1
|
Sasaki Y, Maeda T, Hojo M, Miura T, Ishikawa K, Funayama E, Okada K, Yamamoto Y. Synergistic anti-tumor effects of oncolytic virus and anti-programmed cell death protein 1 antibody combination therapy: For suppression of lymph node and distant metastasis in a murine melanoma model. Biochem Biophys Res Commun 2024; 740:151011. [PMID: 39571230 DOI: 10.1016/j.bbrc.2024.151011] [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: 08/20/2024] [Revised: 11/10/2024] [Accepted: 11/14/2024] [Indexed: 12/01/2024]
Abstract
It is believed that oncolytic viruses (OVs) exert both direct anti-tumor effects by intratumoral injection as well as indirect anti-tumor effects by activating systemic immunity. In phase III clinical trials, OV and anti-programmed cell death-1 (aPD-1) antibody combination therapy showed no significant differences in overall survival and progression-free survival in patients with unresectable advanced melanoma. In the study, OVs can exert only indirect anti-tumor effects in non-injected, systemic lesions. If the tumor is at a stage where both direct and indirect anti-tumor effects of OVs can be expected, OVs may further enhance the therapeutic effect, in addition to the clinically expected therapeutic effect. Therefore, we investigated whether canerpaturev (C-REV) and aPD-1 antibody combination therapy suppresses tumor progression in a murine melanoma model. Our findings showed that the C-REV and aPD-1 antibody combination therapy suppressed tumor progression in a murine melanoma model. The combination therapy stimulated systemic immunity in lymphoid tissues by activating helper T cells and B cells to enhance adaptive and humoral immunity, as well as by increasing effector/memory T cell fractions. Synergistically enhanced systemic anti-tumor effects suppressed lymph node and lung metastases. These findings suggest that direct anti-tumor effects by infecting and destroying cancer cells from within and indirect anti-tumor effects enhanced by the combination therapy worked simultaneously to suppress tumor progression. Our results may provide evidence to support the usefulness of OV and aPD-1 antibody combination therapy as a neoadjuvant therapy in the surgical treatment of melanoma.
Collapse
Affiliation(s)
- Yuki Sasaki
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Japan.
| | - Taku Maeda
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Japan.
| | - Masahiro Hojo
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Japan.
| | - Takahiro Miura
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Japan.
| | - Kosuke Ishikawa
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Japan.
| | - Emi Funayama
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Japan.
| | - Kazufumi Okada
- Data Science Center, Promotion Unit, Institute of Health Science Innovation for Medical Care, Hokkaido University Hospital, Japan.
| | - Yuhei Yamamoto
- Department of Plastic and Reconstructive Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Japan.
| |
Collapse
|
2
|
Brito-Orama S, Sheth RA. The Contemporary Landscape and Future Directions of Intratumoral Immunotherapy. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2023; 6:84-90. [PMID: 37214205 PMCID: PMC10195020 DOI: 10.36401/jipo-22-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 11/07/2022] [Accepted: 11/28/2022] [Indexed: 05/24/2023]
Abstract
Systemically administered immunotherapies have revolutionized the care of patients with cancer; however, for many cancer types, most patients do not exhibit objective responses. Intratumoral immunotherapy is a burgeoning strategy that is designed to boost the effectiveness of cancer immunotherapies across the spectrum of malignancies. By locally administering immune-activating therapies into the tumor itself, immunosuppressive barriers in the tumor microenvironment can be broken. Moreover, therapies too potent for systemic delivery can be safely administered to target location to maximize efficacy and minimize toxicity. In order for these therapies to be effective, though, they must be effectively delivered into the target tumor lesion. In this review, we summarize the current landscape of intratumoral immunotherapies and highlight key concepts that influence intratumoral delivery, and by extension, efficacy. We also provide an overview of the breadth and depth of approved minimally invasive delivery devices that can be considered to improve delivery of intratumoral therapies.
Collapse
Affiliation(s)
- Sebastian Brito-Orama
- Department of Interventional Radiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rahul A. Sheth
- Department of Interventional Radiology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
3
|
Tang C, Li L, Mo T, Na J, Qian Z, Fan D, Sun X, Yao M, Pan L, Huang Y, Zhong L. Oncolytic viral vectors in the era of diversified cancer therapy: from preclinical to clinical. Clin Transl Oncol 2022; 24:1682-1701. [PMID: 35612653 PMCID: PMC9131313 DOI: 10.1007/s12094-022-02830-x] [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: 01/31/2022] [Accepted: 03/21/2022] [Indexed: 12/19/2022]
Abstract
With the in-depth research and wide application of immunotherapy recently, new therapies based on oncolytic viruses are expected to create new prospects for cancer treatment via eliminating the suppression of the immune system by tumors. Currently, an increasing number of viruses are developed and engineered, and various virus vectors based on effectively stimulating human immune system to kill tumor cells have been approved for clinical treatment. Although the virus can retard the proliferation of tumor cells, the choice of oncolytic viruses in biological cancer therapy is equally critical given their therapeutic efficacy, safety and adverse effects. Moreover, previously known oncolytic viruses have not been systematically classified. Therefore, in this review, we summarized and distinguished the characteristics of several common types of oncolytic viruses: herpes simplex virus, adenovirus, measles virus, Newcastle disease virus, reovirus and respiratory syncytial virus. Subsequently, we outlined that these oncolytic viral vectors have been transformed from preclinical studies in combination with immunotherapy, radiotherapy, chemotherapy, and nanoparticles into clinical therapeutic strategies for various advanced solid malignancies or circulatory system cancers.
Collapse
Affiliation(s)
- Chao Tang
- 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, 530021, Guangxi, China
| | - Lan Li
- 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, 530021, Guangxi, China
| | - Tong Mo
- 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, 530021, Guangxi, China
| | - Jintong Na
- 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, 530021, Guangxi, China
| | - Zhangbo Qian
- 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, 530021, Guangxi, China
| | - Dianfa Fan
- 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, 530021, Guangxi, China
| | - Xinjun Sun
- 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, 530021, Guangxi, China
| | - Min Yao
- 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, 530021, Guangxi, China
| | - Lina Pan
- 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, 530021, Guangxi, China
| | - Yong Huang
- 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, 530021, Guangxi, 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, 530021, Guangxi, China.
| |
Collapse
|
4
|
Scanlan H, Coffman Z, Bettencourt J, Shipley T, Bramblett DE. Herpes simplex virus 1 as an oncolytic viral therapy for refractory cancers. Front Oncol 2022; 12:940019. [PMID: 35965554 PMCID: PMC9364694 DOI: 10.3389/fonc.2022.940019] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/27/2022] [Indexed: 11/25/2022] Open
Abstract
The need for efficacious and non-toxic cancer therapies is paramount. Oncolytic viruses (OVs) are showing great promise and are introducing new possibilities in cancer treatment with their ability to selectively infect tumor cells and trigger antitumor immune responses. Herpes Simplex Virus 1 (HSV-1) is a commonly selected OV candidate due to its large genome, relative safety profile, and ability to infect a variety of cell types. Talimogene laherparevec (T-VEC) is an HSV-1-derived OV variant and the first and only OV therapy currently approved for clinical use by the United States Food and Drug Administration (FDA). This review provides a concise description of HSV-1 as an OV candidate and the genomic organization of T-VEC. Furthermore, this review focuses on the advantages and limitations in the use of T-VEC compared to other HSV-1 OV variants currently in clinical trials. In addition, approaches for future directions of HSV-1 OVs as cancer therapy is discussed.
Collapse
Affiliation(s)
- Hayle Scanlan
- Rowan School of Medicine, RowanSOM-Jefferson Health-Virtua Our Lady of Lourdes Hospital, Stratford, NJ, United States
| | - Zachary Coffman
- Monroe Clinic Rural Family Medicine Program, The University of Illinois College of Medicine Rockford, Monroe, WI, United States
| | - Jeffrey Bettencourt
- Department of Biomedical Sciences, Burrell College of Osteopathic Medicine, Las Cruces, NM, United States
| | - Timothy Shipley
- Department of Biomedical Sciences, A.T. Still University School of Osteopathic Medicine in Arizona, Mesa, AZ, United States
| | - Debra E. Bramblett
- Department of Biomedical Sciences, Burrell College of Osteopathic Medicine, Las Cruces, NM, United States
- *Correspondence: Debra E. Bramblett,
| |
Collapse
|
5
|
The Effect of Herpes Simplex Virus-Type-1 (HSV-1) Oncolytic Immunotherapy on the Tumor Microenvironment. Viruses 2021; 13:v13071200. [PMID: 34206677 PMCID: PMC8310320 DOI: 10.3390/v13071200] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/11/2021] [Accepted: 06/16/2021] [Indexed: 12/19/2022] Open
Abstract
The development of cancer causes disruption of anti-tumor immunity required for surveillance and elimination of tumor cells. Immunotherapeutic strategies aim for the restoration or establishment of these anti-tumor immune responses. Cancer immunotherapies include immune checkpoint inhibitors (ICIs), adoptive cellular therapy (ACT), cancer vaccines, and oncolytic virotherapy (OVT). The clinical success of some of these immunotherapeutic modalities, including herpes simplex virus type-1 derived OVT, resulted in Food and Drug Administration (FDA) approval for use in treatment of human cancers. However, a significant proportion of patients do not respond or benefit equally from these immunotherapies. The creation of an immunosuppressive tumor microenvironment (TME) represents an important barrier preventing success of many immunotherapeutic approaches. Mechanisms of immunosuppression in the TME are a major area of current research. In this review, we discuss how oncolytic HSV affects the tumor microenvironment to promote anti-tumor immune responses. Where possible we focus on oncolytic HSV strains for which clinical data is available, and discuss how these viruses alter the vasculature, extracellular matrix and immune responses in the tumor microenvironment.
Collapse
|
6
|
Koch MS, Lawler SE, Chiocca EA. HSV-1 Oncolytic Viruses from Bench to Bedside: An Overview of Current Clinical Trials. Cancers (Basel) 2020; 12:E3514. [PMID: 33255871 PMCID: PMC7760226 DOI: 10.3390/cancers12123514] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/18/2020] [Accepted: 11/23/2020] [Indexed: 12/13/2022] Open
Abstract
Herpes simplex virus 1 (HSV-1) provides a genetic chassis for several oncolytic viruses (OVs) currently in clinical trials. Oncolytic HSV1 (oHSV) have been engineered to reduce neurovirulence and enhance anti-tumor lytic activity and immunogenicity to make them attractive candidates in a range of oncology indications. Successful clinical data resulted in the FDA-approval of the oHSV talimogene laherparepvec (T-Vec) in 2015, and several other variants are currently undergoing clinical assessment and may expand the landscape of future oncologic therapy options. This review offers a detailed overview of the latest results from clinical trials as well as an outlook on newly developed HSV-1 oncolytic variants with improved tumor selectivity, replication, and immunostimulatory capacity and related clinical studies.
Collapse
Affiliation(s)
| | - Sean E. Lawler
- Harvey Cushing Neurooncology Research Laboratories, Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (M.S.K.); (E.A.C.)
| | | |
Collapse
|
7
|
Generation of an Oncolytic Herpes Simplex Viral Vector Completely Retargeted to the GDNF Receptor GFRα1 for Specific Infection of Breast Cancer Cells. Int J Mol Sci 2020; 21:ijms21228815. [PMID: 33233403 PMCID: PMC7700293 DOI: 10.3390/ijms21228815] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 11/18/2020] [Indexed: 12/17/2022] Open
Abstract
Oncolytic herpes simplex viruses (oHSV) are under development for the treatment of a variety of human cancers, including breast cancer, a leading cause of cancer mortality among women worldwide. Here we report the design of a fully retargeted oHSV for preferential infection of breast cancer cells through virus recognition of GFRα1, the cellular receptor for glial cell-derived neurotrophic factor (GDNF). GFRα1 displays a limited expression profile in normal adult tissue, but is upregulated in a subset of breast cancers. We generated a recombinant HSV expressing a completely retargeted glycoprotein D (gD), the viral attachment/entry protein, that incorporates pre-pro-GDNF in place of the signal peptide and HVEM binding domain of gD and contains a deletion of amino acid 38 to eliminate nectin-1 binding. We show that GFRα1 is necessary and sufficient for infection by the purified recombinant virus. Moreover, this virus enters and spreads in GFRα1-positive breast cancer cells in vitro and caused tumor regression upon intratumoral injection in vivo. Given the heterogeneity observed between and within individual breast cancers at the molecular level, these results expand our ability to deliver oHSV to specific tumors and suggest opportunities to enhance drug or viral treatments aimed at other receptors.
Collapse
|
8
|
Scherwitzl I, Opp S, Hurtado AM, Pampeno C, Loomis C, Kannan K, Yu M, Meruelo D. Sindbis Virus with Anti-OX40 Overcomes the Immunosuppressive Tumor Microenvironment of Low-Immunogenic Tumors. Mol Ther Oncolytics 2020; 17:431-447. [PMID: 32478167 PMCID: PMC7251545 DOI: 10.1016/j.omto.2020.04.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 04/29/2020] [Indexed: 01/03/2023] Open
Abstract
Despite remarkable responses to cancer immunotherapy in a subset of patients, many patients remain resistant to therapies. It is now clear that elevated levels of tumor-infiltrating T cells as well as a systemic anti-tumor immune response are requirements for successful immunotherapies. However, the tumor microenvironment imposes an additional resistance mechanism to immunotherapy. We have developed a practical and improved strategy for cancer immunotherapy using an oncolytic virus and anti-OX40. This strategy takes advantage of a preexisting T cell immune repertoire in vivo, removing the need to know about present tumor antigens. We have shown in this study that the replication-deficient oncolytic Sindbis virus vector expressing interleukin-12 (IL-12) (SV.IL12) activates immune-mediated tumor killing by inducing OX40 expression on CD4 T cells, allowing the full potential of the agonistic anti-OX40 antibody. The combination of SV.IL12 with anti-OX40 markedly changes the transcriptome signature and metabolic program of T cells, driving the development of highly activated terminally differentiated effector T cells. These metabolically reprogrammed T cells demonstrate enhanced tumor infiltration capacity as well as anti-tumor activity capable of overcoming the repressive tumor microenvironment. Our findings identify SV.IL12 in combination with anti-OX40 to be a novel and potent therapeutic strategy that can cure multiple types of low-immunogenic solid tumors.
Collapse
Affiliation(s)
- Iris Scherwitzl
- Department of Pathology, NYU School of Medicine, New York, NY, USA
| | - Silvana Opp
- Department of Pathology, NYU School of Medicine, New York, NY, USA
| | | | | | - Cynthia Loomis
- Department of Pathology, NYU School of Medicine, New York, NY, USA
| | - Kasthuri Kannan
- Department of Pathology, NYU School of Medicine, New York, NY, USA
| | - Minjun Yu
- Department of Pathology, NYU School of Medicine, New York, NY, USA
| | - Daniel Meruelo
- Department of Pathology, NYU School of Medicine, New York, NY, USA
| |
Collapse
|
9
|
Abstract
Intratumoral immunotherapies aim to trigger local and systemic immunologic responses via direct injection of immunostimulatory agents with the goal of tumor cell lysis, followed by release of tumor‐derived antigens and subsequent activation of tumor‐specific effector T cells. In 2019, a multitude of intratumoral immunotherapies with varied mechanisms of action, including nononcolytic viral therapies such as PV‐10 and toll‐like receptor 9 agonists and oncolytic viral therapies such as CAVATAK, Pexa‐Vec, and HF10, have been extensively evaluated in clinical trials and demonstrated promising antitumor activity with tolerable toxicities in melanoma and other solid tumor types. Talimogene laherparepvec (T‐VEC), a genetically modified herpes simplex virus type 1–based oncolytic immunotherapy, is the first oncolytic virus approved by the U.S. Food and Drug Administration for the treatment of unresectable melanoma recurrent after initial surgery. In patients with unresectable metastatic melanoma, T‐VEC demonstrated a superior durable response rate (continuous complete response or partial response lasting ≥6 months) over subcutaneous GM‐CSF (16.3% vs. 2.1%; p < .001). Responses were seen in both injected and uninjected lesions including visceral lesions, suggesting a systemic antitumor response. When combined with immune checkpoint inhibitors, T‐VEC significantly improved response rates compared with single agent; similar results were seen with combinations of checkpoint inhibitors and other intratumoral therapies such as CAVATAK, HF10, and TLR9 agonists. In this review, we highlight recent results from clinical trials of key intratumoral immunotherapies that are being evaluated in the clinic, with a focus on T‐VEC in the treatment of advanced melanoma as a model for future solid tumor indications. Implications for Practice This review provides oncologists with the latest information on the development of key intratumoral immunotherapies, particularly oncolytic viruses. Currently, T‐VEC is the only U.S. Food and Drug Administration (FDA)‐approved oncolytic immunotherapy. This article highlights the efficacy and safety data from clinical trials of T‐VEC both as monotherapy and in combination with immune checkpoint inhibitors. This review summarizes current knowledge on intratumoral therapies, a novel modality with increased utility in cancer treatment, and T‐VEC, the only U.S. FDA‐approved oncolytic viral therapy, for medical oncologists. This review evaluates approaches to incorporate T‐VEC into daily practice to offer the possibility of response in selected melanoma patients with manageable adverse events as compared with other available immunotherapies. This review highlights recent results from clinical trials of key intratumoral immunotherapies that are being evaluated in the clinic, with a focus on talimogene laherparepvec in the treatment of advanced melanoma as a model for future solid tumor indications.
Collapse
Affiliation(s)
- Omid Hamid
- The Angeles Clinic and Research InstituteLos AngelesCaliforniaUSA
| | | | - Igor Puzanov
- Roswell Park Comprehensive Cancer CenterBuffaloNew YorkUSA
| |
Collapse
|
10
|
Taguchi S, Fukuhara H, Todo T. Oncolytic virus therapy in Japan: progress in clinical trials and future perspectives. Jpn J Clin Oncol 2019; 49:201-209. [PMID: 30462296 DOI: 10.1093/jjco/hyy170] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 09/21/2018] [Indexed: 01/28/2023] Open
Abstract
Oncolytic virus therapy is a promising new option for cancer. It utilizes genetically engineered or naturally occurring viruses that selectively replicate in and kill cancer cells without harming normal cells. T-VEC (talimogene laherparepvec), a second-generation oncolytic herpes simplex virus type 1, was approved by the US Food and Drug Administration for the treatment of inoperable melanoma in 2015 and subsequently approved in Europe in 2016. Other oncolytic viruses using different parental viruses have also been tested in Phase III clinical trials and are ready for drug approval: Pexa-Vec (pexastimogene devacirepvec), an oncolytic vaccinia virus, CG0070, an oncolytic adenovirus, and REOLYSIN (pelareorep), an oncolytic reovirus. In Japan, as of May 2018, several oncolytic viruses have been developed, and some have already proceeded to clinical trials. In this review, we summarize clinical trials assessing oncolytic virus therapy that were conducted or are currently ongoing in Japan, specifically, T-VEC, the abovementioned oncolytic herpes simplex virus type 1, G47Δ, a third-generation oncolytic herpes simplex virus type 1, HF10, a naturally attenuated oncolytic herpes simplex virus type 1, Telomelysin, an oncolytic adenovirus, Surv.m-CRA, another oncolytic adenovirus, and Sendai virus particle. In the near future, oncolytic virus therapy may become an important and major treatment option for cancer in Japan.
Collapse
Affiliation(s)
- Satoru Taguchi
- Department of Urology, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Hiroshi Fukuhara
- Department of Urology, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Tomoki Todo
- Division of Innovative Cancer Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
11
|
The Current Status and Future Prospects of Oncolytic Viruses in Clinical Trials against Melanoma, Glioma, Pancreatic, and Breast Cancers. Cancers (Basel) 2018; 10:cancers10100356. [PMID: 30261620 PMCID: PMC6210336 DOI: 10.3390/cancers10100356] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 09/10/2018] [Accepted: 09/11/2018] [Indexed: 02/06/2023] Open
Abstract
Oncolytic viral therapy has been accepted as a standard immunotherapy since talimogene laherparepvec (T-VEC, Imlygic®) was approved by the Food and Drug Administration (FDA) and European Medicines Agency (EMA) for melanoma treatment in 2015. Various oncolytic viruses (OVs), such as HF10 (Canerpaturev—C-REV) and CVA21 (CAVATAK), are now actively being developed in phase II as monotherapies, or in combination with immune checkpoint inhibitors against melanoma. Moreover, in glioma, several OVs have clearly demonstrated both safety and a promising efficacy in the phase I clinical trials. Additionally, the safety of several OVs, such as pelareorep (Reolysin®), proved their safety and efficacy in combination with paclitaxel in breast cancer patients, but the outcomes of OVs as monotherapy against breast cancer have not provided a clear therapeutic strategy for OVs. The clinical trials of OVs against pancreatic cancer have not yet demonstrated efficacy as either monotherapy or as part of combination therapy. However, there are several oncolytic viruses that have successfully proved their efficacy in different preclinical models. In this review, we mainly focused on the oncolytic viruses that transitioned into clinical trials against melanoma, glioma, pancreatic, and breast cancers. Hence, we described the current status and future prospects of OVs clinical trials against melanoma, glioma, pancreatic, and breast cancers.
Collapse
|
12
|
Hirooka Y, Kasuya H, Ishikawa T, Kawashima H, Ohno E, Villalobos IB, Naoe Y, Ichinose T, Koyama N, Tanaka M, Kodera Y, Goto H. A Phase I clinical trial of EUS-guided intratumoral injection of the oncolytic virus, HF10 for unresectable locally advanced pancreatic cancer. BMC Cancer 2018; 18:596. [PMID: 29801474 PMCID: PMC5970460 DOI: 10.1186/s12885-018-4453-z] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 04/30/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Prognosis of pancreatic cancer is poor with a 5-year survival rate of only 7%. Although several new chemotherapy treatments have shown promising results, all patients will eventually progress, and we need to develop newer chemotherapy treatments to improve response rates and overall survival (OS). HF10 is a spontaneously mutated oncolytic virus derived from a herpes simplex virus-1, and it has potential to show strong antitumor effect against malignancies without damaging normal tissue. We aimed to evaluate the safety and anti-tumor effectiveness in phase I dose-escalation trial of direct injection of HF10 into unresectable locally advanced pancreatic cancer under endoscopic ultrasound (EUS)-guidance in combination with erlotinib and gemcitabine administration. The mid-term results have been previously reported and here we report the final results of our study. METHODS This was a single arm, open-label Phase I trial. HF10 was injected once every 2 weeks and continued up to four times in total unless dose-limiting toxicity (DLT) appears. A total of nine subjects in three Cohorts with dose-escalation were planned to be enrolled in this trial. The primary endpoint was the safety assessment and the secondary endpoint was the efficacy assessment. RESULTS Twelve patients enrolled in this clinical trial, and ten subjects received this therapy. Five patients showed Grade III myelosuppression and two patients developed serious adverse events (AEs) (perforation of duodenum, hepatic dysfunction). However, all of these events were judged as AEs unrelated to HF10. Tumor responses were three partial responses (PR), four stable diseases (SD), and two progressive diseases (PD) out of nine subjects who completed the treatment. Target lesion responses were three PRs and six SDs. The median progression free survival (PFS) was 6.3 months, whereas the median OS was 15.5 months. Two subjects from Cohort 1 and 2 showed downstaging and finally achieved surgical complete response (CR). CONCLUSIONS HF10 direct injection under EUS-guidance in combination with erlotinib and gemcitabine was a safe treatment for locally advanced pancreatic cancer. Combination therapy of HF10 and chemotherapy should be explored further in large prospective studies. TRIAL REGISTRATION This study was prospectively registered in UMIN-CTR (UMIN000010150) on March 4th, 2013.
Collapse
Affiliation(s)
- Yoshiki Hirooka
- Department of Endoscopy, Nagoya University Hospital, 65 Tsuruma-cho, Showa-ku, Nagoya, 466-8550 Japan
| | - Hideki Kasuya
- Cancer Immune Therapy Research Center, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takuya Ishikawa
- Department of Gastroenterology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroki Kawashima
- Department of Gastroenterology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Eizaburo Ohno
- Department of Gastroenterology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Itzel B. Villalobos
- Cancer Immune Therapy Research Center, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshinori Naoe
- Cancer Immune Therapy Research Center, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toru Ichinose
- Cancer Immune Therapy Research Center, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | - Yasuhiro Kodera
- Department of Surgery II, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hidemi Goto
- Department of Gastroenterology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
13
|
Abstract
Oncolytic virotherapy is a kind of antitumor therapy using viruses with natural or engineered tumor-selective replication to intentionally infect and kill tumor cells. An early clinical trial has been performed in the 1950s using wild-type and non-engineered in vitro-passaged virus strains and vaccine strains (first generation oncolytic viruses). Because of the advances in biotechnology and virology, the field of virotherapy has rapidly evolved over the past two decades and innovative recombinant selectivity-enhanced viruses (second generation oncolytic viruses). Nowadays, therapeutic transgene-delivering "armed" oncolytic viruses (third generation oncolytic viruses) have been engineered using many kinds of viruses. In this chapter, the history, mechanisms, rationality, and advantages of oncolytic virotherapy by herpes simplex virus (HSV) are mentioned. Past and ongoing clinical trials by oncolytic HSVs (G207, HSV1716, NV1020, HF10, Talimogene laherparepvec (T-VEC, OncoVEXGM-CSF)) are also summarized. Finally, the way of enhancement of oncolytic virotherapy by gene modification or combination therapy with radiation, chemotherapy, or immune checkpoint inhibitors are discussed.
Collapse
|
14
|
Eissa IR, Naoe Y, Bustos-Villalobos I, Ichinose T, Tanaka M, Zhiwen W, Mukoyama N, Morimoto T, Miyajima N, Hitoki H, Sumigama S, Aleksic B, Kodera Y, Kasuya H. Genomic Signature of the Natural Oncolytic Herpes Simplex Virus HF10 and Its Therapeutic Role in Preclinical and Clinical Trials. Front Oncol 2017; 7:149. [PMID: 28770166 PMCID: PMC5509757 DOI: 10.3389/fonc.2017.00149] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 06/26/2017] [Indexed: 12/19/2022] Open
Abstract
Oncolytic viruses (OVs) are opening new possibilities in cancer therapy with their unique mechanism of selective replication within tumor cells and triggering of antitumor immune responses. HF10 is an oncolytic herpes simplex virus-1 with a unique genomic structure that has non-engineered deletions and insertions accompanied by frame-shift mutations, in contrast to the majority of engineered OVs. At the genetic level, HF10 naturally lacks the expression of UL43, UL49.5, UL55, UL56, and latency-associated transcripts, and overexpresses UL53 and UL54. In preclinical studies, HF10 replicated efficiently within tumor cells with extensive cytolytic effects and induced increased numbers of activated CD4+ and CD8+ T cells and natural killer cells within the tumor, leading to a significant reduction in tumor growth and prolonged survival rates. Investigator-initiated clinical studies of HF10 have been completed in recurrent breast carcinoma, head and neck cancer, and unresectable pancreatic cancer in Japan. Phase I trials were subsequently completed in refractory superficial cancers and melanoma in the United States. HF10 has been demonstrated to have a high safety margin with low frequency of adverse effects in all treated patients. Interestingly, HF10 antigens were detected in pancreatic carcinoma over 300 days after treatment with infiltration of CD4+ and CD8+ T cells, which enhanced the immune response. To date, preliminary results from a Phase II trial have indicated that HF10 in combination with ipilimumab (anti-CTLA-4) is safe and well tolerated, with high antitumor efficacy. Improvement of the effect of ipilimumab was observed in patients with stage IIIb, IIIc, or IV unresectable or metastatic melanoma. This review provides a concise description of the genomic functional organization of HF10 compared with talimogene laherparepvec. Furthermore, this review focuses on HF10 in cancer treatment as monotherapy as well as in combination therapy through a concise description of all preclinical and clinical data. In addition, we will address approaches for future directions in HF10 studies as cancer therapy.
Collapse
Affiliation(s)
- Ibrahim Ragab Eissa
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya, Japan.,Department of Surgery II, Graduate School of Medicine, Nagoya University, Nagoya, Japan.,Faculty of Science, Tanta University, Tanta, Egypt
| | - Yoshinori Naoe
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Itzel Bustos-Villalobos
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Toru Ichinose
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | | | - Wu Zhiwen
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya, Japan.,Department of Surgery II, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Nobuaki Mukoyama
- Department of Otolaryngology, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Taishi Morimoto
- Department of Surgery II, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Noriyuki Miyajima
- Department of Transplantation and Endocrine Surgery, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Hasegawa Hitoki
- Office of International Affairs, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Seiji Sumigama
- Office of International Affairs, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Branko Aleksic
- Office of International Affairs, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Yasuhiro Kodera
- Department of Surgery II, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Hideki Kasuya
- Cancer Immune Therapy Research Center, Graduate School of Medicine, Nagoya University, Nagoya, Japan
| |
Collapse
|
15
|
Asad AS, Moreno Ayala MA, Gottardo MF, Zuccato C, Nicola Candia AJ, Zanetti FA, Seilicovich A, Candolfi M. Viral gene therapy for breast cancer: progress and challenges. Expert Opin Biol Ther 2017; 17:945-959. [DOI: 10.1080/14712598.2017.1338684] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Antonela S. Asad
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED-CONICET/UBA), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Mariela A. Moreno Ayala
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED-CONICET/UBA), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - M. Florencia Gottardo
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED-CONICET/UBA), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Camila Zuccato
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED-CONICET/UBA), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Alejandro Javier Nicola Candia
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED-CONICET/UBA), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Flavia A. Zanetti
- Instituto de Ciencia y Tecnología César Milstein (ICT Milstein), Unidad Ejecutora del Consejo Nacional de Investigaciones Científicas y Técnicas, Fundación Pablo Cassará, Buenos Aires, Argentina
| | - Adriana Seilicovich
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED-CONICET/UBA), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marianela Candolfi
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- Instituto de Investigaciones Biomédicas (INBIOMED-CONICET/UBA), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
16
|
Presage of oncolytic virotherapy for oral cancer with herpes simplex virus. JAPANESE DENTAL SCIENCE REVIEW 2016; 53:53-60. [PMID: 28479936 PMCID: PMC5405200 DOI: 10.1016/j.jdsr.2016.10.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 10/02/2016] [Accepted: 10/08/2016] [Indexed: 12/30/2022] Open
Abstract
A virus is a pathogenic organism that causes a number of infectious diseases in humans. The oral cavity is the site at which viruses enter and are excreted from the human body. Herpes simplex virus type 1 (HSV-1) produces the primary infectious disease, gingivostomatitis, and recurrent disease, labial herpes. HSV-1 is one of the most extensively investigated viruses used for cancer therapy. In principle, HSV-1 infects epithelial cells and neuronal cells and exhibits cytotoxicity due to its cytopathic effects on these cells. If the replication of the virus occurs in tumor cells, but not normal cells, the virus may be used as an antitumor agent. Therefore, HSV-1 genes have been modified by genetic engineering, and in vitro and in vivo studies with the oncolytic virus have demonstrated its efficiency against head and neck cancer including oral cancer. The oncolytic abilities of other viruses such as adenovirus and reovirus have also been demonstrated. In clinical trials, HSV-1 is the top runner and is now available for the treatment of patients with advanced melanoma. Thus, melanoma in the oral cavity is the target of oncolytic HSV-1. Oncolytic virotherapy is a hopeful and realistic modality for the treatment of oral cancer.
Collapse
|
17
|
Zamarin D, Pesonen S. Replication-Competent Viruses as Cancer Immunotherapeutics: Emerging Clinical Data. Hum Gene Ther 2016; 26:538-49. [PMID: 26176173 PMCID: PMC4968310 DOI: 10.1089/hum.2015.055] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Replication-competent (oncolytic) viruses (OV) as cancer immunotherapeutics have gained an increasing level of attention over the last few years while the clinical evidence of virus-mediated antitumor immune responses is still anecdotal. Multiple clinical studies are currently ongoing and more immunomonitoring results are expected within the next five years. All viruses can be recognized by the immune system and are therefore potential candidates for immune therapeutics. However, each virus activates innate immune system by using different combination of recognition receptors/pathways which leads to qualitatively different adaptive immune responses. This review summarizes immunological findings in cancer patients following treatment with replication-competent viruses.
Collapse
Affiliation(s)
- Dmitriy Zamarin
- 1 Memorial Sloan Kettering Cancer Center , New York, New York
| | | |
Collapse
|
18
|
Ishihara M, Seo N, Mitsui J, Muraoka D, Tanaka M, Mineno J, Ikeda H, Shiku H. Systemic CD8+ T cell-mediated tumoricidal effects by intratumoral treatment of oncolytic herpes simplex virus with the agonistic monoclonal antibody for murine glucocorticoid-induced tumor necrosis factor receptor. PLoS One 2014; 9:e104669. [PMID: 25105508 PMCID: PMC4126744 DOI: 10.1371/journal.pone.0104669] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Accepted: 07/11/2014] [Indexed: 02/07/2023] Open
Abstract
Oncolytic virotherapy combined with immunomodulators is a novel noninvasive strategy for cancer treatment. In this study, we examined the tumoricidal effects of oncolytic HF10, a naturally occurring mutant of herpes simplex virus type-1, combined with an agonistic DTA-1 monoclonal antibody specific for the glucocorticoid-induced tumor necrosis factor receptor. Two murine tumor models were used to evaluate the therapeutic efficacies of HF10 virotherapy combined with DTA-1. The kinetics and immunological mechanisms of DTA-1 in HF10 infection were examined using flow cytometry and immunohistochemistry. Intratumoral administration of HF10 in combination with DTA-1 at a low dose resulted in a more vigorous attenuation of growth of the untreated contralateral as well as the treated tumors than treatment with either HF10 or DTA-1 alone. An accumulation of CD8+ T cells, including tumor- and herpes simplex virus type-1-specific populations, and a decrease in the number of CD4+ Foxp3+ T regulatory cells were seen in both HF10- and DTA-1-treated tumors. Studies using Fc-digested DTA-1 and Fcγ receptor knockout mice demonstrated the direct participation of DTA-1 in regulatory T cell depletion by antibody-dependent cellular cytotoxicity primarily via macrophages. These results indicated the potential therapeutic efficacy of a glucocorticoid-induced tumor necrosis factor receptor-specific monoclonal antibody in oncolytic virotherapy at local tumor sites.
Collapse
Affiliation(s)
- Mikiya Ishihara
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie, Japan
| | - Naohiro Seo
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie, Japan
- * E-mail: (NS); (HS)
| | - Jun Mitsui
- Department of Gastroenterological Surgery II, Hokkaido University Graduate School of Medicine, Hokkaido, Japan
| | - Daisuke Muraoka
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie, Japan
| | - Maki Tanaka
- Gene Medicine Business Unit, Takara Bio Inc., Shiga, Japan
| | - Junichi Mineno
- Gene Medicine Business Unit, Takara Bio Inc., Shiga, Japan
| | - Hiroaki Ikeda
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie, Japan
| | - Hiroshi Shiku
- Department of Immuno-Gene Therapy, Mie University Graduate School of Medicine, Mie, Japan
- * E-mail: (NS); (HS)
| |
Collapse
|
19
|
Braidwood L, Graham SV, Graham A, Conner J. Oncolytic herpes viruses, chemotherapeutics, and other cancer drugs. Oncolytic Virother 2013; 2:57-74. [PMID: 27512658 PMCID: PMC4918355 DOI: 10.2147/ov.s52601] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Oncolytic viruses are emerging as a potential new way of treating cancers. They are selectively replication-competent viruses that propagate only in actively dividing tumor cells but not in normal cells and, as a result, destroy the tumor cells by consequence of lytic infection. At least six different oncolytic herpes simplex viruses (oHSVs) have undergone clinical trials worldwide to date, and they have demonstrated an excellent safety profile and intimations of efficacy. The first pivotal Phase III trial with an oHSV, talimogene laherparepvec (T-Vec [OncoVex(GM-CSF)]), is almost complete, with extremely positive early results reported. Intuitively, therapeutically beneficial interactions between oHSV and chemotherapeutic and targeted therapeutic drugs would be limited as the virus requires actively dividing cells for maximum replication efficiency and most anticancer agents are cytotoxic or cytostatic. However, combinations of such agents display a range of responses, with antagonistic, additive, or, perhaps most surprisingly, synergistic enhancement of antitumor activity. When synergistic interactions in cancer cell killing are observed, chemotherapy dose reductions that achieve the same overall efficacy may be possible, resulting in a valuable reduction of adverse side effects. Therefore, the combination of an oHSV with "standard-of-care" drugs makes a logical and reasonable approach to improved therapy, and the addition of a targeted oncolytic therapy with "standard-of-care" drugs merits further investigation, both preclinically and in the clinic. Numerous publications report such studies of oncolytic HSV in combination with other drugs, and we review their findings here. Viral interactions with cellular hosts are complex and frequently involve intracellular signaling networks, thus creating diverse opportunities for synergistic or additive combinations with many anticancer drugs. We discuss potential mechanisms that may lead to synergistic interactions.
Collapse
Affiliation(s)
- Lynne Braidwood
- Virttu Biologics Ltd, Department of Neurology, Southern General Hospital, Glasgow, UK
| | - Sheila V Graham
- MRC-University of Glasgow Centre for Virus Research, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, Jarrett Building, University of Glasgow, Glasgow, UK
| | - Alex Graham
- Virttu Biologics Ltd, Department of Neurology, Southern General Hospital, Glasgow, UK
| | - Joe Conner
- Virttu Biologics Ltd, Department of Neurology, Southern General Hospital, Glasgow, UK
| |
Collapse
|
20
|
Meshii N, Takahashi G, Okunaga S, Hamada M, Iwai S, Takasu A, Ogawa Y, Yura Y. Enhancement of systemic tumor immunity for squamous cell carcinoma cells by an oncolytic herpes simplex virus. Cancer Gene Ther 2013; 20:493-8. [PMID: 23887644 DOI: 10.1038/cgt.2013.45] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 06/07/2013] [Accepted: 06/15/2013] [Indexed: 01/13/2023]
Abstract
RH2 is a neurovirulent γ134.5 gene-deficient herpes simplex virus type 1 (HSV-1) with a lytic ability in human squamous cell carcinoma (SCC) cells; it is related to spontaneously occurring HSV-1 mutant HF10. The effect of RH2 on SCC was examined using a syngeneic C3H mouse model. After infection of mouse SCCVII cells with RH2, cell viability was decreased at first, but recovered by prolonged culture, indicating the limited replication of RH2. The antitumor ability of RH2 was examined using a bilateral SCCVII tumor model. The growth of the RH2-injected tumors was suppressed compared with that of phosphate-buffered saline-injected tumors. Moreover, the growth of contralateral tumor of RH2-treated mice was also suppressed significantly. The splenocytes of C3H mice treated with RH2 lysed more SCCVII cells than NFSaY83 cells and YAC-1 cells. The cytotoxicity of the splenocytes on SCCVII cells was significantly greater than that of splenocytes from tumor-bearing mice. Removal of CD8(+) T cells from splenocytes decreased their cell killing activity remarkably. The antitumor effect of RH2 on SCCVII xenografts in nude mice was not demonstrated. These results indicate that RH2 exhibited a suppressive effect on mouse SCC, even if the replication of RH2 was limited. This is ascribed to the ability of RH2 to enhance existing tumor-specific cytotoxic T lymphocyte activity.
Collapse
Affiliation(s)
- N Meshii
- Department of Oral and Maxillofacial Surgery, Osaka University Graduate School of Dentistry, Osaka, Japan
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Takahashi G, Meshii N, Hamada M, Iwai S, Yura Y. Sequence of a fusogenic herpes simplex virus, RH2, for oncolytic virotherapy. J Gen Virol 2012; 94:726-737. [PMID: 23239570 DOI: 10.1099/vir.0.044834-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
RH2 is a novel oncolytic herpes simplex virus type 1 (HSV-1) produced by simultaneous infection with neurovirulent γ134.5 gene-deficient HSV-1 R849 derived from strain F and the spontaneously occurring, fusogenic HSV-1 HF in cell culture. The genome of RH2 was studied using Genome Sequencer FLX. RH2 comprised 149 64 bp and it was shown that the lacZ gene was inserted into the γ134.5 gene of R849. Comparison of ORFs revealed that RH2 had 100 % identity with strain F in 21/58 unique long (UL) genes (36.2%) and 1/13 unique short (US) genes (7.7%). RH2 had 100% amino acid identity with HF10 in 24/58 UL genes (41.4%) and 9/13 US genes (69.2%). Twelve genes, including UL27 (gB), US4 (gG) and UL6 (gD), had amino acid changes unique to RH2. Amino acid changes in gB occurred at positions 459 (T→A) and 817 (L→P). Other unique features were the amino acids missing in UL36 (VP1/2) and UL46 (VP11/12). Thus, RH2 is an HF10-based vector preserving the fusogenic amino acid changes of gB but lacking the γ134.5 gene. RH2 is expected to be a version of HF10 useful for the treatment of brain tumours as well as oral squamous cell carcinoma. Spontaneously occurring HSV-1 mutants may also be useful clinically, as their genome sequences can easily be determined by this genome sequencing system.
Collapse
Affiliation(s)
- Gen Takahashi
- Department of Oral and Maxillofacial Surgery, Osaka University Graduate School of Dentistry, Osaka University, 1-8 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Noritoshi Meshii
- Department of Oral and Maxillofacial Surgery, Osaka University Graduate School of Dentistry, Osaka University, 1-8 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Masakazu Hamada
- Department of Oral and Maxillofacial Surgery, Osaka University Graduate School of Dentistry, Osaka University, 1-8 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Soichi Iwai
- Department of Oral and Maxillofacial Surgery, Osaka University Graduate School of Dentistry, Osaka University, 1-8 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yoshiaki Yura
- Department of Oral and Maxillofacial Surgery, Osaka University Graduate School of Dentistry, Osaka University, 1-8 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
22
|
Esaki S, Goshima F, Kimura H, Murakami S, Nishiyama Y. Enhanced antitumoral activity of oncolytic herpes simplex virus with gemcitabine using colorectal tumor models. Int J Cancer 2012; 132:1592-601. [PMID: 22949155 DOI: 10.1002/ijc.27823] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Accepted: 08/24/2012] [Indexed: 01/10/2023]
Abstract
To enhance the oncolytic activity of herpes simplex viruses (HSVs) control of immune-suppression and immune-resistance by cancer cells is important. Myeloid-derived suppressor cells (MDSCs), which interfere with tumor-suppressive environments, are inhibited by gemcitabine (GEM) treatment. We investigated the oncolytic activity and systemic antitumor immunity induced by oncolytic HSVs in combination with GEM treatment. A mouse model with subcutaneous tumors on both sides of the lateral flanks was used. A highly attenuated HSV type 1, strain HF10, was inoculated into one side of each tumor three times following intraperitoneal injection of GEM. Histopathological changes and IFN-γ secretion of the tumor and leukocytes in the spleen were analyzed. These treatments were repeated to enhance oncolytic activity. HF10 inoculation reduced tumor growth only on the HF10-treated side. HF10 inoculation following GEM treatment resulted in greater reduction of tumor growth on the HF10-treated tumor; furthermore, reduction of tumors on the contralateral untreated side was also observed. Necrosis of the tumor was observed in areas where HSV-infected cells were detected. F4/80(+) macrophages around the tumor were eliminated, and CD4(+) T and CD8(+) T cells increased in the spleen. A single injection of GEM decreased CD11b(+) /Gr-1(+) MDSCs while retaining CD4(+) T cells and CD8(+) T cells. Repetition of this treatment regimen resulted in even greater reduction of tumor growth on both sides and complete rejection in some of the mice. Intratumoral injection of oncolytic HSVs following GEM injection reduced MDSCs. Repeated treatment with oncolytic HSVs following GEM resulted in enhanced oncolytic activity.
Collapse
Affiliation(s)
- Shinichi Esaki
- Department of Virology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | | | | | | | | |
Collapse
|
23
|
Antitumor efficacy of oncolytic herpes simplex virus adsorbed onto antigen-specific lymphocytes. Cancer Gene Ther 2012; 19:292-8. [PMID: 22281756 DOI: 10.1038/cgt.2011.91] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
24
|
Fink DJ, Wechuck J, Mata M, Glorioso JC, Goss J, Krisky D, Wolfe D. Gene therapy for pain: results of a phase I clinical trial. Ann Neurol 2011; 70:207-12. [PMID: 21796661 DOI: 10.1002/ana.22446] [Citation(s) in RCA: 128] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 04/01/2011] [Accepted: 04/01/2011] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Preclinical evidence indicates that gene transfer to the dorsal root ganglion using replication-defective herpes simplex virus (HSV)-based vectors can reduce pain-related behavior in animal models of pain. This clinical trial was carried out to assess the safety and explore the potential efficacy of this approach in humans. METHODS We conducted a multicenter, dose-escalation, phase I clinical trial of NP2, a replication-defective HSV-based vector expressing human preproenkephalin (PENK) in subjects with intractable focal pain caused by cancer. NP2 was injected intradermally into the dermatome(s) corresponding to the radicular distribution of pain. The primary outcome was safety. As secondary measures, efficacy of pain relief was assessed using a numeric rating scale (NRS), the Short Form McGill Pain Questionnaire (SF-MPQ), and concurrent opiate usage. RESULTS Ten subjects with moderate to severe intractable pain despite treatment with >200mg/day of morphine (or equivalent) were enrolled into the study. Treatment was well tolerated with no study agent-related serious adverse events observed at any point in the study. Subjects receiving the low dose of NP2 reported no substantive change in pain. Subjects in the middle- and high-dose cohorts reported pain relief as assessed by NRS and SF-MPQ. INTERPRETATION Treatment of intractable pain with NP2 was well tolerated. There were no placebo controls in this relatively small study, but the dose-responsive analgesic effects suggest that NP2 may be effective in reducing pain and warrants further clinical investigation.
Collapse
Affiliation(s)
- David J Fink
- Department of Neurology, University of Michigan and VA Ann Arbor Healthcare System, Ann Arbor, MI, USA.
| | | | | | | | | | | | | |
Collapse
|
25
|
Takaoka H, Takahashi G, Ogawa F, Imai T, Iwai S, Yura Y. A novel fusogenic herpes simplex virus for oncolytic virotherapy of squamous cell carcinoma. Virol J 2011; 8:294. [PMID: 21663640 PMCID: PMC3131258 DOI: 10.1186/1743-422x-8-294] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2011] [Accepted: 06/10/2011] [Indexed: 01/09/2023] Open
Abstract
Background R849 is a neurovirulent γ134.5 gene-deficient form of herpes simplex virus type 1 (HSV-1) and has LacZ genes at the deleted sites of the γ134.5 gene. HF is a spontaneously occurring, fusogenic HSV-1 strain. The purpose of this work was to generate a virus that has the syncytial character of HF, while preserving the γ134.5 gene inactivation profile of R849 virus. Results Vero cells were infected with R849 and HF simultaneously and two viruses, RH1 and RH2, expressing the LacZ gene and inducing extensive cell fusion were selected. A polymerase chain reaction (PCR)-based analysis suggested that one copy of the γ134.5 gene is lost in RH1, whereas both copies are lost in RH2, and that the γ134.5 gene is replaced by a R849-derived DNA fragment with the LacZ gene. These viruses produced larger plaques and more progeny than the parental viruses. Infection with RH2 decreased the viability of oral squamous cell carcinoma (SCC) cells most strongly. When RH2 was injected into xenografts of oral SCC in nude mice, multinucleated cells were produced and the growth of the tumors was suppressed significantly. Conclusion These results indicate that novel oncolytic HSV-1 vectors can be produced with the genetic background of the oncolytic HSV-1 HF, and that RH2 is deficient in γ134.5 genes and shows extensive cytopathic effects in oral SCC cells. RH2 may be useful in oncolytic virotherapy for oral SCC.
Collapse
Affiliation(s)
- Hiroo Takaoka
- Department of Oral and Maxillofacial Surgery, Osaka University Graduate School of Dentistry, Osaka University, Suita, Osaka, Japan
| | | | | | | | | | | |
Collapse
|
26
|
Campadelli-Fiume G, De Giovanni C, Gatta V, Nanni P, Lollini PL, Menotti L. Rethinking herpes simplex virus: the way to oncolytic agents. Rev Med Virol 2011; 21:213-26. [PMID: 21626603 DOI: 10.1002/rmv.691] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Revised: 03/15/2011] [Accepted: 03/16/2011] [Indexed: 12/22/2022]
Abstract
Oncolytic viruses infect, replicate in and kill cancer cells. HSV has emerged as a most promising candidate because it exerts a generally moderate pathogenicity in humans; it is amenable to attenuation and tropism retargeting; the ample genome provides space for heterologous genes; specific antiviral therapy is available in a worst case scenario. The first strategy to convert HSV into an oncolytic agent consisted in deletion of the γ(1) 34.5 gene which counteracts the protein kinase R (PKR) response, and of the UL39 gene which encodes the large ribonucleotide reductase subunit. Tumor specificity resided in low PKR activity, and high deoxyribonucleotides content of cancer cells. These highly attenuated viruses have been and presently are in clinical trials with encouraging results. The preferred route of administration has been intratumor or in tissues adjacent to resected tumors. Although the general population has a high seroprevalence of antibodies to HSV, studies in animals and humans demonstrate that prior immunity is not an obstacle to systemic routes of administration, and that oncolytic HSV (o-HSVs) do populate tumors. As the attenuated viruses undergo clinical experimentation, the research pipeline is developing novel, more potent and highly tumor-specific o-HSVs. These include viruses which overcome tumor heterogeneity in PKR level by insertion of anti-PKR genes, viruses which reinforce the host tumor clearance capacity by encoding immune cytokines (IL-12 or granulocyte-macrophage colony-stimulating factor), and non-attenuated viruses fully retargeted to tumor specific receptors. A strategy to generate o-HSVs fully retargeted to human epidermal growth factor receptor-2 (HER-2) or other cancer-specific surface receptors is detailed.
Collapse
Affiliation(s)
- Gabriella Campadelli-Fiume
- Department of Experimental Pathology, Section on Microbiology and Virology, Alma Mater Studiorum - University of Bologna, Italy.
| | | | | | | | | | | |
Collapse
|
27
|
A phase I dose-escalation clinical trial of intraoperative direct intratumoral injection of HF10 oncolytic virus in non-resectable patients with advanced pancreatic cancer. Cancer Gene Ther 2010; 18:167-75. [PMID: 21102422 DOI: 10.1038/cgt.2010.65] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In 2005, we initiated a clinical trial that examined the efficacy of the oncolytic virus HF10 to treat pancreatic cancer. Pancreatic cancer continues to have a high mortality rate, despite multimodal treatments for patients, and new therapeutic methods are greatly needed. The current mainstream methods for cancer treatment include biological therapeutics such as trastuzumab (Herceptin) for breast cancer or erlotinib (Tarceva) for non-small cell lung cancer. Oncolytic virus therapy is a new and promising treatment strategy for cancer. Oncolytic viruses are novel biological therapeutics for advanced cancer that appear to have a wide spectrum of anticancer activity with minimal human toxicity. To examine the efficacy of oncolytic virus therapy for pancreatic cancer, we initiated pilot studies by injecting six patients with non-resectable pancreatic cancer with three doses of HF10. All patients were monitored for 30 days for local and systemic adverse effects and were not administered any other therapeutics during this period. There were no adverse side-effects, and we observed some therapeutic potential based on tumor marker levels, survival, pathological findings and diagnostic radiography. The tumors were classified as stable disease in three patients, partial response in one patient and progressive disease in two patients.
Collapse
|
28
|
Carrier cell-based delivery of replication-competent HSV-1 mutants enhances antitumor effect for ovarian cancer. Cancer Gene Ther 2010; 18:77-86. [PMID: 20885447 PMCID: PMC3025316 DOI: 10.1038/cgt.2010.53] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Oncolytic viruses capable of tumor-selective replication and cytolysis have shown early promise as cancer therapeutics. We have developed replication-competent attenuated herpes simplex virus type 1 (HSV-1) mutants, named HF10 and Hh101, which have been evaluated for their oncolytic activities. However, the host immune system remains a significant obstacle to effective intraperitoneal administration of these viruses in the clinical setting. In this study, we investigated the use of these HSV-1 mutants as oncolytic agents against ovarian cancer and the use of human peritoneal mesothelial cells (MCs) as carrier cells for intraperitoneal therapy. MCs were efficiently infected with HSV-1 mutants, and MCs loaded with HSV-1 mutants caused cell killing adequately when cocultured with cancer cells in the presence or absence of HSV antibodies. In a mouse xenograft model of ovarian cancer, the injection of infected carrier cells led to a significant reduction of tumor volume and prolonged survival in comparison with the injection of virus alone. Our results indicate that replication-competent attenuated HSV-1 exerts a potent oncolytic effect on ovarian cancer, which may be further enhanced by the utilization of a carrier cell delivery system, based on amplification of viral load and possibly on avoidance of neutralizing antibodies.
Collapse
|
29
|
Ishida D, Nawa A, Tanino T, Goshima F, Luo CH, Iwaki M, Kajiyama H, Shibata K, Yamamoto E, Ino K, Tsurumi T, Nishiyama Y, Kikkawa F. Enhanced cytotoxicity with a novel system combining the paclitaxel-2'-ethylcarbonate prodrug and an HSV amplicon with an attenuated replication-competent virus, HF10 as a helper virus. Cancer Lett 2009; 288:17-27. [PMID: 19604626 DOI: 10.1016/j.canlet.2009.06.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2009] [Revised: 05/22/2009] [Accepted: 06/16/2009] [Indexed: 11/15/2022]
Abstract
We previously demonstrated that HF10, which is a natural, non-engineered HSV-1, has potent oncolytic activity in the treatment of solid malignant tumors in vitro and in vivo [H. Takakuwa, F. Goshima, N. Nozawa, T. Yoshikawa, H. Kimata, A. Nakao, et al., Oncolytic viral therapy using a spontaneously generated herpes simplex virus type 1 variant for disseminated peritoneal tumor in immunocompetent mice, Arch. Virol. 148 (2003) 813-825; S. Kohno, C. Lou, F. Goshima, Y. Nishiyama, T. Sata, Y. Ono, Herpes simplex virus type 1 mutant HF10 oncolytic viral therapy for bladder cancer, Urology 66 (2005) 1116-1121; D. Watanabe, F. Goshima, I. Mori, Y. Tamada, Y. Matsumoto, Y. Nishiyama, Oncolytic virotherapy for malignant melanoma with herpes simplex virus type 1 mutant HF10, J. Dermatol. Sci. 50 (2008) 185-196; A. Nawa, C. Luo, L. Zhang, Y. Ushijima, D. Ishida, M. Kamakura, et al., Non-engineered, naturally oncolytic herpes simplex virus HSV1 HF10: applications for cancer gene therapy, Curr. Gene. Ther. 8 (2008) 208-221]. Previous reports have also shown that a combination of HF10 and paclitaxel (TAX) was more efficacious than either regimen alone for some types of malignant tumors [S. Shimoyama, F. Goshima, O. Teshigahara, H. Kasuya, Y. Kodera, A. Nakao, et al., Enhanced efficacy of herpes simplex virus mutant HF10 combined with paclitaxel in peritoneal cancer dissemination models, Hepatogastroenterology 54 (2007) 1038-1042]. In this study, we investigated the efficacy of gene-directed enzyme prodrug therapy (GDEPT) using a novel system that combines the paclitaxel-2'-ethylcarbonate prodrug (TAX-2'-Et) and an HSV amplicon expressing rabbit-carboxylesterase (CES) with HF10 as a helper virus. This GDEPT system aims to produce high level of CES at the tumor site, resulting in efficient local conversion of the TAX-2'-Et prodrug into the active drug TAX [A. Nawa, T. Tanino, C. Lou, M. Iwaki, H. Kajiyama, K. Shibata, et al., Gene directed enzyme prodrug therapy for ovarian cancer: could GDEPT become a promising treatment against ovarian cancer?, Anti-Cancer Agents Med Chem 8 (2008) 232-239]. We demonstrated that the green fluorescent protein (GFP) gene, as a trace maker, was more efficiently introduced by the HSV amplicon compared to the expression vector, pHGCX, and that the HSV amplicon system expressed an active CES enzyme that could convert TAX-2'-Et to TAX in Cos7 cells. Furthermore, although the cytotoxicity of this amplicon system was not enhanced in virus-sensitive tumor cells, it was significantly enhanced in low virus-sensitive tumor cells in the presence of the prodrug in a concentration-dependent manner, compared to the control virus alone (p<0.05). These results indicate that the addition of a prodrug converting enzyme may be a feasible approach to further enhance the efficacy of HF10 as a cancer therapeutics in low HF10-sensitive malignancies.
Collapse
Affiliation(s)
- Daisuke Ishida
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Shouwa-ku, Nagoya 466-8550, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
Gene transfer to the dorsal root ganglion using replication defective herpes simplex virus (HSV)-based vectors reduces pain-related behaviors in rodent models having inflammatory pain, neuropathic pain and pain caused by cancer in bone. HSV vectors engineered to produce inhibitory neurotransmitters, including the delta opioid agonist peptide enkephalin, the mu opioid agonist peptide endomorphin-2 and glutamic acid decarboxylase (GAD), to effect the release of gamma amino butyric acid (GABA) act to inhibit nociceptive neurotransmission at the first synapse between primary nociceptive and second-order neuron in the dorsal horn of the spinal cord. HSV vectors engineered to release anti-inflammatory peptides, including interleukin (IL)-4, IL-10 and the p55 soluble tumor necrosis factor alpha (TNFalpha) receptor reduce neuroimmune activation in the spinal dorsal horn. The path leading from preclinical animal studies to the ongoing phase 1 human trial of the enkephalin-producing vector in patients with pain from cancer, and plans for an efficacy trial with an opioid-producing vector in inflammatory pain and an efficacy trial with a GAD-producing vector in diabetic neuropathic pain are outlined.
Collapse
|
31
|
Nomura N, Kasuya H, Watanabe I, Shikano T, Shirota T, Misawa M, Sugimoto H, Kanazumi N, Nomoto S, Takeda S, Nakao A. Considerations for intravascular administration of oncolytic herpes virus for the treatment of multiple liver metastases. Cancer Chemother Pharmacol 2008; 63:321-30. [PMID: 18575868 DOI: 10.1007/s00280-008-0742-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2007] [Accepted: 03/17/2008] [Indexed: 11/25/2022]
Abstract
PURPOSE Oncolytic viral therapy is a newly developed modality for treating tumors. Many clinical trials using oncolytic virus have been performed worldwide, but most of them have used local injection in the tumor. Determination of the effect and safety of intravascular virus injection instead of local injection is necessary for clinical use against multiple liver metastases and systemic metastases. METHODS To evaluate the efficacy and safety of intravascular virus therapy, mice bearing multiple liver metastases were treated by intraportal or intravenous administration of the herpes simplex virus type 1 (HSV-1) mutant, hrR3. Mice treated with hrR3 were killed and organs were harvested for lacZ staining and PCR analysis. Inactivation of oncolytic virus in bloodstream was assessed by neutralization assay in vitro. Infectious activity of hrR3 with vascular endothelial cells was evaluated by replication and cytotoxicity assay. RESULTS The survival rate of animals treated by hrR3 was significantly improved compared with the untreated group. lacZ staining and PCR analysis demonstrated detectable virus in the tumor but not in normal tissue or other organs except for the adrenal glands. We also showed that vascular endothelial cells allowed virus replication, while normal hepatocytes did not, and human anti-HSV antibody revealed attenuation of the infectious activity of hrR3. CONCLUSIONS Intravascular delivery of hrR3 is effective in treating multiple liver metastases, however, several points must be kept in mind at the time of human clinical trials using intravascular virus administration in order to avoid critical side effects.
Collapse
Affiliation(s)
- Naohiro Nomura
- Department of Surgery II, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Watanabe D, Goshima F, Mori I, Tamada Y, Matsumoto Y, Nishiyama Y. Oncolytic virotherapy for malignant melanoma with herpes simplex virus type 1 mutant HF10. J Dermatol Sci 2008; 50:185-96. [PMID: 18226503 DOI: 10.1016/j.jdermsci.2007.12.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2007] [Revised: 11/13/2007] [Accepted: 12/03/2007] [Indexed: 12/22/2022]
Abstract
BACKGROUND Many viruses have been engineered and evaluated for their potential as therapeutic agents in the treatment of malignant neoplasm, including malignant melanoma. OBJECTIVE In this study, we investigated the efficacy of HF10, an attenuated, replication-competent HSV, in immunocompetent animal models with malignant melanoma. METHODS For in vitro study, viral cytotoxicity assays and replication assays were performed both in human and mouse melanoma cells. For the study in vivo, intraperitoneally disseminated or subcutaneous melanoma models were prepared in DBA/2 mice using clone M3 cells, then HF10 was inoculated intraperitoneally or intratumorally. Therapeutic efficacy of HF10 was assessed by survival, tumor growth, and histopathological analysis. RESULTS HF10 infection produced cytolytic effects in melanoma cells at various multiplicities of infection (MOI). In the intraperitoneal melanoma model, all mice survived when given intraperitoneal injections of HF10 compared with 100% fatality in the control mice. In the subcutaneous tumor model, intratumoral inoculation of HF10 significantly reduced tumor growth. Histology and immunohistochemistry showed tumor lysis and inflammatory cell infiltration after intratumoral HF10 inoculation. Viral antigen was retained at the inoculation site until 7 days post-infection. HF10-treated intraperitoneal tumor mice were also protected against tumor rechallenge. HF10 also affected the non-inoculated contralateral tumor when injected into the ipsilateral tumor of mice, suggesting that HF10 can induce systemic antitumor immune responses in mice. CONCLUSION Oncolytic viral therapy using HF10 was effective in melanoma mouse models, and intratumoral injection of HF10 induced systemic antitumor responses. These results suggest that HF10 is a promising agent for the treatment of advanced melanoma.
Collapse
Affiliation(s)
- Daisuke Watanabe
- Department of Dermatology, Aichi Medical University, Nagakute, Aichi, Japan.
| | | | | | | | | | | |
Collapse
|
33
|
Luo C, Mori I, Goshima F, Ushijima Y, Nawa A, Kimura H, Nishiyama Y. Replication-competent, oncolytic herpes simplex virus type 1 mutants induce a bystander effect following ganciclovir treatment. J Gene Med 2007; 9:875-83. [PMID: 17685493 DOI: 10.1002/jgm.1085] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Cells expressing herpes simplex virus (HSV) thymidine kinase (tk) are killed by ganciclovir (GCV). Adjacent cells without HSV-tk also die, a phenomenon known as the 'bystander effect'. However, there is no evidence that replication-competent HSV induces a bystander effect in the presence of GCV. Therefore, we investigated the bystander effect in HEp-2 cells infected with replication-competent, oncolytic HSV-1 mutants, hrR3 and HF10. In cells infected at a multiplicity of infection (MOI) of 3, GCV did not induce apoptosis. At low MOIs of 0.3 and 0.03, however, a number of adjacent, uninfected cells apoptosed following GCV treatment. Irrespective of GCV treatment, HEp-2 cells expressed minimal levels of connexin 43 (Cx43). However, Cx43 expression was enhanced by GCV in response to infection with HF10 at an MOI of 0.3, but not at an MOI of 3. Expression of other proteins involved in gap junctions, including Cx26 and Cx40, was not augmented under these conditions. The PKA and PI3K signal transduction pathways are likely involved in enhanced Cx43 expression as inhibitors of these pathways prevented Cx43 upregulation. These results suggest that infection with replication-competent HSV-1 induces the bystander effect in cells treated with GCV because of efficient intercellular transport of active GCV through abundant gap junctions.
Collapse
Affiliation(s)
- Chenhong Luo
- Department of Virology, Nagoya University Graduate School of Medicine, Tsuruma, Nagoya 466-8550, Japan
| | | | | | | | | | | | | |
Collapse
|
34
|
[Oncolytic virotherapy using replication-competent herpes simplex viruses]. Uirusu 2007; 57:57-65. [PMID: 18040155 DOI: 10.2222/jsv.57.57] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Oncolytic virotherapy using replication-competent viruses has attracted us as a new modality for cancer treatment. The fundamental concept of oncolytic virotherapy is that the viruses selectively replicate in and lyse tumor cells. Since 1997, numbers of clinical trials have been done in over 500 cancer patients. However, the results of those trials have been disappointing in most cases. We have isolated a spontaneously occurring herpes simplex virus type 1 mutant, designated HF10, which efficiently replicates and induces cell fusion in most transformed cells, but is highly attenuated in mice. HF10 has a number of deletions and insertions in the genome, resulting in the lack of the functional expression of UL43, UL49.5, UL55, UL56 and latency-associated transcripts. We have found that HF10 can be used as an oncolytic virus for treatment of malignant tumors in various animal models. Clinical trials have shown that intratumoral injection of HF10 can induce extensive tumor cell death in patients with recurrent breast cancer and head and neck squamous cell carcinoma without significant adverse effects. HF10 is a promising agent for use in oncolytic virotherapy in non-central nervous system malignancies.
Collapse
|
35
|
Watanabe I, Kasuya H, Nomura N, Shikano T, Shirota T, Kanazumi N, Takeda S, Nomoto S, Sugimoto H, Nakao A. Effects of tumor selective replication-competent herpes viruses in combination with gemcitabine on pancreatic cancer. Cancer Chemother Pharmacol 2007; 61:875-82. [PMID: 17726607 DOI: 10.1007/s00280-007-0567-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2007] [Accepted: 07/25/2007] [Indexed: 10/22/2022]
Abstract
PURPOSE Pancreatic cancer still has a poor prognosis, even if aggressive therapy is pursued. Currently, new modalities of oncolytic virus therapy are being tested against this cancer. The combination of one of two representative mutant herpes simplex viruses (R3616: gamma(1)34.5 inactivated, hrR3: UL39 inactivated) with a standard anti-pancreatic cancer chemotherapy drug (gemcitabine), was investigated in this study. EXPERIMENTAL DESIGN The intracellular concentration of ribonucleotide reductase was estimated by Western blotting. The effect of gemcitabine on viral replication and the total cytotoxic effect of the combination therapy were investigated on pancreatic cancer cell lines. We compared the results of two oncolytic viruses, R3616 and hrR3. A mouse model of pancreatic cancer with peritoneal dissemination was used to evaluate the in vivo effect of the combination therapy. RESULTS Although the replication of both viruses was inhibited by gemcitabine, the combination caused more tumor cell cytotoxicity than did virus alone in vitro. The results with R3616 were more striking. Although the difference was not statistically significant, R3616 with gemcitabine had a greater effect than did R3616 alone, while hrR3 with gemcitabine had a weaker effect than did hrR3 alone in vivo experiments. CONCLUSION The combination of oncolytic virus with gemcitabine is a promising new strategy against advanced pancreatic cancer. Each virus has different functional characteristics, and can affect the results of the combination of viruses and chemotherapy drugs. The results indicate that there is a complicated interaction among viruses, cells, and chemotherapy drugs and that the best combination of oncolytic virus and chemotherapeutic agents should be studied more extensively before embarking on a clinical trial.
Collapse
Affiliation(s)
- Izuru Watanabe
- Department of Surgery II, Nagoya University School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Kohno SI, Luo C, Nawa A, Fujimoto Y, Watanabe D, Goshima F, Tsurumi T, Nishiyama Y. Oncolytic virotherapy with an HSV amplicon vector expressing granulocyte–macrophage colony-stimulating factor using the replication-competent HSV type 1 mutant HF10 as a helper virus. Cancer Gene Ther 2007; 14:918-26. [PMID: 17693992 DOI: 10.1038/sj.cgt.7701070] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Direct viral infection of solid tumors can cause tumor cell death, but these techniques offer the opportunity to express exogenous factors to enhance the antitumor response. We investigated the antitumor effects of a herpes simplex virus (HSV) amplicon expressing mouse granulocyte-macrophage colony-stimulating factor (mGM-CSF) using the replication-competent HSV type 1 mutant HF10 as a helper virus. HF10-packaged mGM-CSF-expressing amplicon (mGM-CSF amplicon) was used to infect subcutaneously inoculated murine colorectal tumor cells (CT26 cells) and the antitumor effects were compared to tumors treated with only HF10. The mGM-CSF amplicon efficiently replicated in CT26 cells with similar oncolytic activity to HF10 in vitro. However, when mice subcutaneously inoculated with CT26 cells were intratumorally injected with HF10 or mGM-CSF amplicon, greater tumor regression was seen in mGM-CSF amplicon-treated animals. Furthermore, mGM-CSF amplicon treatment prolonged mouse survival. Immunohistochemical analysis revealed increased inflammatory cell infiltration in the solid tumor in the mGM-CSF amplicon-treated animals. These results suggest that expression of GM-CSF enhances the antitumor effects of HF10, and HF10-packaged GM-CSF-expressing amplicon is a promising agent for the treatment of subcutaneous tumors.
Collapse
Affiliation(s)
- S-I Kohno
- Department of Virology, Graduate School of Medicine, Nagoya University, Showa-ku, Nagoya, Japan
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Kasuya H, Nishiyama Y, Nomoto S, Goshima F, Takeda S, Watanabe I, Nomura N, Shikano T, Fujii T, Kanazumi N, Nakao A. Suitability of a US3-inactivated HSV mutant (L1BR1) as an oncolytic virus for pancreatic cancer therapy. Cancer Gene Ther 2007; 14:533-42. [PMID: 17415379 DOI: 10.1038/sj.cgt.7701049] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Recently, the use of oncolytic viruses against cancer has attracted considerable attention. We studied the potential of the US3 locus-deficient herpes simplex virus (HSV), L1BR1, for oncolytic virus therapy. Its high specificity and potency indicate that L1BR1 is a promising candidate as a new oncolytic virus against pancreatic cancer. Moreover, the virus exhibited the unique characteristic of increasing apoptosis when used in combination with anticancer drugs. We assessed the feasibility of using the US3 locus-deficient HSV named L1BR1 as a new replication-competent oncolytic virus for the treatment of pancreatic cancer. The US3 locus of HSV has been shown to be a key gene in producing a multifunctional protein kinase that inhibits apoptosis induced by viral infections, chemicals and ultraviolet (UV) light. L1BR1 has been reported to be more than 10 000-fold less virulent than the parental virus in mice. In this study, we examined the tumor specificity and oncolytic effect of this attenuated replication-competent virus, L1BR1, in pancreatic cancers derived from SW1990, Capan2 and Bxpc-3cells compared with the parent virus and other well-known oncolytic herpes viruses (R3616 and hrR3). We also studied the efficacy of L1BR1 for the induction of apoptosis as an attribute of this virus in combination with the anticancer drugs 5FU and cisplatin. The combined treatment of the pancreatic cancer cells with L1BR1 and these anticancer drugs enhanced apoptosis significantly. More importantly, L1BR1 showed the lowest replication capacity in normal human hepatocytes, but the highest tumor-reducing effect in vivo among the oncolytic herpes viruses tested. In addition, L1BR1 significantly increased the induction of apoptosis of cancer cells when treated in combination with anticancer drugs although the parental virus inhibited the induction of apoptosis. These results suggest that L1BR1 is promising as a new anticancer oncolytic virus.
Collapse
Affiliation(s)
- H Kasuya
- Department of Surgery II, Nagoya University School of Medicine, Nagoya, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Vähä-Koskela MJ, Heikkilä JE, Hinkkanen AE. Oncolytic viruses in cancer therapy. Cancer Lett 2007; 254:178-216. [PMID: 17383089 PMCID: PMC7126325 DOI: 10.1016/j.canlet.2007.02.002] [Citation(s) in RCA: 220] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2006] [Revised: 02/01/2007] [Accepted: 02/05/2007] [Indexed: 12/26/2022]
Abstract
Oncolytic virotherapy is a promising form of gene therapy for cancer, employing nature’s own agents to find and destroy malignant cells. The purpose of this review is to provide an introduction to this very topical field of research and to point out some of the current observations, insights and ideas circulating in the literature. We have strived to acknowledge as many different oncolytic viruses as possible to give a broader picture of targeting cancer using viruses. Some of the newest additions to the panel of oncolytic viruses include the avian adenovirus, foamy virus, myxoma virus, yaba-like disease virus, echovirus type 1, bovine herpesvirus 4, Saimiri virus, feline panleukopenia virus, Sendai virus and the non-human coronaviruses. Although promising, virotherapy still faces many obstacles that need to be addressed, including the emergence of virus-resistant tumor cells.
Collapse
Affiliation(s)
- Markus J.V. Vähä-Koskela
- Åbo Akademi University, Department of Biochemistry and Pharmacy and Turku Immunology Centre, Turku, Finland
- Turku Graduate School of Biomedical Sciences, Turku, Finland
- Corresponding author. Address: Åbo Akademi University, Department of Biochemistry and Pharmacy and Turku Immunology Centre, Turku, Finland. Tel.: +358 2 215 4018; fax: +358 2 215 4745.
| | - Jari E. Heikkilä
- Åbo Akademi University, Department of Biochemistry and Pharmacy and Turku Immunology Centre, Turku, Finland
| | - Ari E. Hinkkanen
- Åbo Akademi University, Department of Biochemistry and Pharmacy and Turku Immunology Centre, Turku, Finland
| |
Collapse
|
39
|
Ushijima Y, Luo C, Goshima F, Yamauchi Y, Kimura H, Nishiyama Y. Determination and analysis of the DNA sequence of highly attenuated herpes simplex virus type 1 mutant HF10, a potential oncolytic virus. Microbes Infect 2006; 9:142-9. [PMID: 17218138 DOI: 10.1016/j.micinf.2006.10.019] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2006] [Revised: 10/29/2006] [Accepted: 10/31/2006] [Indexed: 02/04/2023]
Abstract
A spontaneously occurring herpes simplex virus type 1 (HSV-1) mutant, designated HF10, replicates very efficiently and induces extensive cell fusion in most transformed cells as well as Vero cells, but is highly attenuated in mice when inoculated by peripheral routes of infection. Recent studies have shown that HF10 is a promising agent for use in oncolytic virotherapy. In this study, we sequenced the genome of HF10 and compared it with that of HSV-1 strain 17, a reference strain with the syn+ phenotype. The sequencing covered whole regions corresponding to all open reading frames of strain 17, and the overall putative amino acid identity between HF10 and strain 17 was 99.1% except for proteins encoded by three genes with frame-shift mutations. HF10 had a number of deletions and insertions in the genome, resulting in the lack of the functional expression of UL43, UL49.5, UL55, UL56 and latency-associated transcripts. Additionally, HF10 had amino acid changes in genes involved in the regulation of syncytium formation, including UL1, UL20, UL22, UL24, UL27 and UL53. The proteins encoded by UL1, UL2, UL11, UL44, US1, US7, US8.5, US10 and US12 exhibited a relatively high divergence. These data provide the genetic background of HF10 and insight into the molecular mechanism of HSV-1 replication and pathogenicity.
Collapse
Affiliation(s)
- Yoko Ushijima
- Department of Virology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | | | | | | | | | | |
Collapse
|
40
|
Zhang L, Daikoku T, Ohtake K, Ohtsuka J, Nawa A, Kudoh A, Iwahori S, Isomura H, Nishiyama Y, Tsurumi T. Establishment of a novel foreign gene delivery system combining an HSV amplicon with an attenuated replication-competent virus, HSV-1 HF10. J Virol Methods 2006; 137:177-83. [PMID: 16854473 DOI: 10.1016/j.jviromet.2006.06.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2006] [Revised: 06/09/2006] [Accepted: 06/13/2006] [Indexed: 11/18/2022]
Abstract
Herpes simplex virus type 1 (HSV-1)-based amplicon vectors have been used widely in genetic engineering with many advantages for gene delivery, being easily constructed. An attenuated and replication-competent HSV-1 HF10 clone demonstrating an oncolytic effect on cancer cells in vitro and in vivo has been applied recently for clinical virotherapy of breast cancers and the present studies were conducted to test its efficacy in combination with an HSV-1 amplicon. For this purpose, a new system was developed to produce high titers of the HSV-1 amplicon vector and the results showed that its package efficiency and the titer ratio to HF10 were improved by passage through two cell lines. A high ratio of amplicon/helper virus HF10 (A/H) (>1) was required to express the foreign gene efficiently. Furthermore, in order to express the foreign gene conditionally, an HSV-1 ICP8 promoter was introduced in place of the human cytomegalovirus MIE promoter, this driving expression of the transgene when replication of HF10 progressed. The methodology for simple preparation of mixtures of viruses containing the amplicon with the oncolytic virus is documented. This system should find application for studies of cancer therapy.
Collapse
Affiliation(s)
- Lumin Zhang
- Division of Virology, Aichi Cancer Center Research Institute, 1-1, Kanokoden, Chikusa-ku, Nagoya 464-8681, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Fujimoto Y, Mizuno T, Sugiura S, Goshima F, Kohno SI, Nakashima T, Nishiyama Y. Intratumoral injection of herpes simplex virus HF10 in recurrent head and neck squamous cell carcinoma. Acta Otolaryngol 2006; 126:1115-7. [PMID: 16923721 DOI: 10.1080/00016480600702100] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
We have developed a novel replication-competent, oncolytic herpes simplex virus (HSV), named HF10, and have evaluated its anticancer efficacy in a variety of animal models. We report a pilot study of intratumoral injection of HF10 into subcutaneous nodules in patients with head and neck squamous cell carcinoma (HNSCC). HF10 efficiently infected human HNSCC cells and caused extensive tumor cell death without any significant adverse effects, suggesting that HF10 represents a promising therapy for HNSCC in humans. To assess the therapeutic potential of HF10 in human HNSCC, we performed a preliminary study of toxicity and efficacy in two patients with recurrent metastatic HNSCC. For each patient, a metastatic skin nodule was injected with HF10 once a day for 3 days. They were monitored for systemic adverse effects, and the injected nodules were excised at day 13 (patient 1) or day 15 (patient 2) after injection for histochemical examination. HF10 replicated, spread well in the tumor nodules, and caused cell death in a considerable population of tumor cells without any significant adverse effects.
Collapse
Affiliation(s)
- Yasushi Fujimoto
- Department of Otorhinolaryngology, Graduate School of Medicine, Nagoya University, Nagoya, Japan.
| | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Cancer remains a serious threat to human health, causing over 500 000 deaths each year in US alone, exceeded only by heart diseases. Many new technologies are being developed to fight cancer, among which are gene therapies and oncolytic virotherapies. Herpes simplex virus type 1 (HSV-1) is a neurotropic DNA virus with many favorable properties both as a delivery vector for cancer therapeutic genes and as a backbone for oncolytic viruses. Herpes simplex virus type 1 is highly infectious, so HSV-1 vectors are efficient vehicles for the delivery of exogenous genetic materials to cells. The inherent cytotoxicity of this virus, if harnessed and made to be selective by genetic manipulations, makes this virus a good candidate for developing viral oncolytic approach. Furthermore, its large genome size, ability to infect cells with a high degree of efficiency, and the presence of an inherent replication controlling mechanism, the thymidine kinase gene, add to its potential capabilities. This review briefly summarizes the biology of HSV-1, examines various strategies that have been used to genetically modify the virus, and discusses preclinical as well as clinical results of the HSV-1-derived vectors in cancer treatment.
Collapse
Affiliation(s)
- Y Shen
- Mary Crowley Medical Research Center, Dallas, TX 75201, USA
| | | |
Collapse
|
43
|
Kohno SI, Luo C, Goshima F, Nishiyama Y, Sata T, Ono Y. Herpes simplex virus type 1 mutant HF10 oncolytic viral therapy for bladder cancer. Urology 2005; 66:1116-21. [PMID: 16286150 DOI: 10.1016/j.urology.2005.05.041] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2005] [Revised: 04/27/2005] [Accepted: 05/16/2005] [Indexed: 10/25/2022]
Abstract
OBJECTIVES To investigate the antitumor effects of the oncolytic herpes simplex virus (HSV) type 1 mutant HF10 on human and murine bladder cancer cells (T24 and MBT-2) in vitro and in immunocompetent mouse models. METHODS In vitro viral oncolytic activity and the replication ability of HF10 were measured in T24 and MBT-2 cells. To evaluate the therapeutic efficacy of HF10, disseminated peritoneal and bladder cancer models using MBT-2 cells were established in C3H/HeJ mice. The therapeutic efficacy was estimated from the survival rates and histopathologic analyses. RESULTS HF10 replicated well in both T24 and MBT-2 cells, and it induced extensive cell lysis. Treatment with HF10 significantly prolonged the survival periods and increased the survival rates in both models tested. Immunohistochemical studies showed that HSV antigens were detected in the bladders 1 and 3 days after intravesical treatment with HF10 in nonimmunized mice, but only at 1 day after HF10 treatment in preimmunized, HSV-1 antibody-positive mice. A large number of inflammatory cells infiltrated into the bladder mucosa at 3 days after HF10 treatment in the preimmunized mice. CONCLUSIONS These results suggest that HF10, a novel oncolytic HSV-1 mutant, is a promising agent for the treatment of superficial bladder cancer.
Collapse
Affiliation(s)
- Shin-ichi Kohno
- Department of Virology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | | | | | |
Collapse
|
44
|
Kasuya H, Takeda S, Nomoto S, Nakao A. The potential of oncolytic virus therapy for pancreatic cancer. Cancer Gene Ther 2005; 12:725-36. [PMID: 15818382 DOI: 10.1038/sj.cgt.7700830] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The objective of this paper was to review a new category of gene therapy using oncolytic viruses for the treatment of pancreatic cancer. The eligibility and feasibility of oncolytic virus therapy as a novel therapeutic agent against pancreatic cancer are discussed as well as basic research for clinical trials, including a historical perspective and the current status of these novel agents. Even combination therapy, such as surgery with radiation and chemotherapy, has not significantly improved the survival rate of pancreatic cancer. Recently, a clinical trial (phase I and II) using an oncolytic adenovirus, ONYX-015, was completed in patients with pancreatic cancer. The phase II trial yielded beneficial results (tumor reduction or stabilization) in about 50% of the patients. A phase I study of the efficacy of oncolytic herpes viruses, G207, OncoVEX GM-CSF, and 1716 against a variety of tumors has been completed, and G207 is in phase II trials for use against brain tumors. In addition, a phase I trial using the herpesvirus showed good tolerance at all dosages. We discuss the basic scientific principles and current results of the above clinical trials with respect to these oncolytic viruses, and then compare the relative advantages and disadvantages of adenoviruses and herpesviruses as oncolytic agents. We also review the published literature on newly developed oncolytic viruses. The concept of oncolytic therapy has been studied for a century. Recent technological developments have made these oncolytic viruses more tumor-specific by exploiting the tumor cell environments. In addition, these viruses have been reported to increase the immunosusceptibility of the tumor cells, and have been designed to express other genes to increase the susceptibility of tumor cells to other therapeutic agents. Oncolytic virus therapy certainly appears to be a feasible treatment for pancreatic cancer.
Collapse
Affiliation(s)
- Hideki Kasuya
- Surgery II, Nagoya University, 65 Tsurumai-cho, Showa-ku, Nagoya, Japan.
| | | | | | | |
Collapse
|
45
|
Chun YS, Adusumilli PS, Fong Y. Employing tumor hypoxia for oncolytic therapy in breast cancer. J Mammary Gland Biol Neoplasia 2005; 10:311-8. [PMID: 16826462 DOI: 10.1007/s10911-006-9004-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
Abstract
Hypoxia is a common tumor condition associated with metastases, therapeutic resistance, and poor patient survival. Forty percent of breast cancers are hypoxic, with a median oxygen concentration of 3.9%, and a third of tumors have regions less than 0.3%. Normal breast tissue is reported to have oxygen concentrations greater than 9%. This tumor hypoxia in breast cancer confers resistance to conventional radiation therapy and chemotherapy, as well as making estrogen-receptor-positive tumors less sensitive to hormonal therapy. Novel treatment modalities are needed to target hypoxic tumor cells. Lower tumor oxygen levels compared with surrounding normal tissues may be utilized to target and enhance herpes oncolytic viral therapy in breast cancer. Attenuated oncolytic herpes simplex viruses offer a unique cancer treatment by specifically infecting, replicating within, and lysing tumor cells. They carry genetically engineered mutations to reduce their virulence and attenuate their ability to infect normal tissues. Studies have shown the safety and efficacy of oncolytic herpes simplex viruses in treating breast cancer both in humans and in preclinical models. The placement of essential viral genes under the control of a hypoxia-responsive enhancer, which is upregulated selectively in hypoxic tissue, represents a promising strategy to target oncolytic viruses precisely to hypoxic cancer cells. In this review we describe strategies to harness hypoxia as a trigger for oncolytic viral gene expression in breast cancer, thereby increasing the specificity of viral infection, replication, and cytotoxicity to hypoxic areas of tumor. Such a targeted approach will increase efficacy in the therapy of hypoxic tumors while achieving a reduction in total dose of viral therapy.
Collapse
Affiliation(s)
- Yun Shin Chun
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA
| | | | | |
Collapse
|
46
|
Mori I, Liu B, Goshima F, Ito H, Koide N, Yoshida T, Yokochi T, Kimura Y, Nishiyama Y. HF10, an attenuated herpes simplex virus (HSV) type 1 clone, lacks neuroinvasiveness and protects mice against lethal challenge with HSV types 1 and 2. Microbes Infect 2005; 7:1492-500. [PMID: 16054416 DOI: 10.1016/j.micinf.2005.05.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2005] [Revised: 05/09/2005] [Accepted: 05/10/2005] [Indexed: 11/30/2022]
Abstract
Herpes simplex virus (HSV), a neurotropic virus, establishes life-long and, although rare, life-threatening infection in humans, and it may precipitate substantial medical and psychosocial morbidity. Here we show that HSV-1 strain HF clone 10 (HF10) exhibits impaired neuroinvasiveness in peripheral olfactory, vomeronasal and trigeminal conduits following intranasal as well as corneal inoculation. HF10 attenuation likely arises from multiple defects of HSV genes, so that HF10 will not revert to a virulent phenotype. Intranasal vaccination of mice with HF10 conferred significant protection against lethal challenge with HSV-1 and HSV-2 via the intranasal and intravaginal routes. Thus, we propose that HF10 explicitly meets the prerequisites for a candidate live attenuated HSV vaccine.
Collapse
Affiliation(s)
- Isamu Mori
- Department of Microbiology and Immunology, Aichi Medical University School of Medicine, 480-1195, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|