha D. Viral Vectors Expressing Interleukin 2 for Cancer Immunotherapy. Hum Gene Ther 2023; 34:878-895. [PMID: 37578106 PMCID: PMC10623065 DOI: 10.1089/hum.2023.099] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/09/2023] [Indexed: 08/15/2023] Open
Abstract
Interleukin 2 (IL-2) plays a crucial role in T cell growth and survival, enhancing the cytotoxic activity of natural killer and cytotoxic T cells and thus functioning as a versatile master proinflammatory anticancer cytokine. The FDA has approved IL-2 cytokine therapy for the treatment of metastatic melanoma and metastatic renal cell carcinoma. However, IL-2 therapy has significant constraints, including a short serum half-life, low tumor accumulation, and life-threatening toxicities associated with high doses. Oncolytic viruses (OVs) offer a promising option for cancer immunotherapy, selectively targeting and destroying cancer cells while sparing healthy cells. Numerous studies have demonstrated the successful delivery of IL-2 to the tumor microenvironment without compromising safety using OVs such as vaccinia, Sendai, parvo, Newcastle disease, tanapox, and adenoviruses. Additionally, by engineering OVs to coexpress IL-2 with other anticancer transgenes, the immune properties of IL-2 can be further enhanced. Preclinical and clinical studies have shown promising antitumor effects of IL-2-expressing viral vectors, either alone or in combination with other anticancer therapies. This review summarizes the therapeutic potential of IL-2-expressing viral vectors and their antitumor mechanisms of action.
Collapse
Affiliation(s)
- Hongbin Wang
- Department of Pharmaceutical and Biomedical Sciences, California Northstate University College of Pharmacy, Elk Grove, California, USA
- Master of Pharmaceutical Sciences Program, College of Graduate Studies, California Northstate University, Elk Grove, California, USA
| | - Mia Borlongan
- Master of Pharmaceutical Sciences Program, College of Graduate Studies, California Northstate University, Elk Grove, California, USA
| | - 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, Helsinki, Finland
| | - Saru Basnet
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Naresh Sah
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center School of Pharmacy, Abilene, Texas, USA
| | - Howard L. Kaufman
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
- Ankyra Therapeutics, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Samuel D. Rabkin
- Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Dipongkor Saha
- Department of Pharmaceutical and Biomedical Sciences, California Northstate University College of Pharmacy, Elk Grove, California, USA
| |
Collapse
|
11
|
Pakola S, Quixabeira DCA, Kudling TV, Clubb JHA, Grönberg-Vähä-Koskela S, Basnet S, Jirovec E, Arias V, Haybout L, Heiniö C, Santos JM, Cervera-Carrascon V, Havunen R, Anttila M, Hemminki A. An oncolytic adenovirus coding for a variant interleukin 2 cytokine improves response to chemotherapy through enhancement of effector lymphocyte cytotoxicity, fibroblast compartment modulation and mitotic slippage. Front Immunol 2023; 14:1171083. [PMID: 37475863 PMCID: PMC10354511 DOI: 10.3389/fimmu.2023.1171083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 06/20/2023] [Indexed: 07/22/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly treatment-resistant cancer. Currently, the only curative treatment for PDAC is surgery, but most patients are diagnosed with metastatic disease and thus outside the scope of surgery. The majority of metastatic patients receive chemotherapy, but responses are limited. New therapeutics are thus urgently needed for PDAC. One major limitation in treating PDAC has been the highly immunosuppressive tumor microenvironment (TME) which inhibits anti-cancer immune responses. We have constructed an oncolytic adenovirus coding for a variant the interleukin 2 molecule, Ad5/3-E2F-d24-vIL2 (also known as TILT-452, and "vIL-2 virus"), with preferential binding to IL-2 receptors on the surface of effector lymphocytes over T regulatory cells (T regs). In the present study this virus was evaluated in combination with nab-paclitaxel and gemcitabine chemotherapy in Panc02 mouse model. Ad5/3-E2F-d24-vIL2 showed marked PDAC cell killing in vitro, alongside induction of mitotic slippage and immunogenic cell death in PDAC cell lines, when combined with chemotherapy. Increased survival was seen in vivo with 80% of animals surviving long term, when compared to chemotherapy alone. Moreover, combination therapy mediated enhanced tumor growth control, without observable toxicities in internal organs or external features. Survival and tumor control benefits were associated with activation of tumor infiltrating immune cells, downregulation of inhibitory signals, change in fibroblast populations in the tumors and changes in intratumoral cytokines, with increased chemokine amounts (CCL2, CCL3, CCL4) and anti-tumor cytokines (IFN-γ and TNFα). Furthermore, vIL-2 virus in combination with chemotherapy efficiently induced tumor protection upon rechallenge, that was extended to a previously non-encountered cancer cell line. In conclusion, Ad5/3-E2F-d24-vIL2 is a promising immunotherapy candidate when combined with nab-paclitaxel and gemcitabine.
Collapse
Affiliation(s)
- Santeri Pakola
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Dafne C. A. Quixabeira
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd., Helsinki, Finland
| | - Tatiana V. Kudling
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - James H. A. Clubb
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd., Helsinki, Finland
| | - Susanna Grönberg-Vähä-Koskela
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Saru Basnet
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Elise Jirovec
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd., Helsinki, Finland
| | - Victor Arias
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Lyna Haybout
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd., Helsinki, Finland
| | - Camilla Heiniö
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Joao M. Santos
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd., Helsinki, Finland
| | - Victor Cervera-Carrascon
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd., Helsinki, Finland
| | - Riikka Havunen
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd., Helsinki, Finland
| | | | - Akseli Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd., Helsinki, Finland
- Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| |
Collapse
|
12
|
Duan S, Wang S, Qiao L, Yu X, Wang N, Chen L, Zhang X, Zhao X, Liu H, Wang T, Wu Y, Li N, Liu F. Oncolytic Virus-Driven Biotherapies from Bench to Bedside. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206948. [PMID: 36879416 DOI: 10.1002/smll.202206948] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 01/17/2023] [Indexed: 06/08/2023]
Abstract
With advances in cancer biology and an ever-deepening understanding of molecular virology, oncolytic virus (OV)-driven therapies have developed rapidly and become a promising alternative to traditional cancer therapies. In recent years, satisfactory results for oncolytic virus therapy (OVT) are achieved at both the cellular and organismal levels, and efforts are being increasingly directed toward clinical trials. Unfortunately, OVT remains ineffective in these trials, especially when performed using only a single OV reagent. In contrast, integrated approaches, such as using immunotherapy, chemotherapy, or radiotherapy, alongside OVT have demonstrated considerable efficacy. The challenges of OVT in clinical efficacy include the restricted scope of intratumoral injections and poor targeting of intravenous administration. Further optimization of OVT delivery is needed before OVs become a viable therapy for tumor treatment. In this review, the development process and antitumor mechanisms of OVs are introduced. The advances in OVT delivery routes to provide perspectives and directions for the improvement of OVT delivery are highlighted. This review also discusses the advantages and limitations of OVT monotherapy and combination therapy through the lens of recent clinical trials and aims to chart a course toward safer and more effective OVT strategies.
Collapse
Affiliation(s)
- Shijie Duan
- 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, Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Shuhang Wang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lei Qiao
- Colorectal and Henia Minimally Invasive Surgery Unit, Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Xinbo Yu
- 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, Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Nan Wang
- 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, Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Liting Chen
- 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, Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Xinyuan Zhang
- 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, Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Xu Zhao
- 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, Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Hongyu 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, Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Tianye Wang
- 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, Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Ying Wu
- Phase I Clinical Trials Center, The First Hospital of China Medical University, Department of General Practice, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Ning Li
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - 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, Phase I Clinical Trials Center, The First Hospital of China Medical University, Shenyang, 110001, China
| |
Collapse
|
13
|
Lin D, Shen Y, Liang T. Oncolytic virotherapy: basic principles, recent advances and future directions. Signal Transduct Target Ther 2023; 8:156. [PMID: 37041165 PMCID: PMC10090134 DOI: 10.1038/s41392-023-01407-6] [Citation(s) in RCA: 85] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 03/05/2023] [Accepted: 03/14/2023] [Indexed: 04/13/2023] Open
Abstract
Oncolytic viruses (OVs) have attracted growing awareness in the twenty-first century, as they are generally considered to have direct oncolysis and cancer immune effects. With the progress in genetic engineering technology, OVs have been adopted as versatile platforms for developing novel antitumor strategies, used alone or in combination with other therapies. Recent studies have yielded eye-catching results that delineate the promising clinical outcomes that OVs would bring about in the future. In this review, we summarized the basic principles of OVs in terms of their classifications, as well as the recent advances in OV-modification strategies based on their characteristics, biofunctions, and cancer hallmarks. Candidate OVs are expected to be designed as "qualified soldiers" first by improving target fidelity and safety, and then equipped with "cold weapons" for a proper cytocidal effect, "hot weapons" capable of activating cancer immunotherapy, or "auxiliary weapons" by harnessing tactics such as anti-angiogenesis, reversed metabolic reprogramming and decomposing extracellular matrix around tumors. Combinations with other cancer therapeutic agents have also been elaborated to show encouraging antitumor effects. Robust results from clinical trials using OV as a treatment congruously suggested its significance in future application directions and challenges in developing OVs as novel weapons for tactical decisions in cancer treatment.
Collapse
Affiliation(s)
- Danni Lin
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, Zhejiang, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Yinan Shen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, Zhejiang, China
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, Zhejiang, China.
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, Zhejiang, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
14
|
Basnet S, Santos JM, Quixabeira DCA, Clubb JHA, Grönberg-Vähä-Koskela SAM, Arias V, Pakola S, Kudling TV, Heiniö C, Havunen R, Cervera-Carrascon V, Sorsa S, Anttila M, Kanerva A, Hemminki A. Oncolytic adenovirus coding for bispecific T cell engager against human MUC-1 potentiates T cell response against solid tumors. Mol Ther Oncolytics 2023; 28:59-73. [PMID: 36699617 PMCID: PMC9842968 DOI: 10.1016/j.omto.2022.12.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022] Open
Abstract
Immunotherapy with bispecific T cell engagers has shown efficacy in patients with hematologic malignancies and uveal melanoma. Antitumor effects of bispecific T cell engagers in most solid tumors are limited due to their short serum half-life and insufficient tumor concentration. We designed a novel serotype 5/3 oncolytic adenovirus encoding a human mucin1 antibody and the human CD3 receptor, Ad5/3-E2F-d24-aMUC1aCD3 (TILT-321). TILT-321 is engineered to replicate only in cancer cells, leading to a high concentration of the aMUC1aCD3 molecule in the tumor microenvironment. Infection and cell viability assays were performed to determine the oncolytic potential of the novel construct. The functionality of the virus-derived aMUC1aCD3 was evaluated in vitro. When TILT-321 was combined with allogeneic T cells, rapid tumor cell lysis was observed. TILT-321-infected cells secreted functional aMUC1aCD3, as shown by increased T cell activity and its binding to MUC1 and CD3. In vivo, TILT-321 treatment led to effective antitumor efficacy mediated by increased intratumoral T cell activity in an A549 and patient-derived ovarian cancer xenograft mouse model humanized with peripheral blood mononuclear cells (PBMC). This study provides a proof of concept for an effective strategy to overcome the key limitations of recombinant bispecific T cell engager delivery for solid tumor treatment.
Collapse
Affiliation(s)
- Saru Basnet
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland
| | - Joao M Santos
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland.,TILT Biotherapeutics Ltd, 00290, Helsinki, Finland
| | - Dafne C A Quixabeira
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland
| | - James H A Clubb
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland.,TILT Biotherapeutics Ltd, 00290, Helsinki, Finland
| | - Susanna A M Grönberg-Vähä-Koskela
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland.,Helsinki University Hospital (HUS), 00029, Helsinki, Finland
| | - Victor Arias
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland
| | - Santeri Pakola
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland.,Helsinki University Hospital (HUS), 00029, Helsinki, Finland
| | - Tatiana V Kudling
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland
| | - Camilla Heiniö
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland
| | - Riikka Havunen
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland.,TILT Biotherapeutics Ltd, 00290, Helsinki, Finland
| | - Victor Cervera-Carrascon
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland.,TILT Biotherapeutics Ltd, 00290, Helsinki, Finland
| | - Suvi Sorsa
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland.,TILT Biotherapeutics Ltd, 00290, Helsinki, Finland
| | - Marjukka Anttila
- Department of Pathology, Finnish Food Authority, 00790, Helsinki, Finland
| | - Anna Kanerva
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland.,Department of Gynecology and Obstetrics, Helsinki University Hospital, 00290, Helsinki, Finland
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, 00014, Helsinki, Finland.,TILT Biotherapeutics Ltd, 00290, Helsinki, Finland.,Department of Oncology, Comprehensive Cancer Center, Helsinki University Hospital, and University of Helsinki, 00029, Helsinki, Finland
| |
Collapse
|
15
|
Jia Y, Wang Y, Dunmall LSC, Lemoine NR, Wang P, Wang Y. Syrian hamster as an ideal animal model for evaluation of cancer immunotherapy. Front Immunol 2023; 14:1126969. [PMID: 36923404 PMCID: PMC10008950 DOI: 10.3389/fimmu.2023.1126969] [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: 12/18/2022] [Accepted: 02/09/2023] [Indexed: 03/02/2023] Open
Abstract
Cancer immunotherapy (CIT) has emerged as an exciting new pillar of cancer treatment. Although benefits have been achieved in individual patients, the overall response rate is still not satisfactory. To address this, an ideal preclinical animal model for evaluating CIT is urgently needed. Syrian hamsters present similar features to humans with regard to their anatomy, physiology, and pathology. Notably, the histological features and pathological progression of tumors and the complexity of the tumor microenvironment are equivalent to the human scenario. This article reviews the current tumor models in Syrian hamster and the latest progress in their application to development of tumor treatments including immune checkpoint inhibitors, cytokines, adoptive cell therapy, cancer vaccines, and oncolytic viruses. This progress strongly advocates Syrian hamster as an ideal animal model for development and assessment of CIT for human cancer treatments. Additionally, the challenges of the Syrian hamster as an animal model for CIT are also discussed.
Collapse
Affiliation(s)
- Yangyang Jia
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yanru Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Louisa S Chard Dunmall
- Centre for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Nicholas R. Lemoine
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- Centre for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Pengju Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yaohe Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- Centre for Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
16
|
Tripodi L, Sasso E, Feola S, Coluccino L, Vitale M, Leoni G, Szomolay B, Pastore L, Cerullo V. Systems Biology Approaches for the Improvement of Oncolytic Virus-Based Immunotherapies. Cancers (Basel) 2023; 15:1297. [PMID: 36831638 PMCID: PMC9954314 DOI: 10.3390/cancers15041297] [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] [Received: 01/04/2023] [Revised: 02/06/2023] [Accepted: 02/10/2023] [Indexed: 02/22/2023] Open
Abstract
Oncolytic virus (OV)-based immunotherapy is mainly dependent on establishing an efficient cell-mediated antitumor immunity. OV-mediated antitumor immunity elicits a renewed antitumor reactivity, stimulating a T-cell response against tumor-associated antigens (TAAs) and recruiting natural killer cells within the tumor microenvironment (TME). Despite the fact that OVs are unspecific cancer vaccine platforms, to further enhance antitumor immunity, it is crucial to identify the potentially immunogenic T-cell restricted TAAs, the main key orchestrators in evoking a specific and durable cytotoxic T-cell response. Today, innovative approaches derived from systems biology are exploited to improve target discovery in several types of cancer and to identify the MHC-I and II restricted peptide repertoire recognized by T-cells. Using specific computation pipelines, it is possible to select the best tumor peptide candidates that can be efficiently vectorized and delivered by numerous OV-based platforms, in order to reinforce anticancer immune responses. Beyond the identification of TAAs, system biology can also support the engineering of OVs with improved oncotropism to reduce toxicity and maintain a sufficient portion of the wild-type virus virulence. Finally, these technologies can also pave the way towards a more rational design of armed OVs where a transgene of interest can be delivered to TME to develop an intratumoral gene therapy to enhance specific immune stimuli.
Collapse
Affiliation(s)
- Lorella Tripodi
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80138 Naples, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80131 Naples, Italy
| | - Emanuele Sasso
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80138 Naples, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80131 Naples, Italy
| | - Sara Feola
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, 00100 Helsinki, Finland
- Translational Immunology Research Program (TRIMM), University of Helsinki, 00100 Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, 00100 Helsinki, Finland
| | - Ludovica Coluccino
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80138 Naples, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80131 Naples, Italy
| | - Maria Vitale
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80131 Naples, Italy
| | - Guido Leoni
- Nouscom Srl, via Castel Romano 100, 00128 Rome, Italy
| | - Barbara Szomolay
- Systems Immunity Research Institute, Cardiff University School of Medicine, Cardiff CF14 4YS, UK
| | - Lucio Pastore
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80138 Naples, Italy
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80131 Naples, Italy
| | - Vincenzo Cerullo
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, 00100 Helsinki, Finland
- Translational Immunology Research Program (TRIMM), University of Helsinki, 00100 Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, 00100 Helsinki, Finland
| |
Collapse
|
17
|
Mansouri S, Daniel L, Amhis N, Leveille M, Boudreau JE, Alkayyal AA, Collin Y, Tai LH. Perioperative oncolytic virotherapy to counteract surgery-induced immunosuppression and improve outcomes in pancreatic ductal adenocarcinoma. Front Oncol 2023; 13:1071751. [PMID: 36874130 PMCID: PMC9978493 DOI: 10.3389/fonc.2023.1071751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a high fatality cancer with one of the worst prognoses in solid tumors. Most patients present with late stage, metastatic disease and are not eligible for potentially curative surgery. Despite complete resection, the majority of surgical patients will recur within the first two years following surgery. Postoperative immunosuppression has been described in different digestive cancers. While the underlying mechanism is not fully understood, there is compelling evidence to link surgery with disease progression and cancer metastasis in the postoperative period. However, the idea of surgery-induced immunosuppression as a facilitator of recurrence and metastatic spread has not been explored in the context of pancreatic cancer. By surveying the existing literature on surgical stress in mostly digestive cancers, we propose a novel practice-changing paradigm: alleviate surgery-induced immunosuppression and improve oncological outcome in PDAC surgical patients by administering oncolytic virotherapy in the perioperative period.
Collapse
Affiliation(s)
- Sarah Mansouri
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, QC, Canada.,Department of Surgery, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Lauren Daniel
- Department of Surgery, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Nawal Amhis
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, QC, Canada.,Department of Surgery, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Maxime Leveille
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Jeanette E Boudreau
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Almohanad A Alkayyal
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, Saudi Arabia.,Immunology Research Program, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Yves Collin
- Department of Surgery, Université de Sherbrooke, Sherbrooke, QC, Canada.,Research Center of the Centre hospitalier universitaire de Sherbrooke (CHUS), Sherbrooke, QC, Canada
| | - Lee-Hwa Tai
- Department of Immunology and Cell Biology, Université de Sherbrooke, Sherbrooke, QC, Canada.,Research Center of the Centre hospitalier universitaire de Sherbrooke (CHUS), Sherbrooke, QC, Canada
| |
Collapse
|
18
|
Clubb JHA, Kudling TV, Girych M, Haybout L, Pakola S, Hamdan F, Cervera-Carrascon V, Hemmes A, Grönberg-Vähä-Koskela S, Santos JM, Quixabeira DCA, Basnet S, Heiniö C, Arias V, Jirovec E, Kaptan S, Havunen R, Sorsa S, Erikat A, Schwartz J, Anttila M, Aro K, Viitala T, Vattulainen I, Cerullo V, Kanerva A, Hemminki A. Development of a Syrian hamster anti-PD-L1 monoclonal antibody enables oncolytic adenoviral immunotherapy modelling in an immunocompetent virus replication permissive setting. Front Immunol 2023; 14:1060540. [PMID: 36817448 PMCID: PMC9936529 DOI: 10.3389/fimmu.2023.1060540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 01/11/2023] [Indexed: 02/05/2023] Open
Abstract
Introduction Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of cancer, but preclinical testing of hypotheses such as combination therapies has been complicated, in part due to species incompatibility issues. For example, one of few known permissive animal models for oncolytic adenoviruses is the Syrian hamster, for which an ICI, mainly an anti-PD-L1 monoclonal antibody (mAb) was not previously available. In this study, we developed an anti-Syrian hamster PD-L1 mAb to enable the evaluation of safety and efficacy, when combining anti-PD-L1 with an oncolytic adenovirus encoding tumour necrosis factor alpha (TNFα) and interleukin-2 (IL-2) (Ad5/3-E2F-D24-hTNFα-IRES-hIL-2 or TILT-123). Methods Recombinant Syrian hamster PD-L1 was expressed and mice immunized for mAb formation using hybridoma technology. Clonal selection through binding and functional studies in vitro, in silico and in vivo identified anti-PD-L1 clone 11B12-1 as the primary mAb candidate for immunotherapy modelling. The oncolytic virus (OV) and ICI combination approach was then evaluated using 11B12-1 and TILT-123 in a Syrian hamster model of pancreatic ductal adenocarcinoma (PDAC). Results Supernatants from hybridoma parent subclone 11B12B4 provided the highest positive PD-L1 signal, on Syrian hamster PBMCs and three cancer cell lines (HT100, HapT1 and HCPC1). In vitro co-cultures revealed superior immune modulated profiles of cell line matched HT100 tumour infiltrating lymphocytes when using subclones of 7G2, 11B12 and 12F1. Epitope binning and epitope prediction using AlphaFold2 and ColabFold revealed two distinct functional epitopes for clone 11B12-1 and 12F1-1. Treatment of Syrian hamsters bearing HapT1 tumours, with 11B12-1 induced significantly better (p<0.05) tumour growth control than isotype control by day 12. 12F1-1 did not induce significant tumour growth control. The combination of 11B12-1 with oncolytic adenovirus TILT-123 improved tumour growth control further, when compared to monotherapy (p<0.05) by day 26. Conclusions Novel Syrian hamster anti-PD-L1 clone 11B12-1 induces tumour growth control in a hamster model of PDAC. Combining 11B12-1 with oncolytic adenovirus TILT-123 improves tumour growth control further and demonstrates good safety and toxicity profiles.
Collapse
Affiliation(s)
- James H A Clubb
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,R&D Department, TILT Biotherapeutics Ltd, Helsinki, Finland.,Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Tatiana V Kudling
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Mykhailo Girych
- Department of Physics, University of Helsinki, Helsinki, Finland
| | - Lyna Haybout
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Santeri Pakola
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Firas Hamdan
- Laboratory of ImmunoViroTherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Víctor Cervera-Carrascon
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,R&D Department, TILT Biotherapeutics Ltd, Helsinki, Finland.,Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Annabrita Hemmes
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Sciences (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Susanna Grönberg-Vähä-Koskela
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Research Program Unit (RPU), University of Helsinki, Helsinki, Finland.,Comprehensive Cancer Centre, Helsinki University Hospital, Helsinki, Finland
| | - João Manuel Santos
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,R&D Department, TILT Biotherapeutics Ltd, Helsinki, Finland.,Research Program Unit (RPU), 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.,R&D Department, TILT Biotherapeutics Ltd, Helsinki, Finland.,Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Saru Basnet
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Camilla Heiniö
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Victor Arias
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Elise Jirovec
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Shreyas Kaptan
- Department of Physics, University of Helsinki, Helsinki, Finland
| | - Riikka Havunen
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,R&D Department, TILT Biotherapeutics Ltd, Helsinki, Finland.,Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Suvi Sorsa
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,R&D Department, TILT Biotherapeutics Ltd, Helsinki, Finland.,Research Program Unit (RPU), University of Helsinki, Helsinki, Finland
| | - Abdullah Erikat
- Department of Chemistry and the Randall Centre for Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Joel Schwartz
- Chicago Department of Oral Medicine and Diagnostic Science, University of Illinois, Chicago, IL, United States
| | | | - Katri Aro
- Comprehensive Cancer Centre, Helsinki University Hospital, Helsinki, Finland.,Department of Otorhinolaryngology - Head and Neck Surgery, Helsinki Head and Neck Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Tapani Viitala
- Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Ilpo Vattulainen
- Department of Physics, University of Helsinki, Helsinki, Finland
| | - Vincenzo Cerullo
- Laboratory of ImmunoViroTherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,Drug Research Program, Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Anna Kanerva
- Department of Gynecology and Obstetrics, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.,R&D Department, TILT Biotherapeutics Ltd, Helsinki, Finland.,Research Program Unit (RPU), University of Helsinki, Helsinki, Finland.,Comprehensive Cancer Centre, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
19
|
Lu SC, Barry MA. Locked and loaded: engineering and arming oncolytic adenoviruses to enhance anti-tumor immune responses. Expert Opin Biol Ther 2022; 22:1359-1378. [DOI: 10.1080/14712598.2022.2139601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
| | - Michael A Barry
- Division of Infectious Diseases, Department of Medicine
- Department of Immunology
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
20
|
Spunde K, Korotkaja K, Zajakina A. Recombinant Viral Vectors for Therapeutic Programming of Tumour Microenvironment: Advantages and Limitations. Biomedicines 2022; 10:2142. [PMID: 36140243 PMCID: PMC9495732 DOI: 10.3390/biomedicines10092142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 11/30/2022] Open
Abstract
Viral vectors have been widely investigated as tools for cancer immunotherapy. Although many preclinical studies demonstrate significant virus-mediated tumour inhibition in synergy with immune checkpoint molecules and other drugs, the clinical success of viral vector applications in cancer therapy currently is limited. A number of challenges have to be solved to translate promising vectors to clinics. One of the key elements of successful virus-based cancer immunotherapy is the understanding of the tumour immune state and the development of vectors to modify the immunosuppressive tumour microenvironment (TME). Tumour-associated immune cells, as the main component of TME, support tumour progression through multiple pathways inducing resistance to treatment and promoting cancer cell escape mechanisms. In this review, we consider DNA and RNA virus vectors delivering immunomodulatory genes (cytokines, chemokines, co-stimulatory molecules, antibodies, etc.) and discuss how these viruses break an immunosuppressive cell development and switch TME to an immune-responsive "hot" state. We highlight the advantages and limitations of virus vectors for targeted therapeutic programming of tumour immune cell populations and tumour stroma, and propose future steps to establish viral vectors as a standard, efficient, safe, and non-toxic cancer immunotherapy approach that can complement other promising treatment strategies, e.g., checkpoint inhibitors, CAR-T, and advanced chemotherapeutics.
Collapse
Affiliation(s)
| | | | - Anna Zajakina
- Cancer Gene Therapy Group, Latvian Biomedical Research and Study Centre, Ratsupites Str. 1, k.1, LV-1067 Riga, Latvia
| |
Collapse
|
21
|
Nisar M, Paracha RZ, Adil S, Qureshi SN, Janjua HA. An Extensive Review on Preclinical and Clinical Trials of Oncolytic Viruses Therapy for Pancreatic Cancer. Front Oncol 2022; 12:875188. [PMID: 35686109 PMCID: PMC9171400 DOI: 10.3389/fonc.2022.875188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 04/12/2022] [Indexed: 12/12/2022] Open
Abstract
Chemotherapy resistance and peculiar tumor microenvironment, which diminish or mitigate the effects of therapies, make pancreatic cancer one of the deadliest malignancies to manage and treat. Advanced immunotherapies are under consideration intending to ameliorate the overall patient survival rate in pancreatic cancer. Oncolytic viruses therapy is a new type of immunotherapy in which a virus after infecting and lysis the cancer cell induces/activates patients’ immune response by releasing tumor antigen in the blood. The current review covers the pathways and molecular ablation that take place in pancreatic cancer cells. It also unfolds the extensive preclinical and clinical trial studies of oncolytic viruses performed and/or undergoing to design an efficacious therapy against pancreatic cancer.
Collapse
Affiliation(s)
- Maryum Nisar
- School of Interdisciplinary Engineering & Sciences (SINES), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Rehan Zafar Paracha
- School of Interdisciplinary Engineering & Sciences (SINES), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Sidra Adil
- School of Interdisciplinary Engineering & Sciences (SINES), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | | | - Hussnain Ahmed Janjua
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Sector H-12, Islamabad, Pakistan
| |
Collapse
|
22
|
Li Q, Oduro PK, Guo R, Li R, Leng L, Kong X, Wang Q, Yang L. Oncolytic Viruses: Immunotherapy Drugs for Gastrointestinal Malignant Tumors. Front Cell Infect Microbiol 2022; 12:921534. [PMID: 35719333 PMCID: PMC9203847 DOI: 10.3389/fcimb.2022.921534] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
Oncolytic virus therapy has advanced rapidly in recent years. Natural or transgenic viruses can target tumor cells and inhibit tumor growth and metastasis in various ways without interfering with normal cell and tissue function. Oncolytic viruses have a high level of specificity and are relatively safe. Malignant tumors in the digestive system continue to have a high incidence and mortality rate. Although existing treatment methods have achieved some curative effects, they still require further improvement due to side effects and a lack of specificity. Many studies have shown that oncolytic viruses can kill various tumor cells, including malignant tumors in the digestive system. This review discusses how oncolytic virus therapy improves malignant tumors in the digestive system from the point-of-view of basic and clinical studies. Also, the oncolytic virus anti-tumor mechanisms underpinning the therapeutic potential of oncolytic viruses are expounded. In all, we argue that oncolytic viruses might eventually provide therapeutic solutions to malignant tumors in the digestive system.
Collapse
Affiliation(s)
- Qingbo Li
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Patrick Kwabena Oduro
- Research Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine & State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education, Tianjin, China
| | - Rui Guo
- Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ruiqiao Li
- Research Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine & State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education, Tianjin, China
| | - Ling Leng
- Research Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine & State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education, Tianjin, China
| | - Xianbin Kong
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Xianbin Kong, ; Qilong Wang, ; Long Yang,
| | - Qilong Wang
- Research Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine & State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education, Tianjin, China
- *Correspondence: Xianbin Kong, ; Qilong Wang, ; Long Yang,
| | - Long Yang
- Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Xianbin Kong, ; Qilong Wang, ; Long Yang,
| |
Collapse
|
23
|
Controlling Cell Trafficking: Addressing Failures in CAR T and NK Cell Therapy of Solid Tumours. Cancers (Basel) 2022; 14:cancers14040978. [PMID: 35205725 PMCID: PMC8870056 DOI: 10.3390/cancers14040978] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/09/2022] [Accepted: 02/11/2022] [Indexed: 02/04/2023] Open
Abstract
The precision guiding of endogenous or adoptively transferred lymphocytes to the solid tumour mass is obligatory for optimal anti-tumour effects and will improve patient safety. The recognition and elimination of the tumour is best achieved when anti-tumour lymphocytes are proximal to the malignant cells. For example, the regional secretion of soluble factors, cytotoxic granules, and cell-surface molecule interactions are required for the death of tumour cells and the suppression of neovasculature formation, tumour-associated suppressor, or stromal cells. The resistance of individual tumour cell clones to cellular therapy and the hostile environment of the solid tumours is a major challenge to adoptive cell therapy. We review the strategies that could be useful to overcoming insufficient immune cell migration to the tumour cell mass. We argue that existing 'competitive' approaches should now be revisited as complementary approaches to improve CAR T and NK cell therapy.
Collapse
|
24
|
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
|