1
|
Nikrad JA, Galvin RT, Sheehy MM, Novacek EL, Jacobsen KL, Corbière SM, Beckmann PJ, Jubenville TA, Yamamoto M, Largaespada DA. Conditionally replicative adenovirus as a therapy for malignant peripheral nerve sheath tumors. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200783. [PMID: 38595983 PMCID: PMC10959710 DOI: 10.1016/j.omton.2024.200783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 02/14/2024] [Accepted: 02/26/2024] [Indexed: 04/11/2024]
Abstract
Oncolytic adenoviruses (Ads) stand out as a promising strategy for the targeted infection and lysis of tumor cells, with well-established clinical utility across various malignancies. This study delves into the therapeutic potential of oncolytic Ads in the context of neurofibromatosis type 1 (NF1)-associated malignant peripheral nerve sheath tumors (MPNSTs). Specifically, we evaluate conditionally replicative adenoviruses (CRAds) driven by the cyclooxygenase 2 (COX2) promoter, as selective agents against MPNSTs, demonstrating their preferential targeting of MPNST cells compared with non-malignant Schwann cell control. COX2-driven CRAds, particularly those with modified fiber-knobs exhibit superior binding affinity toward MPNST cells and demonstrate efficient and preferential replication and lysis of MPNST cells, with minimal impact on non-malignant control cells. In vivo experiments involving intratumoral CRAd injections in immunocompromised mice with human MPNST xenografts significantly extend survival and reduce tumor growth rate compared with controls. Moreover, in immunocompetent mouse models with MPNST-like allografts, CRAd injections induce a robust infiltration of CD8+ T cells into the tumor microenvironment (TME), indicating the potential to promote a pro-inflammatory response. These findings underscore oncolytic Ads as promising, selective, and minimally toxic agents for MPNST therapy, warranting further exploration.
Collapse
Affiliation(s)
- Julia A. Nikrad
- Department of Pediatrics, Medical School, University of Minnesota, 420 Delaware Street SE, Mayo Mail Code 484, Minneapolis, MN 55455, USA
| | - Robert T. Galvin
- Department of Pediatrics, Medical School, University of Minnesota, 420 Delaware Street SE, Mayo Mail Code 484, Minneapolis, MN 55455, USA
| | - Mackenzie M. Sheehy
- Department of Pediatrics, Medical School, University of Minnesota, 420 Delaware Street SE, Mayo Mail Code 484, Minneapolis, MN 55455, USA
| | - Ethan L. Novacek
- Department of Pediatrics, Medical School, University of Minnesota, 420 Delaware Street SE, Mayo Mail Code 484, Minneapolis, MN 55455, USA
| | - Kari L. Jacobsen
- Department of Surgery, University of Minnesota, 516 Delaware Street SE, Minneapolis, MN 55455, USA
| | - Stanislas M.A.S. Corbière
- Institute for Research in Immunology and Cancer, Université de Montréal, 2950 Chemin de Polytechnique Marcelle-Coutu Pavilion, Montréal, QC H3T1J4, Canada
| | - Pauline J. Beckmann
- Department of Pediatrics, Medical School, University of Minnesota, 420 Delaware Street SE, Mayo Mail Code 484, Minneapolis, MN 55455, USA
| | - Tyler A. Jubenville
- Department of Pediatrics, Medical School, University of Minnesota, 420 Delaware Street SE, Mayo Mail Code 484, Minneapolis, MN 55455, USA
| | - Masato Yamamoto
- Department of Surgery, University of Minnesota, 516 Delaware Street SE, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN 55455, USA
| | - David A. Largaespada
- Department of Pediatrics, Medical School, University of Minnesota, 420 Delaware Street SE, Mayo Mail Code 484, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN 55455, USA
| |
Collapse
|
2
|
Ogasawara S. Replication-competent influenza virus with a protein-responsive multiplication ability. N Biotechnol 2023; 77:100-110. [PMID: 37586547 DOI: 10.1016/j.nbt.2023.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 07/30/2023] [Accepted: 08/12/2023] [Indexed: 08/18/2023]
Abstract
Applications of influenza A viruses (IAV) for virotherapy and biotechnology have accelerated substantially with the development of reverse genetic technology and advances in the understanding of packaging signals. While the use of a replication-competent IAV is particularly promising, owing to its efficient transmission to organ depths with high infectivity, there is also a risk that its multiplication cannot be controlled in a cell-type-specific manner, causing an infectious disease. Therefore, here a simple and effective replication-competent IAV-based cell-targeting system has been developed. It was demonstrated that the activity of the ribonucleoprotein complex (RNP) of IAV could be regulated by the interaction between the endogenous protein and a nanobody fused to the subunit of RNA-dependent RNA polymerase (RdRp). To validate the feasibility of the method, it was demonstrated that RNP containing RdRp fused with Nb139, a nanobody against p53, is inactive in HEK293T cells expressing endogenous p53, but active in p53-defective Saos-2 cells. Finally, a replication-competent IAV was successfully generated that multiplies only in p53-defective tumor cells and an IAV vector was developed that can deliver a foreign gene in cell type-specific manner. The method is flexible because the nanobody can be easily altered to target a different cell type, offering a valuable platform for virotherapy and biotechnology.
Collapse
Affiliation(s)
- Shinzi Ogasawara
- Department of Biology, Faculty of Science, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan.
| |
Collapse
|
3
|
Garcia G, Chakravarty N, Paiola S, Urena E, Gyani P, Tse C, French SW, Danielpour M, Breunig JJ, Nathanson DA, Arumugaswami V. Differential Susceptibility of Ex Vivo Primary Glioblastoma Tumors to Oncolytic Effect of Modified Zika Virus. Cells 2023; 12:2384. [PMID: 37830597 PMCID: PMC10572118 DOI: 10.3390/cells12192384] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/14/2023] Open
Abstract
Glioblastoma (GBM), the most common primary malignant brain tumor, is a highly lethal form of cancer with a very limited set of treatment options. High heterogeneity in the tumor cell population and the invasive nature of these cells decrease the likely efficacy of traditional cancer treatments, thus requiring research into novel treatment options. The use of oncolytic viruses as potential therapeutics has been researched for some time. Zika virus (ZIKV) has demonstrated oncotropism and oncolytic effects on GBM stem cells (GSCs). To address the need for safe and effective GBM treatments, we designed an attenuated ZIKV strain (ZOL-1) that does not cause paralytic or neurological diseases in mouse models compared with unmodified ZIKV. Importantly, we found that patient-derived GBM tumors exhibited susceptibility (responders) and non-susceptibility (non-responders) to ZOL-1-mediated tumor cell killing, as evidenced by differential apoptotic cell death and cell viability upon ZOL-1 treatment. The oncolytic effect observed in responder cells was seen both in vitro in neurosphere models and in vivo upon xenograft. Finally, we observed that the use of ZOL-1 as combination therapy with multiple PI3K-AKT inhibitors in non-responder GBM resulted in enhanced chemotherapeutic efficacy. Altogether, this study establishes ZOL-1 as a safe and effective treatment against GBM and provides a foundation to conduct further studies evaluating its potential as an effective adjuvant with other chemotherapies and kinase inhibitors.
Collapse
Affiliation(s)
- Gustavo Garcia
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA 90095, USA; (G.G.J.); (D.A.N.)
| | - Nikhil Chakravarty
- Department of Epidemiology, University of California Los Angeles, Los Angeles, CA 90095, USA;
| | - Sophia Paiola
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA 90095, USA; (G.G.J.); (D.A.N.)
| | - Estrella Urena
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA 90095, USA; (G.G.J.); (D.A.N.)
| | - Priya Gyani
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA 90095, USA; (G.G.J.); (D.A.N.)
| | - Christopher Tse
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA 90095, USA; (G.G.J.); (D.A.N.)
| | - Samuel W. French
- Department of Pathology and Laboratory Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA;
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Moise Danielpour
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (M.D.); (J.J.B.)
| | - Joshua J. Breunig
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; (M.D.); (J.J.B.)
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - David A. Nathanson
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA 90095, USA; (G.G.J.); (D.A.N.)
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Vaithilingaraja Arumugaswami
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA 90095, USA; (G.G.J.); (D.A.N.)
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
- California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
4
|
You L, Liu Y, Chen N, Zhu L, Xu G, Lv Z, Zhou Y, Li C, Tong H, Jin J, Meng H. Influenza A (H1N1) virus induced long-term remission in a refractory acute myeloid leukaemia. Br J Haematol 2023; 202:745-748. [PMID: 37230766 DOI: 10.1111/bjh.18876] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 05/01/2023] [Accepted: 05/09/2023] [Indexed: 05/27/2023]
Abstract
There have been reports of haematological cancer patients achieving spontaneous remission after being infected with the influenza A or SARS-COV-2 virus. Here, we present the first case of long-term complete remission (CR) induced by influenza A (IAV, H1N1 subtype) in a refractory AML patient and have functionally validated this finding in two different animal disease models. We observed a significant increase in the proportion of helper T cells in the patient after IAV infection. The levels of cytokines, including IL-2, IL-4, IL-6, IL-10, IL-17A, IFN-γ and TNF-α, were higher in IAV-infected patients compared with control groups. These findings indicate that the anti-tumour effects induced by IAV are closely related to the modification of the immune response. Our study provides new evidence of the anti-tumour effects of IAV from a clinical practice perspective.
Collapse
Affiliation(s)
- Liangshun You
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
- Zhejiang Province Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou, Zhejiang, P.R. China
| | - Yi Liu
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Nianci Chen
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Li Zhu
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Gaixiang Xu
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
- Zhejiang Province Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou, Zhejiang, P.R. China
| | - Zuopo Lv
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Yile Zhou
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
- Zhejiang Province Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou, Zhejiang, P.R. China
| | - Chenying Li
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
- Zhejiang Province Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou, Zhejiang, P.R. China
| | - Hongyan Tong
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
- Zhejiang Province Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou, Zhejiang, P.R. China
| | - Jie Jin
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
- Zhejiang Province Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou, Zhejiang, P.R. China
| | - Haitao Meng
- Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
- Zhejiang Province Key Laboratory of Hematopoietic Malignancy, Zhejiang University, Hangzhou, Zhejiang, P.R. China
- Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou, Zhejiang, P.R. China
| |
Collapse
|
5
|
Fang C, Xiao G, Wang T, Song L, Peng B, Xu B, Zhang K. Emerging Nano-/Biotechnology Drives Oncolytic Virus-Activated and Combined Cancer Immunotherapy. RESEARCH 2023; 6:0108. [PMID: 37040283 PMCID: PMC10079287 DOI: 10.34133/research.0108] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 03/15/2023] [Indexed: 04/05/2023]
Abstract
Oncolytic viruses (OVs) as one promising antitumor methods have made important contributions to tumor immunotherapy, which arouse increasing attention. They provide the dual mechanisms including direct killing effect toward tumor cells and immune activation for elevating antitumor responses, which have been proved in many preclinical studies. Especially, natural or genetically modified viruses as clinical immune preparations have emerged as a new promising approach objective to oncology treatment. The approval of talimogene laherparepvec (T-VEC) by the U.S. Food and Drug Administration (FDA) for the therapy of advanced melanoma could be considered as a milestone achievement in the clinical translation of OV. In this review, we first discussed the antitumor mechanisms of OVs with an emphasis on targeting, replication, and propagation. We further outlined the state of the art of current OVs in tumor and underlined the activated biological effects especially including immunity. More significantly, the enhanced immune responses based on OVs were systematically discussed from different perspectives such as combination with immunotherapy, genetic engineering of OVs, integration with nanobiotechnology or nanoparticles, and antiviral response counteraction, where their principles were shed light on. The development of OVs in the clinics was also highlighted to analyze the actuality and concerns of different OV applications in clinical trials. At last, the future perspectives and challenges of OVs as an already widely accepted treatment means were discussed. This review will provide a systematic review and deep insight into OV development and also offer new opportunities and guidance pathways to drive the further clinical translation.
Collapse
Affiliation(s)
- Chao Fang
- Central Laboratory and Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine,
Tongji University, No. 301 Yan-chang-zhong Road, Shanghai 200072, China
| | - Gaozhe Xiao
- National Center for International Research of Bio-targeting Theranostics,
Guangxi Medical University, No. 22 Shuangyong Road 22, Nanning, Guangxi 530021, China
| | - Taixia Wang
- Central Laboratory and Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine,
Tongji University, No. 301 Yan-chang-zhong Road, Shanghai 200072, China
| | - Li Song
- Central Laboratory and Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine,
Tongji University, No. 301 Yan-chang-zhong Road, Shanghai 200072, China
| | - Bo Peng
- Central Laboratory and Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine,
Tongji University, No. 301 Yan-chang-zhong Road, Shanghai 200072, China
| | - Bin Xu
- Department of Urology, Shanghai Ninth People’s Hospital,
Shanghai Jiaotong University School of Medicine, No. 639 Zhizaoju Road, Huangpu, Shanghai 200011, China
| | - Kun Zhang
- Central Laboratory and Department of Urology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine,
Tongji University, No. 301 Yan-chang-zhong Road, Shanghai 200072, China
- National Center for International Research of Bio-targeting Theranostics,
Guangxi Medical University, No. 22 Shuangyong Road 22, Nanning, Guangxi 530021, China
| |
Collapse
|
6
|
Li S, Li Q, Ren Y, Yi J, Guo J, Kong X. HSV: The scout and assault for digestive system tumors. Front Mol Biosci 2023; 10:1142498. [PMID: 36926680 PMCID: PMC10011716 DOI: 10.3389/fmolb.2023.1142498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 02/15/2023] [Indexed: 03/08/2023] Open
Abstract
More than 25% of all malignant tumors are digestive system tumors (DSTs), which mostly include esophageal cancer, gastric cancer, pancreatic cancer, liver cancer, gallbladder cancer and cholangiocarcinoma, and colorectal cancer. DSTs have emerged as one of the prominent reasons of morbidity and death in many nations and areas around the world, posing a serious threat to human life and health. General treatments such as radiotherapy, chemotherapy, and surgical resection can poorly cure the patients and have a bad prognosis. A type of immunotherapy known as oncolytic virus therapy, have recently shown extraordinary anti-tumor effectiveness. One of the viruses that has been the subject of the greatest research in this field, the herpes simplex virus (HSV), has shown excellent potential in DSTs. With a discussion of HSV-1 based on recent studies, we outline the therapeutic effects of HSV on a number of DSTs in this review. Additionally, the critical function of HSV in the detection of cancers is discussed, and some HSV future possibilities are shown.
Collapse
Affiliation(s)
- Sheng Li
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qingbo Li
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yi Ren
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jia Yi
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jinhe Guo
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xianbin Kong
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
7
|
Gospel of malignant Glioma: Oncolytic virus therapy. Gene 2022; 818:146217. [PMID: 35093451 DOI: 10.1016/j.gene.2022.146217] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 12/09/2021] [Accepted: 01/13/2022] [Indexed: 12/12/2022]
Abstract
Glioma accounts for nearly 80% of all intracranial malignant tumors. It is a major challenge to society as it is causes to impaired brain function in many patients. Currently, gliomas are mainly treated with surgery, postoperative radiotherapy, and chemotherapy. However, the curative effects of these treatments are not satisfactory. Oncolytic virus (OV) is a novel treatment which works by activating the immune functions and inducing apoptosis of tumor cells. The OV propagates indefinitely in the host cell, eventually leading to the death of host cell. Subsequently, a large number of antigens and signal molecules are released which exert antitumor immunity. Several preclinical and clinical studies have shown that G207, DNX2401, Zika and other viruses have important roles in malignant tumors. For example, these viruses can reduce the growth of tumor cells without causing severe complications. However, the known OVs have not been clearly classified. Herein, we divided OVs into neurotropic and non-neurophilic OVs based on whether the OVs are naturally neurotropic or not. The therapeutic effects of each group were compared. Finally, challenges encountered in the clinical application of OVs in the treatment of malignant gliomas were summarized.
Collapse
|
8
|
Cai J, Yan G. The Identification and Development of a Novel Oncolytic Virus: Alphavirus M1. Hum Gene Ther 2021; 32:138-149. [PMID: 33261513 DOI: 10.1089/hum.2020.271] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Oncolytic virotherapy represents an ideal therapeutic platform for cancer in which natural or engineered viruses selectively replicate in and destroy tumor cells, whereas sparing normal cells. Oncolytic virotherapy is considered as a key contributor in modern immunotherapy. However, several challenges remain with regard to exploiting the potential of oncolytic viruses, such as the lack of biomarkers for precise treatment, the difficulty of systemic administration because of pre-existing neutralizing antibodies to popular oncolytic viral vectors in human serum and the lack of mature lyophilization technology for convenient transport and preservation of viral preparations. The M1 strain, which was isolated on Hainan Island of China in the 1960s, is a member of the alphavirus genus Togaviridae family. It was identified as a novel oncolytic virus in 2014. During the development of M1 virus, many challenges have been overcome: several biomarkers have been identified for precise treatment; systematic administration of M1 is suitable and feasible because of the extremely low percentage of pre-existing neutralizing antibodies in the general population, and a lyophilized powder that maintains high biological stability has been developed. This review provides an encyclopedia of studies supporting M1 as an oncolytic virus, including the biological characteristics, tumor selectivity and its mechanism, tumor killing mechanism, combination therapy, and nonclinical pharmacokinetics of M1 virus. The future development direction of oncolytic virus M1 is also discussed at the end of the review.
Collapse
Affiliation(s)
- Jing Cai
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Guangmei Yan
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
9
|
Liu S, Zhang J, Fang S, Zhang Q, Zhu G, Tian Y, Zhao M, Liu F. Macrophage polarization contributes to the efficacy of an oncolytic HSV-1 targeting human uveal melanoma in a murine xenograft model. Exp Eye Res 2021; 202:108285. [PMID: 33039456 DOI: 10.1016/j.exer.2020.108285] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 09/04/2020] [Accepted: 09/29/2020] [Indexed: 12/28/2022]
Abstract
Uveal melanoma (UM), the most common primary malignant tumor of the eye in adults, is difficult-to-treat. UM has a relatively high mortality secondary to distant metastasis and poor overall survival with existing therapies. The oncolytic virus herpes simplex virus type-1 (HSV-1) has been approved for clinical use in melanoma. This double-stranded DNA virus was suspected to directly activate lysis specifically in neoplastic cells. We tested the antitumor efficacy of recombinant oncolytic HSV-1-EGFP (oHSV-EGFP) in UM and characterized the local and systemic antitumor innate immune response in a murine xenograft model. We first determined the efficacy of the oncolytic virus in 92.1, MUM2B and MP41 cell lines. In murine xenograft models, oHSV-EGFP reduced intraocular tumors as well as systemic subcutaneous tumors. A significant change in cytokines was observed in viral infected cells, especially a rise in IFNγ. In vivo analyses showed increased anti-tumorigenic M1 macrophages and decreased pro-tumorigenic M2 macrophages in peripheral blood, and intraocular and distant tumors after intravitreal viral treatment. Increased infiltration of natural killer cells and mature dendritic cells was also detected after viral treatment. In addition, no virus was detected in major organs after the treatment. Our data support that oHSV-EGFP is effective, neoplasm specific, immune active and safe, providing possible clinical translatable options to treat ocular and metastatic UM.
Collapse
Affiliation(s)
- Sisi Liu
- Department of Ophthalmology, Peking University People's Hospital, Eye Diseases and Optometry Institute, Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center, Beijing, China
| | - Junwen Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Sheng Fang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Qing Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Guidong Zhu
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Yifu Tian
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing Laboratory of Biomedical Materials, Beijing, China
| | - Mingwei Zhao
- Department of Ophthalmology, Peking University People's Hospital, Eye Diseases and Optometry Institute, Beijing Key Laboratory of Diagnosis and Therapy of Retinal and Choroid Diseases, College of Optometry, Peking University Health Science Center, Beijing, China.
| | - Fusheng Liu
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Department of Neurosurgery, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing Laboratory of Biomedical Materials, Beijing, China.
| |
Collapse
|
10
|
Niedźwiedzka-Rystwej P, Grywalska E, Hrynkiewicz R, Wołącewicz M, Becht R, Roliński J. The Double-Edged Sword Role of Viruses in Gastric Cancer. Cancers (Basel) 2020; 12:cancers12061680. [PMID: 32599870 PMCID: PMC7352989 DOI: 10.3390/cancers12061680] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/14/2020] [Accepted: 06/23/2020] [Indexed: 02/06/2023] Open
Abstract
Due to its high morbidity and mortality, gastric cancer is a topic of a great concern throughout the world. Major ways of treatment are gastrectomy and chemotherapy, unfortunately they are not always successful. In a search for more efficient therapy strategies, viruses and their potential seem to be an important issue. On one hand, several oncogenic viruses have been noticed in the case of gastric cancer, making the positive treatment even more advantageous, but on the other, viruses exist with a potential therapeutic role in this malignancy.
Collapse
Affiliation(s)
- Paulina Niedźwiedzka-Rystwej
- Institute of Biology, University of Szczecin, Felczaka 3c, 71-412 Szczecin, Poland; (R.H.); (M.W.)
- Correspondence:
| | - Ewelina Grywalska
- Department of Clinical Immunology and Immunotherapy, Medical University of Lublin, 20-093 Lublin, Poland; (E.G.); (J.R.)
| | - Rafał Hrynkiewicz
- Institute of Biology, University of Szczecin, Felczaka 3c, 71-412 Szczecin, Poland; (R.H.); (M.W.)
| | - Mikołaj Wołącewicz
- Institute of Biology, University of Szczecin, Felczaka 3c, 71-412 Szczecin, Poland; (R.H.); (M.W.)
| | - Rafał Becht
- Clinical Department of Oncology, Chemotherapy and Cancer Immunotherapy, Pomeranian Medical University of Szczecin, 70-204 Szczecin, Poland;
| | - Jacek Roliński
- Department of Clinical Immunology and Immunotherapy, Medical University of Lublin, 20-093 Lublin, Poland; (E.G.); (J.R.)
| |
Collapse
|
11
|
Gray Z, Tabarraei A, Moradi A, Kalani MR. M51R and Delta-M51 matrix protein of the vesicular stomatitis virus induce apoptosis in colorectal cancer cells. Mol Biol Rep 2019; 46:3371-3379. [PMID: 31006094 DOI: 10.1007/s11033-019-04799-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 04/05/2019] [Indexed: 12/17/2022]
Abstract
Colorectal cancer (CRC) is the third most common cancer in both men and women. Oncolytic viral-based therapy methods seem to be promising for CRC treatment. Vesicular stomatitis virus (VSV) is considered as a potent candidate in viral therapy for several tumors. VSV particles with mutated matrix (M) protein are capable of initiating cell death cascades while not being harmful to the immune system. In the current study, the effects of the VSV M-protein was investigated on the apoptosis of the colorectal cancer SW480 cell. Wild-type, M51R, and ΔM51 mutants VSV M-protein genes were cloned into the PCDNA3.1 vector and transfected into the SW480 cells. The results of the MTT assay, Western blotting, and Caspase 3, 8, and 9 measurement, illustrated that both wild and M51R mutant M-proteins can destroy the SW480 colorectal cancer cells. DAPI/TUNEL double-staining reconfirmed the apoptotic effects of the M-protein expression. The ΔM51 mutant M-protein is effective likewise M51R, somehow it can be considered as a safer substitution.
Collapse
Affiliation(s)
- Zahra Gray
- Department of Microbiology, College of Medicine, Golestan University of Medical Science, 1 Shastcola Ave, 5 km Sari Rd, Gorgan, Iran
| | - Alijan Tabarraei
- Department of Microbiology, College of Medicine, Golestan University of Medical Science, 1 Shastcola Ave, 5 km Sari Rd, Gorgan, Iran
| | - Abdolvahab Moradi
- Department of Microbiology, College of Medicine, Golestan University of Medical Science, 1 Shastcola Ave, 5 km Sari Rd, Gorgan, Iran.
| | - Mohamad R Kalani
- Cell and Molecular Research center, Golestan University of Medical Science, 1 Shastcola Ave, 5 km Sari Rd, Gorgan, Iran. .,Molecular and Cell Biology, RAL, University of Illinois at Urbana-Champaign, 600 S Goodwin Ave #325, Urbana, IL, 61801, USA.
| |
Collapse
|
12
|
A Recombinant Antibody-Expressing Influenza Virus Delays Tumor Growth in a Mouse Model. Cell Rep 2019; 22:1-7. [PMID: 29298413 DOI: 10.1016/j.celrep.2017.12.025] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 11/09/2017] [Accepted: 12/06/2017] [Indexed: 12/27/2022] Open
Abstract
Influenza A virus (IAV) has shown promise as an oncolytic agent. To improve IAV as an oncolytic virus, we sought to design a transgenic virus expressing an immune checkpoint-inhibiting antibody during the viral life cycle. To test whether it was possible to express an antibody during infection, an influenza virus was constructed encoding the heavy chain of an antibody on the PB1 segment and the light chain of an antibody on the PA segment. This antibody-expressing IAV grows to high titers, and the antibodies secreted from infected cells exhibit comparable functionality with hybridoma-produced antibodies. To enhance the anti-cancer activity of IAV, an influenza virus was engineered to express a single-chain antibody antagonizing the immune checkpoint CTLA4 (IAV-CTLA4). In mice implanted with the aggressive B16-F10 melanoma, intratumoral injection with IAV-CTLA4 delayed the growth of treated tumors, mediated an abscopal effect, and increased overall survival.
Collapse
|
13
|
Song J, Zhang F, Ji J, Chen M, Li Q, Weng Q, Gu S, Kogut MJ, Yang X. Orthotopic hepatocellular carcinoma: molecular imaging-monitored intratumoral hyperthermia-enhanced direct oncolytic virotherapy. Int J Hyperthermia 2019; 36:344-350. [PMID: 30776922 PMCID: PMC6988576 DOI: 10.1080/02656736.2019.1569731] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Objective: To validate the feasibility of molecular imaging-monitored intratumoral radiofrequency hyperthermia (RFH) enhanced direct oncolytic virotherapy for hepatocellular carcinoma (HCC). Methods: This study included in vitro experiments using luciferase-labeled rat HCC cells and in vivo validation experiments on rat models with orthotopic HCCs. Both cells and HCCs in four groups (n = 6/group) were treated by: (1) combination therapy of oncolytic virotherapy (T-VEC) plus RFH at 42 °C for 30 min; (2) oncolytic virotherapy alone; (3) RFH alone; and (4) saline. For in vitro confirmation, confocal microscopy and bioluminescence optical imaging were used to evaluate the cell viabilities. For in vivo validation, oncolytic viruses were directly infused into rat HCCs through a multi-functional perfusion-thermal RF electrode, followed by RFH. Ultrasound and optical imaging were used to follow up size and bioluminescence signal changes of tumors overtime, which were correlated with subsequent laboratory examinations. Results: For in vitro experiments, confocal microscopy showed the lowest number of viable cells, as well as a significant decrease of bioluminescence signal intensity of cells with combination therapy group, compared to other three groups (p < .001). For in vivo experiments, ultrasound and optical imaging showed the smallest tumor volume, and significantly decreased bioluminescence signal intensity in combination therapy group compared to other three groups (p < .05), which were well correlated with pathologic analysis. Conclusion: It is feasible of using molecular imaging to guide RFH-enhanced intratumoral oncolytic virotherapy of HCC, which may open new avenues to prevent residual or recurrent disease of thermally ablated intermediate-to-large HCCs.
Collapse
Affiliation(s)
- Jingjing Song
- a Image-Guided Bio-Molecular Interventions Section, Division of Interventional Radiology, Department of Radiology , University of Washington School of Medicine , Seattle , WA , USA.,b Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Affiliated Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital , Zhejiang , China
| | - Feng Zhang
- a Image-Guided Bio-Molecular Interventions Section, Division of Interventional Radiology, Department of Radiology , University of Washington School of Medicine , Seattle , WA , USA
| | - Jiansong Ji
- b Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Affiliated Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital , Zhejiang , China
| | - Minjiang Chen
- b Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Affiliated Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital , Zhejiang , China
| | - Qiang Li
- c Department of Radiology , Yinzhou People's Hospital Ningbo , Ningbo , Zhejiang , China
| | - Qiaoyou Weng
- b Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Affiliated Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital , Zhejiang , China
| | - Shannon Gu
- a Image-Guided Bio-Molecular Interventions Section, Division of Interventional Radiology, Department of Radiology , University of Washington School of Medicine , Seattle , WA , USA
| | - Matthew J Kogut
- a Image-Guided Bio-Molecular Interventions Section, Division of Interventional Radiology, Department of Radiology , University of Washington School of Medicine , Seattle , WA , USA
| | - Xiaoming Yang
- a Image-Guided Bio-Molecular Interventions Section, Division of Interventional Radiology, Department of Radiology , University of Washington School of Medicine , Seattle , WA , USA.,d Department of Radiology , Sir Run Run Shaw Hospital, Zhejiang University School of Medicine , Hangzhou , Zhejiang , China
| |
Collapse
|
14
|
Abstract
Convection-enhanced delivery (CED) is a promising technique that generates a pressure gradient at the tip of an infusion catheter to deliver therapeutics directly through the interstitial spaces of the central nervous system. It addresses and offers solutions to many limitations of conventional techniques, allowing for delivery past the blood-brain barrier in a targeted and safe manner that can achieve therapeutic drug concentrations. CED is a broadly applicable technique that can be used to deliver a variety of therapeutic compounds for a diversity of diseases, including malignant gliomas, Parkinson's disease, and Alzheimer's disease. While a number of technological advances have been made since its development in the early 1990s, clinical trials with CED have been largely unsuccessful, and have illuminated a number of parameters that still need to be addressed for successful clinical application. This review addresses the physical principles behind CED, limitations in the technique, as well as means to overcome these limitations, clinical trials that have been performed, and future developments.
Collapse
Affiliation(s)
- A M Mehta
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY, 10032, USA
| | - A M Sonabend
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY, 10032, USA
| | - J N Bruce
- Department of Neurological Surgery, Columbia University Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
15
|
Abstract
Oncolytic viruses represent a new class of therapeutic agents that promote anti-tumour responses through a dual mechanism of action that is dependent on selective tumour cell killing and the induction of systemic anti-tumour immunity. The molecular and cellular mechanisms of action are not fully elucidated but are likely to depend on viral replication within transformed cells, induction of primary cell death, interaction with tumour cell antiviral elements and initiation of innate and adaptive anti-tumour immunity. A variety of native and genetically modified viruses have been developed as oncolytic agents, and the approval of the first oncolytic virus by the US Food and Drug Administration (FDA) is anticipated in the near future. This Review provides a comprehensive overview of the basic biology supporting oncolytic viruses as cancer therapeutic agents, describes oncolytic viruses in advanced clinical trials and discusses the unique challenges in the development of oncolytic viruses as a new class of drugs for the treatment of cancer.
Collapse
Affiliation(s)
- Howard L. Kaufman
- Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, Room 2004, New Brunswick, 08901 New Jersey USA
| | - Frederick J. Kohlhapp
- Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, Room 2004, New Brunswick, 08901 New Jersey USA
| | - Andrew Zloza
- Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, Room 2004, New Brunswick, 08901 New Jersey USA
| |
Collapse
|
16
|
Brader P, Wong RJ, Horowitz G, Gil Z. Combination of pet imaging with viral vectors for identification of cancer metastases. Adv Drug Deliv Rev 2012; 64:749-55. [PMID: 21565234 DOI: 10.1016/j.addr.2011.04.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2011] [Revised: 04/22/2011] [Accepted: 04/24/2011] [Indexed: 02/05/2023]
Abstract
There are three main ways for dissemination of solid tumors: direct invasion, lymphatic spread and hematogenic spread. The presence of metastases is the most significant factor in predicting prognosis and therefore evidence of metastases will influence decision-making regarding treatment. Conventional imaging techniques are limited in the evaluation and localization of metastases due to their restricted ability to identify subcentimeter neoplastic disease. Hence, there is a need for an effective noninvasive modality that can accurately identify occult metastases in cancer patients. One such method is the combination of positron emission tomography (PET) with vectors designed for delivery of reporter genes into target cells. Vectors expressing the herpes simplex virus-1 thymidine kinase (HSV1-tk) reporter system have recently been shown to allow localization of micrometastases in animal models of cancer using non invasive imaging. Combination of HSV1-tk and PET imaging is based on the virtues of vectors which can carry and selectively express the HSV1-tk reporter gene in a variety of cancer cells but not in normal tissue. A radioactive tracer which is applied systemically is phosphorylated by the HSV1-tk enzyme, and as a consequence, the tracer accumulates in proportion to the level of HSV1-tk expression which can be imaged by PET. In this paper we review the recent developments in molecular imaging of micrometastases using replication-competent viral or nonviral vectors carrying the HSV1-tk gene using PET imaging. These diagnostic paradigms introduce an advantageous new concept in noninvasive molecular imaging with the potential benefits for improving patient care by providing guidance for therapy to patients with risk for metastases.
Collapse
Affiliation(s)
- Peter Brader
- Molecular and Gender Imaging, Universitätsklinik für Radiologie, Medical University Vienna, General Hospital Vienna, Austria
| | | | | | | |
Collapse
|
17
|
Brader P, Kelly K, Gang S, Shah JP, Wong RJ, Hricak H, Blasberg RG, Fong Y, Gil Z. Imaging of lymph node micrometastases using an oncolytic herpes virus and [F]FEAU PET. PLoS One 2009; 4:e4789. [PMID: 19274083 PMCID: PMC2651472 DOI: 10.1371/journal.pone.0004789] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2008] [Accepted: 02/02/2009] [Indexed: 12/11/2022] Open
Abstract
Background In patients with melanoma, knowledge of regional lymph node status provides important information on outlook. Since lymph node status can influence treatment, surgery for sentinel lymph node (SLN) biopsy became a standard staging procedure for these patients. Current imaging modalities have a limited sensitivity for detection of micrometastases in lymph nodes and, therefore, there is a need for a better technique that can accurately identify occult SLN metastases. Methodology/Principal Findings B16-F10 murine melanoma cells were infected with replication-competent herpes simplex virus (HSV) NV1023. The presence of tumor-targeting and reporter-expressing virus was assessed by [18F]-2′-fluoro-2′-deoxy-1-β-D-β-arabinofuranosyl-5-ethyluracil ([18F]FEAU) positron emission tomography (PET) and confirmed by histochemical assays. An animal foot pad model of melanoma lymph node metastasis was established. Mice received intratumoral injections of NV1023, and 48 hours later were imaged after i.v. injection of [18F]FEAU. NV1023 successfully infected and provided high levels of lacZ transgene expression in melanoma cells. Intratumoral injection of NV1023 resulted in viral trafficking to melanoma cells that had metastasized to popliteal and inguinal lymph nodes. Presence of virus-infected tumor cells was successfully imaged with [18F]FEAU-PET, that identified 8 out of 8 tumor-positive nodes. There was no overlap between radioactivity levels (lymph node to surrounding tissue ratio) of tumor-positive and tumor-negative lymph nodes. Conclusion/Significance A new approach for imaging SLN metastases using NV1023 and [18F]FEAU-PET was successful in a murine model. Similar studies could be translated to the clinic and improve the staging and management of patients with melanoma.
Collapse
Affiliation(s)
- Peter Brader
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- Department of Radiology, Medical University Graz, Graz, Austria
| | - Kaitlyn Kelly
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Sheng Gang
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Jatin P. Shah
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Richard J. Wong
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Hedvig Hricak
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Ronald G. Blasberg
- Department of Neurology, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Yuman Fong
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
| | - Ziv Gil
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, United States of America
- The Laboratory for Applied Cancer Research, Tel Aviv Sourasky Medical Center, Tel Aviv University, Tel Aviv, Israel
- * E-mail:
| |
Collapse
|
18
|
|
19
|
Smith KD, Shao MY, Posner MC, Weichselbaum RR. Tumor genotype determines susceptibility to oncolytic herpes simplex virus mutants: strategies for clinical application. Future Oncol 2007; 3:545-56. [DOI: 10.2217/14796694.3.5.545] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Oncolytic Herpes simplex virus -1 (HSV-1) mutants based on deletion of the γ134.5 gene are promising therapies for cancer. Δγ134.5 mutant replication and cytolysis is tumor cell type specific and severely attenuated in normal tissues. The basis for attenuation lies in the activation of the protein kinase R (PKR)-mediated host cellular defense pathway, which inhibits protein synthesis in infected cells. Tumor cells which overexpress MAPK kinase (MEK) activity support robust replication of Δγ134.5 mutants via MEK-mediated inhibition of PKR, resulting in tumor oncolysis. Systemic delivery of γ134.5 mutants may allow selective targeting and destruction of metastases from a broad range of solid human tumors that overexpress MEK. Barriers to systemic HSV-1 oncolytic therapy include innate immunity, adaptive immunity and hepatic adsorption. Immunomodulating agents may overcome innate immunity to HSV-1-based vectors. Preclinical data combined with the pervasiveness of HSV-1 despite widespread immunity suggest that preexisting immunity may not eliminate oncolytic efficacy. In the future, biopsy-determined tumor MEK status may select patients for Δγ134.5 oncolytic therapy.
Collapse
Affiliation(s)
- Kerrington D Smith
- MD Anderson Cancer Center, Department of Surgical Oncology, 1515 Holcombe Blvd. Unit 444, Houston TX 77030, USA
| | - Michael Y Shao
- University of Chicago Medical Center, Department of General Surgery, 5841 S. Maryland Avenue, MC 6040, Chicago, IL 60637, USA
| | - Mitchell C Posner
- University of Chicago Medical Center, 5841 S. Maryland Avenue, MC 5031, Chicago, IL 60637, USA
| | - Ralph R Weichselbaum
- Center for Advanced Medicine 1338, Department of Radiation & Cellular Oncology, 5758 S. Maryland Avenue, MC 9006, Chicago, IL 60637, USA
| |
Collapse
|
20
|
Yu Z, Adusumilli PS, Eisenberg DP, Darr E, Ghossein RA, Li S, Liu S, Singh B, Shah JP, Fong Y, Wong RJ. Nectin-1 Expression by Squamous Cell Carcinoma is a Predictor of Herpes Oncolytic Sensitivity. Mol Ther 2007; 15:103-13. [PMID: 17164781 DOI: 10.1038/sj.mt.6300009] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Oncolytic viruses based on herpes simplex virus type 1 (HSV-1) are able to infect and lyse a variety of malignant cell lines. However, there is variability in the degree of tumor susceptibility, and the cancer cell determinants of HSV sensitivity are poorly defined. Nectin-1 is a cell surface adhesion molecule that functions as a cellular receptor to HSV envelope glycoprotein D (gD). We assessed tumor nectin-1 expression as a predictor of oncolytic HSV sensitivity. A panel of human squamous carcinoma cell lines was evaluated for viral entry, replication, and cytotoxicity to an attenuated, replication-competent, oncolytic HSV (NV1023). Potential tumor determinants of HSV sensitivity were assessed, including nectin-1, herpes viral entry mediator, total gD receptor expression, S-phase fraction, and doubling time. Significant correlations between nectin-1 expression measured by quantitative fluorescence-activated cell sorting and viral sensitivity measures were identified using Pearson's coefficients. Cancer cell nectin-1 receptor blockade and nectin-1 transfection led to inhibition and enhancement of NV1023 viral entry, respectively. Cell lines with varying nectin-1 expression showed corresponding sensitivity to NV1023 therapy in vivo. Immunohistochemistry for nectin-1 was inversely related to E-cadherin staining, suggesting increased herpes sensitivity of E-cadherin-deficient tumors. These results suggest that nectin-1 may be used as a marker to predict the sensitivity of a tumor to herpes oncolytic therapy.
Collapse
MESH Headings
- Animals
- Cadherins/metabolism
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/therapy
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/metabolism
- Cell Line, Tumor
- Cell Transformation, Neoplastic
- Cricetinae
- Gene Expression Regulation, Neoplastic/genetics
- Genetic Therapy
- Herpesvirus 1, Human/physiology
- Humans
- Immunohistochemistry
- Mice
- Mice, Nude
- Nectins
- Oncolytic Viruses/physiology
- Receptors, Tumor Necrosis Factor, Member 14/metabolism
- S Phase
- Sensitivity and Specificity
- Transgenes/genetics
- Viral Envelope Proteins/metabolism
- Virus Internalization
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Zhenkun Yu
- Head and Neck Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Yu Z, Chan MK, O-charoenrat P, Eisenberg DP, Shah JP, Singh B, Fong Y, Wong RJ. Enhanced nectin-1 expression and herpes oncolytic sensitivity in highly migratory and invasive carcinoma. Clin Cancer Res 2005; 11:4889-97. [PMID: 16000587 DOI: 10.1158/1078-0432.ccr-05-0309] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Although a variety of malignant tumors are susceptible to therapy with oncolytic herpes simplex viruses, the determinants of tumor sensitivity to these viruses are poorly understood. Nectin-1 is a cell surface adhesion molecule that is a component of intercellular adherens junctions and also functions as a herpes viral receptor. Because highly invasive cells may have decreased intercellular adhesion, we sought to determine if such cells might also have altered availability of cell surface nectin-1 to act as a herpes receptor. EXPERIMENTAL DESIGN AND RESULTS A series of squamous cell carcinoma lines of increasing migratory and invasive potential, termed MG1-MG14, were selected by serial passages of murine SCC7 through Matrigel invasion chambers. Available cell surface nectin-1 was enhanced on the MG11 and MG14 cell lines in comparison to SCC7 as measured by cellular ELISA and immunofluorescence microscopy. A replication-competent, oncolytic herpes virus (NV1023) showed an increased ability to enter MG11 and MG14 cells as compared with SCC7 cells. Furthermore, MG11 and MG14 supported increased herpes viral replication and cytotoxicity over SCC7. For all three of the cell lines, viral entry assays revealed that the actively migrating cells were significantly more susceptible to herpes infection than the nonmigrating cells. CONCLUSIONS These results show that malignant cells with highly migratory and invasive properties may exhibit increased cell surface nectin-1 availability, which may serve as a herpes viral receptor to enhance the efficacy of herpes oncolytic therapy. This finding has implications regarding patient selection for future clinical trials using these promising therapeutic vectors.
Collapse
Affiliation(s)
- Zhenkun Yu
- Head and Neck Service and Hepatobiliary Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, New York 10021, USA
| | | | | | | | | | | | | | | |
Collapse
|
22
|
|
23
|
|
24
|
COLLIER WA. [The cultivation of yellow fever virus 17D on ascites of the Krebs-2-carcinoma]. Antonie van Leeuwenhoek 1960; 26:209-23. [PMID: 13811248 DOI: 10.1007/bf02539006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
25
|
PIERCE M, CARTER RE, RIGOR E. Studies on the relationship of viral infection to leukemia in mice.I. A comparison of the effects of lethal and sublethal infections due to egypt 101 virus on transmissible leukemia in mice. Cancer 1959; 12:222-31. [PMID: 13638940 DOI: 10.1002/1097-0142(195903/04)12:2<222::aid-cncr2820120204>3.0.co;2-n] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
26
|
|
27
|
|
28
|
|
29
|
|
30
|
KUWATA T. Multiplication of
Rickettsia tsutsugamushi
and Ornithosis Virus in Transplantable Mouse Tumors. Science 1951; 114:640-1. [PMID: 14913128 DOI: 10.1126/science.114.2972.640] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
31
|
|
32
|
|