1
|
Lundstrom K. Viral Vector-Based Cancer Vaccines. Methods Mol Biol 2025; 2926:101-127. [PMID: 40266521 DOI: 10.1007/978-1-0716-4542-0_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Viral vectors have been frequently used as vaccine-delivery vehicles. Both DNA and RNA viruses have been employed for vaccine development. Viral vectors based on adenoviruses (Ad), adeno-associated viruses (AAV), herpes simplex viruses (HSV), lentiviruses (LV), alphaviruses, flaviviruses, measles viruses (MV), rhabdoviruses, Newcastle disease virus (NDV), poxviruses and picornaviruses have been utilized. Approaches have included the expression of tumor-associated antigens and immunostimulatory genes as well as administration of oncolytic viruses. Prophylactic and therapeutic proof-of-concept has been established in preclinical animal tumor models, and therapeutic efficacy has been obtained in clinical trials in human cancer patients. Model viral vector systems and their applications in cancer vaccine development are described here.
Collapse
|
2
|
Forden CA. Phagolysosomal resistance hypothesized to be a danger signal. Scand J Immunol 2024; 100:e13400. [PMID: 39138895 DOI: 10.1111/sji.13400] [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: 01/02/2024] [Revised: 07/09/2024] [Accepted: 07/19/2024] [Indexed: 08/15/2024]
Abstract
Antigen presenting cells sometimes require T cell "help" to kill and decompose microbes they capture, especially when those microbes resist effector molecules including nitric oxide and reactive oxygen species. Pathogens are more likely to resist those effectors, shared by the innate and adaptive immune systems, than are commensals. Does such resistance alert the immune system to the danger posed by those pathogens? Several lines of evidence suggest this occurs. Mouse studies showed a surprising exacerbation, not alleviation of experimental autoimmune encephalomyelitis, by suppression of nitric oxide production, but only when the suppression was applied to animals undergoing vaccination with myelin. In contrast, animals receiving T cells activated by vaccination without suppression of nitric oxide benefitted from reduced autoimmune cytotoxicity when nitric oxide production was suppressed after adoptive transfer. Vaccinia and adenovirus suppress nitric oxide production and have been successful vaccine platforms, also consistent with the above phagolysosomal resistance hypothesis. The hypothesis solves a long-standing quandary-how can nitric oxide protect against both infection and autoimmunity, especially autoimmune diseases for which it seems a major effector? The importance of physical linkage between epitopes, first proposed in Bretscher's Two-Step, Two-Signal theory dependent on B cells, is extended to include phagolysosomal resistance in general, plus a corollary proposition that the immune system detects resistance to dissociation of high-affinity pathogenic ligands from host binding sites to make neutralizing antibodies.
Collapse
|
3
|
Letafati A, Ardekani OS, Naderisemiromi M, Fazeli MM, Jemezghani NA, Yavarian J. Oncolytic viruses against cancer, promising or delusion? Med Oncol 2023; 40:246. [PMID: 37458862 DOI: 10.1007/s12032-023-02106-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 06/23/2023] [Indexed: 07/20/2023]
Abstract
Cancer treatment is one of the most challenging topics in medical sciences. Different methods such as chemotherapy, tumor surgery, and immune checkpoint inhibitors therapy (ICIs) are potential approaches to treating cancer and killing tumor cells, but clinical studies have shown that they have been successful for a limited group of patients. Using viruses as a treatment can be considered as an effective treatment in the field of medicine. This is considered as a potential treatment, especially in comparison to chemotherapy, which has severe side effects related to the immune system. Most oncolytic viruses (OVs) have the potential to multiply in cancer cells, which are more than normal cells in malignant tissue and can induce immune responses. Therefore, tons of efforts and research have been started on the utilization of OVs as a treatment for cancer and have shown promising in treating cancers with less side effects. In this article, we have gathered studies about oncolytic viruses and their effectiveness in cancer treatment.Please confirm if the author names are presented accurately and in the correct sequence (given name, middle name/initial, family name). Author 1 Given name: [Omid Salahi] Last name [Ardekani], Author 2 Given name: [Mohammad Mehdi] Last name [Fazeli], Author 3 Given name: [Nillofar Asadi] Last name [Jemezghani]. Also, kindly confirm the details in the metadata are correct.Confirmed.
Collapse
Affiliation(s)
- Arash Letafati
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Omid Salahi Ardekani
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | - Mina Naderisemiromi
- Department of Immunology, Faculty of Medicine and Health, The University of Manchester, Manchester, UK
| | - Mohammad Mehdi Fazeli
- Research Center for Clinical Virology, Tehran University of Medical Science, Tehran, Iran
| | | | - Jila Yavarian
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Shakiba Y, Vorobyev PO, Yusubalieva GM, Kochetkov DV, Zajtseva KV, Valikhov MP, Kalsin VA, Zabozlaev FG, Semkina AS, Troitskiy AV, Baklaushev VP, Chumakov PM, Lipatova AV. Oncolytic therapy with recombinant vaccinia viruses targeting the interleukin-15 pathway elicits a synergistic response. Mol Ther Oncolytics 2023; 29:158-168. [PMID: 37387795 PMCID: PMC10300409 DOI: 10.1016/j.omto.2023.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 05/10/2023] [Indexed: 07/01/2023] Open
Abstract
We developed recombinant variants of oncolytic vaccinia virus LIVP strain expressing interleukin-15 (IL-15) or its receptor subunit alpha (IL-15Rα) to stimulate IL-15-dependent immune cells. We evaluated their oncolytic activity either alone or in combination with each other in vitro and in vivo using the murine CT26 colon carcinoma and 4T1 breast carcinoma models. We demonstrated that the admixture of these recombinant variants could promote the generation of the IL-15/IL-15Rα complex. In vitro studies indicated that 4T1 breast cancer cells were more susceptible to the developed recombinant viruses. In vivo studies showed significant survival benefits and tumor regression in 4T1 breast cancer syngeneic mice that received a combination of LIVP-IL15-RFP with LIVP-IL15Ra-RFP. Histological analysis showed recruited lymphocytes at the tumor region, while no harmful effects to the liver or spleen of the animals were detected. Evaluating tumor-infiltrated lymphocytes represented profound activation of cytotoxic T cells and macrophages in mice receiving combination therapy. Thus, our experiments showed superior oncolytic effectiveness of simultaneous injection of LIVP-IL15-RFP and LIVP-IL15Ra-RFP in breast cancer-bearing mice. The combined therapy by these recombinant variants represents a potent and versatile approach for developing new immunotherapies for breast cancer.
Collapse
Affiliation(s)
- Yasmin Shakiba
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia
| | - Pavel O. Vorobyev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Gaukhar M. Yusubalieva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Federal Research and Clinical Center for Specialized Types of Medical Care and Medical Technologies, FMBA of Russia, 115682 Moscow, Russian Federation
- Federal Center of Brain Research and Neurotechnologies, FMBA of Russia, 117513 Moscow, Russian Federation
| | - Dmitry V. Kochetkov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Ksenia V. Zajtseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Marat P. Valikhov
- Department of Neurobiology, Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, 119034 Moscow, Russia
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University 117997 Moscow, Russia
| | - Vladimir A. Kalsin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Federal Research and Clinical Center for Specialized Types of Medical Care and Medical Technologies, FMBA of Russia, 115682 Moscow, Russian Federation
| | - Fedor G. Zabozlaev
- Federal Research and Clinical Center for Specialized Types of Medical Care and Medical Technologies, FMBA of Russia, 115682 Moscow, Russian Federation
| | - Alevtina S. Semkina
- Department of Neurobiology, Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, 119034 Moscow, Russia
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University 117997 Moscow, Russia
| | - Alexander V. Troitskiy
- Federal Research and Clinical Center for Specialized Types of Medical Care and Medical Technologies, FMBA of Russia, 115682 Moscow, Russian Federation
| | - Vladimir P. Baklaushev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Federal Research and Clinical Center for Specialized Types of Medical Care and Medical Technologies, FMBA of Russia, 115682 Moscow, Russian Federation
- Federal Center of Brain Research and Neurotechnologies, FMBA of Russia, 117513 Moscow, Russian Federation
| | - Peter M. Chumakov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anastasia V. Lipatova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
5
|
Shakiba Y, Vorobyev PO, Naumenko VA, Kochetkov DV, Zajtseva KV, Valikhov MP, Yusubalieva GM, Gumennaya YD, Emelyanov EA, Semkina AS, Baklaushev VP, Chumakov PM, Lipatova AV. Oncolytic Efficacy of a Recombinant Vaccinia Virus Strain Expressing Bacterial Flagellin in Solid Tumor Models. Viruses 2023; 15:828. [PMID: 37112810 PMCID: PMC10142208 DOI: 10.3390/v15040828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/17/2023] [Accepted: 03/22/2023] [Indexed: 04/29/2023] Open
Abstract
Oncolytic viral therapy is a promising novel approach to cancer treatment. Oncolytic viruses cause tumor regression through direct cytolysis on the one hand and recruiting and activating immune cells on the other. In this study, to enhance the antitumor efficacy of the thymidine kinase-deficient vaccinia virus (VV, Lister strain), recombinant variants encoding bacterial flagellin (subunit B) of Vibrio vulnificus (LIVP-FlaB-RFP), firefly luciferase (LIVP-Fluc-RFP) or red fluorescent protein (LIVP-RFP) were developed. The LIVP-FLuc-RFP strain demonstrated exceptional onco-specificity in tumor-bearing mice, detected by the in vivo imaging system (IVIS). The antitumor efficacy of these variants was explored in syngeneic murine tumor models (B16 melanoma, CT26 colon cancer and 4T1 breast cancer). After intravenous treatment with LIVP-FlaB-RFP or LIVP-RFP, all mice tumor models exhibited tumor regression, with a prolonged survival rate in comparison with the control mice. However, superior oncolytic activity was observed in the B16 melanoma models treated with LIVP-FlaB-RFP. Tumor-infiltrated lymphocytes and the cytokine analysis of the serum and tumor samples from the melanoma-xenografted mice treated with these virus variants demonstrated activation of the host's immune response. Thus, the expression of bacterial flagellin by VV can enhance its oncolytic efficacy against immunosuppressive solid tumors.
Collapse
Affiliation(s)
- Yasmin Shakiba
- Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia;
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Pavel O. Vorobyev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Victor A. Naumenko
- Department of Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, 119034 Moscow, Russia
| | - Dmitry V. Kochetkov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Ksenia V. Zajtseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Marat P. Valikhov
- Department of Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, 119034 Moscow, Russia
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Gaukhar M. Yusubalieva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Federal Research and Clinical Center for Specialized Types of Medical Care and Medical Technologies FMBA of Russia, 115682 Moscow, Russia
| | - Yana D. Gumennaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Egor A. Emelyanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Alevtina S. Semkina
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Vladimir P. Baklaushev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Federal Research and Clinical Center for Specialized Types of Medical Care and Medical Technologies FMBA of Russia, 115682 Moscow, Russia
| | - Peter M. Chumakov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anastasia V. Lipatova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
6
|
Viral Vectors in Gene Therapy: Where Do We Stand in 2023? Viruses 2023; 15:v15030698. [PMID: 36992407 PMCID: PMC10059137 DOI: 10.3390/v15030698] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/23/2023] [Accepted: 03/02/2023] [Indexed: 03/11/2023] Open
Abstract
Viral vectors have been used for a broad spectrum of gene therapy for both acute and chronic diseases. In the context of cancer gene therapy, viral vectors expressing anti-tumor, toxic, suicide and immunostimulatory genes, such as cytokines and chemokines, have been applied. Oncolytic viruses, which specifically replicate in and kill tumor cells, have provided tumor eradication, and even cure of cancers in animal models. In a broader meaning, vaccine development against infectious diseases and various cancers has been considered as a type of gene therapy. Especially in the case of COVID-19 vaccines, adenovirus-based vaccines such as ChAdOx1 nCoV-19 and Ad26.COV2.S have demonstrated excellent safety and vaccine efficacy in clinical trials, leading to Emergency Use Authorization in many countries. Viral vectors have shown great promise in the treatment of chronic diseases such as severe combined immunodeficiency (SCID), muscular dystrophy, hemophilia, β-thalassemia, and sickle cell disease (SCD). Proof-of-concept has been established in preclinical studies in various animal models. Clinical gene therapy trials have confirmed good safety, tolerability, and therapeutic efficacy. Viral-based drugs have been approved for cancer, hematological, metabolic, neurological, and ophthalmological diseases as well as for vaccines. For example, the adenovirus-based drug Gendicine® for non-small-cell lung cancer, the reovirus-based drug Reolysin® for ovarian cancer, the oncolytic HSV T-VEC for melanoma, lentivirus-based treatment of ADA-SCID disease, and the rhabdovirus-based vaccine Ervebo against Ebola virus disease have been approved for human use.
Collapse
|
7
|
Lundstrom K. Gene Therapy Cargoes Based on Viral Vector Delivery. Curr Gene Ther 2023; 23:111-134. [PMID: 36154608 DOI: 10.2174/1566523222666220921112753] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 07/13/2022] [Accepted: 08/05/2022] [Indexed: 11/22/2022]
Abstract
Viral vectors have been proven useful in a broad spectrum of gene therapy applications due to their possibility to accommodate foreign genetic material for both local and systemic delivery. The wide range of viral vectors has enabled gene therapy applications for both acute and chronic diseases. Cancer gene therapy has been addressed by the delivery of viral vectors expressing anti-tumor, toxic, and suicide genes for the destruction of tumors. Delivery of immunostimulatory genes such as cytokines and chemokines has also been applied for cancer therapy. Moreover, oncolytic viruses specifically replicating in and killing tumor cells have been used as such for tumor eradication or in combination with tumor killing or immunostimulatory genes. In a broad meaning, vaccines against infectious diseases and various cancers can be considered gene therapy, which has been highly successful, not the least for the development of effective COVID-19 vaccines. Viral vector-based gene therapy has also demonstrated encouraging and promising results for chronic diseases such as severe combined immunodeficiency (SCID), muscular dystrophy, and hemophilia. Preclinical gene therapy studies in animal models have demonstrated proof-of-concept for a wide range of disease indications. Clinical evaluation of drugs and vaccines in humans has showed high safety levels, good tolerance, and therapeutic efficacy. Several gene therapy drugs such as the adenovirus-based drug Gendicine® for non-small-cell lung cancer, the reovirus-based drug Reolysin® for ovarian cancer, lentivirus-based treatment of SCID-X1 disease, and the rhabdovirus-based vaccine Ervebo against Ebola virus disease, and adenovirus-based vaccines against COVID-19 have been developed.
Collapse
|
8
|
Wei M, Zuo S, Chen Z, Qian P, Zhang Y, Kong L, Gao H, Wei J, Dong J. Oncolytic vaccinia virus expressing a bispecific T-cell engager enhances immune responses in EpCAM positive solid tumors. Front Immunol 2022; 13:1017574. [PMID: 36451817 PMCID: PMC9702515 DOI: 10.3389/fimmu.2022.1017574] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 10/26/2022] [Indexed: 10/27/2023] Open
Abstract
Insufficient intratumoral T-cell infiltration and lack of tumor-specific immune surveillance in tumor microenvironment (TME) hinder the progression of cancer immunotherapy. In this study, we explored a recombinant vaccinia virus encoding an EpCAM BiTE (VV-EpCAM BiTE) to modulate the immune suppressive microenvironment to enhance antitumor immunity in several solid tumors. VV-EpCAM BiTE effectively infected, replicated and lysed malignant cells. The EpCAM BiTE secreted from infected malignants effectively mediated the binding of EpCAM-positive tumor cells and CD3ϵ on T cells, which led to activation of naive T-cell and the release of cytokines, such as IFN-γ and IL-2. Intratumoral administration of VV-EpCAM BiTE significantly enhanced antitumor activity in malignancies with high other than with low EpCAM expression level. In addition, immune cell infiltration was significantly increased in TME upon VV-EpCAM BiTE treatment, CD8+ T cell exhaustion was reduced and T-cell-mediated immune activation was markedly enhanced. Taken together, VV-EpCAM BiTE sophistically combines the antitumor advantages of bispecific antibodies and oncolytic viruses, which provides preclinical evidence for the therapeutic potential of VV-EpCAM BiTE.
Collapse
Affiliation(s)
- Min Wei
- Affiliated Yancheng No.1 People’s Hospital, Medical School of Nanjing University, Yancheng, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Shuguang Zuo
- Liuzhou Key Laboratory of Molecular Diagnosis, Guangxi Key Laboratory of Molecular Diagnosis and Application, Affiliated Liutie Central Hospital of Guangxi Medical University, Liuzhou, Guangxi, China
| | - Zhimin Chen
- Affiliated Yancheng No.1 People’s Hospital, Medical School of Nanjing University, Yancheng, China
| | - Peng Qian
- Affiliated Yancheng No.1 People’s Hospital, Medical School of Nanjing University, Yancheng, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Yenan Zhang
- Affiliated Yancheng No.1 People’s Hospital, Medical School of Nanjing University, Yancheng, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Lingkai Kong
- Affiliated Yancheng No.1 People’s Hospital, Medical School of Nanjing University, Yancheng, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Honglan Gao
- Affiliated Yancheng No.1 People’s Hospital, Medical School of Nanjing University, Yancheng, China
| | - Jiwu Wei
- Affiliated Yancheng No.1 People’s Hospital, Medical School of Nanjing University, Yancheng, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Jie Dong
- Affiliated Yancheng No.1 People’s Hospital, Medical School of Nanjing University, Yancheng, China
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
9
|
Mukesh RK, Kalam AA, Nag J, Jaikumar VS, Kunnakkadan U, Kumar NA, Suma SM, Rajavelu A, Johnson JB. Chandipura virus induces cell death in cancer cell lines of human origin and promotes tumor regression in vivo. MOLECULAR THERAPY-ONCOLYTICS 2021; 23:254-265. [PMID: 34761105 PMCID: PMC8554107 DOI: 10.1016/j.omto.2021.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 09/20/2021] [Accepted: 09/29/2021] [Indexed: 11/25/2022]
Abstract
Chandipura virus (CHPV) is an emerging human pathogen of great clinical significance. In this study, we have investigated the susceptibility pattern of both normal and cancer cell lines of human origin to wild-type (wt) CHPV in order to explore the possibility of developing CHPV as an oncolytic vector (OV). Marked cytopathic effect along with enhanced virus output was observed in cancer cell lines (HeLa, A549, U-138, PC-3, and HepG2) in comparison to normal human adult dermal fibroblast (HADF) cells. At an MOI of 0.1, cancer cell lines were differentially susceptible to CHPV, with cells like HeLa and U-138 having pronounced cell death, while the PC-3 were comparatively resistant. All cell lines used in the study except U-138 restricted CHPV infection to varying degrees with IFN-β pre-treatment and supplementation of interferon (IFN) could neither activate the IFN signaling pathway in U-138 cells. Finally, U-138 tumor xenografts established in non-obese diabetic severe combined immunodeficiency (NOD/SCID) mice showed significant delay in tumor growth in the CHPV-challenged animals. Thus, targeted cytopathic effect in cancer cells at a very low dose with restricted replication in normal cells offers a rationale to exploit CHPV as an oncolytic vector in the future.
Collapse
Affiliation(s)
- Reshma Koolaparambil Mukesh
- Pathogen Biology, Virology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India.,Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Azeem Abdul Kalam
- Pathogen Biology, Virology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India
| | - Joydeep Nag
- Pathogen Biology, Virology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India.,Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Vishnu Sunil Jaikumar
- Animal Research Facility, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India
| | - Umerali Kunnakkadan
- Pathogen Biology, Virology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India.,Department of Biotechnology, University of Kerala, Thiruvananthapuram 695581, Kerala, India
| | - Nisha Asok Kumar
- Pathogen Biology, Virology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India.,Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | | | - Arumugam Rajavelu
- Pathogen Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India
| | - John Bernet Johnson
- Pathogen Biology, Virology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, Kerala, India
| |
Collapse
|
10
|
Zhang S, Rabkin SD. The discovery and development of oncolytic viruses: are they the future of cancer immunotherapy? Expert Opin Drug Discov 2020; 16:391-410. [PMID: 33232188 DOI: 10.1080/17460441.2021.1850689] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Despite diverse treatment modalities and novel therapies, many cancers and patients are not effectively treated. Cancer immunotherapy has recently achieved breakthrough status yet is not effective in all cancer types or patients and can generate serious adverse effects. Oncolytic viruses (OVs) are a promising new therapeutic modality that harnesses virus biology and host interactions to treat cancer. OVs, genetically engineered or natural, preferentially replicate in and kill cancer cells, sparing normal cells/tissues, and mediating anti-tumor immunity.Areas covered: This review focuses on OVs as cancer therapeutic agents from a historical perspective, especially strategies to boost their immunotherapeutic activities. OVs offer a multifaceted platform, whose activities are modulated based on the parental virus and genetic alterations. In addition to direct viral effects, many OVs can be armed with therapeutic transgenes to also act as gene therapy vectors, and/or combined with other drugs or therapies.Expert opinion: OVs are an amazingly versatile and malleable class of cancer therapies. They tend to target cellular and host physiology as opposed to specific genetic alterations, which potentially enables broad responsiveness. The biological complexity of OVs have hindered their translation; however, the recent approval of talimogene laherparepvec (T-Vec) has invigorated the field.
Collapse
Affiliation(s)
- Shunchuan Zhang
- Molecular Neurosurgery Laboratory and the Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Samuel D Rabkin
- Molecular Neurosurgery Laboratory and the Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| |
Collapse
|
11
|
Current Perspectives on Therapies, Including Drug Delivery Systems, for Managing Glioblastoma Multiforme. ACS Chem Neurosci 2020; 11:2962-2977. [PMID: 32945654 DOI: 10.1021/acschemneuro.0c00555] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma multiforme (GBM), a standout among the most dangerous class of central nervous system (CNS) cancer, is most common and is an aggressive malignant brain tumor in adults. In spite of developments in modality therapy, it remains mostly incurable. Consequently, the need for novel systems, strategies, or therapeutic approaches for enhancing the assortment of active agents meant for GBM becomes an important criterion. Currently, cancer research focuses mainly on improving the treatment of GBM via diverse novel drug delivery systems. The treatment options at diagnosis are multimodal and include radiation therapy. Moreover, significant advances in understanding the molecular pathology of GBM and associated cell signaling pathways have opened opportunities for new therapies. Innovative treatment such as immunotherapy also gives hope for enhanced survival. The objective of this work was to collect and report the recent research findings to manage GBM. The present review includes existing novel drug delivery systems and therapies intended for managing GBM. Reported novel drug delivery systems and diverse therapies seem to be precise, secure, and relatively effective, which could lead to a new track for the obliteration of GBM.
Collapse
|
12
|
Stewart JH, Blazer DG, Calderon MJG, Carter TM, Eckhoff A, Al Efishat MA, Fernando DG, Foster JM, Hayes-Jordan A, Johnston FM, Lautz TB, Levine EA, Maduekwe UN, Mangieri CW, Moaven O, Mogal H, Shen P, Votanopoulos KI. The Evolving Management of Peritoneal Surface Malignancies. Curr Probl Surg 2020; 58:100860. [PMID: 33832580 DOI: 10.1016/j.cpsurg.2020.100860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 07/04/2020] [Indexed: 02/07/2023]
Affiliation(s)
| | - Dan G Blazer
- Division of Surgical Oncology, Duke University Medical Center, Durham, NC
| | | | | | | | | | | | - Jason M Foster
- Fred and Pamela Buffet Cancer Center, University of Nebraska, Omaha, NE
| | | | - Fabian M Johnston
- Complex General Surgical Oncology Program, Johns Hopkins University, Baltimore, MD
| | - Timothy B Lautz
- Northwestern University Feinberg School of Medicine, Chicago, IL
| | | | - Ugwuji N Maduekwe
- Division of Surgical Oncology and Endocrine Surgery, University of North Carolina, Chapel Hill, NC
| | | | | | | | - Perry Shen
- Wake Forest University School of Medicine, Winston-Salem, NC
| | | |
Collapse
|
13
|
Hromic-Jahjefendic A, Lundstrom K. Viral Vector-Based Melanoma Gene Therapy. Biomedicines 2020; 8:E60. [PMID: 32187995 PMCID: PMC7148454 DOI: 10.3390/biomedicines8030060] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 02/06/2023] Open
Abstract
Gene therapy applications of oncolytic viruses represent an attractive alternative for cancer treatment. A broad range of oncolytic viruses, including adenoviruses, adeno-associated viruses, alphaviruses, herpes simplex viruses, retroviruses, lentiviruses, rhabdoviruses, reoviruses, measles virus, Newcastle disease virus, picornaviruses and poxviruses, have been used in diverse preclinical and clinical studies for the treatment of various diseases, including colon, head-and-neck, prostate and breast cancer as well as squamous cell carcinoma and glioma. The majority of studies have focused on immunotherapy and several drugs based on viral vectors have been approved. However, gene therapy for malignant melanoma based on viral vectors has not been utilized to its full potential yet. This review represents a summary of the achievements of preclinical and clinical studies using viral vectors, with the focus on malignant melanoma.
Collapse
Affiliation(s)
- Altijana Hromic-Jahjefendic
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, 71000 Sarajevo, Bosnia and Herzegovina;
| | | |
Collapse
|
14
|
Deng L, Fan J, Ding Y, Yang X, Huang B, Hu Z. Target Therapy With Vaccinia Virus Harboring IL-24 For Human Breast Cancer. J Cancer 2020; 11:1017-1026. [PMID: 31956348 PMCID: PMC6959063 DOI: 10.7150/jca.37590] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 10/26/2019] [Indexed: 01/04/2023] Open
Abstract
Background: Breast cancer is a heterogeneous disease with high aggression and novel targeted therapeutic strategies are required. Oncolytic vaccinia virus is an attractive candidate for cancer treatment due to its tumor cell-specific replication causing lysis of tumor cells as well as a delivery vector to overexpress therapeutic transgenes. Interleukin-24 (IL-24) is a novel tumor suppressor cytokine that selectively induces apoptosis in a wide variety of tumor types, including breast cancer. In this study, we used vaccinia virus as a delivery vector to express IL-24 gene and antitumor effects were evaluated both in vitro and in vivo. Methods: The vaccinia virus strain Guang9 armed with IL-24 gene (VG9-IL-24) was constructed via disruption of the viral thymidine kinase (TK) gene region. The cytotoxicity of VG9-IL-24 in various breast cancer cell lines was assessed by MTT and cell cycle progression and apoptosis were examined by flow cytometry. In vivo antitumor effects were further observed in MDA-MB-231 xenograft mouse model. Results: In vitro, VG9-IL-24 efficiently infected and selectively killed breast cancer cells with no strong cytotoxicity to normal cells. VG9-IL-24 induced increased number of apoptotic cells and blocked breast cancer cells in the G2/M phase of the cell cycle. Western blotting results indicated that VG9-IL-24-mediated apoptosis was related to PI3K/β-catenin signaling pathway. In vivo, VG9-IL-24 delayed tumor growth and improved survival. Conclusions: Our findings provided documentation that VG9-IL-24 was targeted in vitro and exhibited enhanced antitumor effects, and it may be an innovative therapy for breast cancer.
Collapse
Affiliation(s)
- Lili Deng
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Jun Fan
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Yuedi Ding
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Xue Yang
- Wuxi Children's Hospital, Wuxi People's Hospital affiliated to Nanjing Medical University, Wuxi 214023, China
| | - Biao Huang
- School of Life Science, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Zhigang Hu
- Wuxi Children's Hospital, Wuxi People's Hospital affiliated to Nanjing Medical University, Wuxi 214023, China
| |
Collapse
|
15
|
Lundstrom K. Viral Vectors in Gene Therapy. Diseases 2018; 6:diseases6020042. [PMID: 29883422 PMCID: PMC6023384 DOI: 10.3390/diseases6020042] [Citation(s) in RCA: 318] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/15/2018] [Accepted: 05/16/2018] [Indexed: 01/02/2023] Open
Abstract
Applications of viral vectors have found an encouraging new beginning in gene therapy in recent years. Significant improvements in vector engineering, delivery, and safety have placed viral vector-based therapy at the forefront of modern medicine. Viral vectors have been employed for the treatment of various diseases such as metabolic, cardiovascular, muscular, hematologic, ophthalmologic, and infectious diseases and different types of cancer. Recent development in the area of immunotherapy has provided both preventive and therapeutic approaches. Furthermore, gene silencing generating a reversible effect has become an interesting alternative, and is well-suited for delivery by viral vectors. A number of preclinical studies have demonstrated therapeutic and prophylactic efficacy in animal models and furthermore in clinical trials. Several viral vector-based drugs have also been globally approved.
Collapse
|
16
|
O'Leary MP, Choi AH, Kim SI, Chaurasiya S, Lu J, Park AK, Woo Y, Warner SG, Fong Y, Chen NG. Novel oncolytic chimeric orthopoxvirus causes regression of pancreatic cancer xenografts and exhibits abscopal effect at a single low dose. J Transl Med 2018; 16:110. [PMID: 29699566 PMCID: PMC5918769 DOI: 10.1186/s12967-018-1483-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 04/17/2018] [Indexed: 02/07/2023] Open
Abstract
Background Pancreatic ductal adenocarcinoma (PDAC) has been increasing by 0.5% per year in the United States. PDAC portends a dismal prognosis and novel therapies are needed. This study describes the generation and characterization of a novel oncolytic chimeric orthopoxvirus for the treatment of pancreatic cancer. Methods After chimerization and high-throughput screening, CF33 was chosen from 100 new chimeric orthopoxvirus isolates for its ability to kill pancreatic cancer cells. In vitro cytotoxicity was assayed in six pancreatic cancer cell lines. In vivo efficacy and toxicity were evaluated in PANC-1 and MIA PaCa-2 xenograft models. Results CF33 caused rapid killing of six pancreatic cancer cells lines in vitro, releasing damage-associated molecular patterns, and regression of PANC-1 injected and non-injected distant xenografts in vivo after a single low intratumoral dose of 103 plaque-forming units. Using luciferase imaging, CF33 was noted to preferentially replicate in tumors which corresponds to the low viral titers found in solid organs. Conclusion The low dose of CF33 required to treat pancreatic cancer in this preclinical study may ease the manufacturing and dosing challenges currently facing oncolytic viral therapy.
Collapse
Affiliation(s)
- Michael P O'Leary
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA
| | - Audrey H Choi
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA
| | - Sang-In Kim
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA
| | - Shyambabu Chaurasiya
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA
| | - Jianming Lu
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA
| | - Anthony K Park
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA
| | - Yanghee Woo
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA
| | - Susanne G Warner
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA
| | - Yuman Fong
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA.,Center for Gene Therapy, Department of Hematologic and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Nanhai G Chen
- Department of Surgery, Division of Surgical Oncology, City of Hope National Medical Center, 1500 Duarte Rd., Duarte, CA, 91010, USA. .,Center for Gene Therapy, Department of Hematologic and Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, 91010, USA. .,Gene Editing and Viral Vector Core, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, 91010, USA.
| |
Collapse
|
17
|
Deng L, Fan J, Ding Y, Zhang J, Zhou B, Zhang Y, Huang B. Oncolytic efficacy of thymidine kinase-deleted vaccinia virus strain Guang9. Oncotarget 2018; 8:40533-40543. [PMID: 28465492 PMCID: PMC5522336 DOI: 10.18632/oncotarget.17125] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 04/04/2017] [Indexed: 11/29/2022] Open
Abstract
Oncolytic virotherapy is being developed as a promising platform for cancer therapy due to its ability to lyse cancer cells in a tumor-specific manner. Vaccinia virus has been used as a live vaccine in the smallpox eradication program and now is being potential in cancer therapy with a great safety profile. Vaccinia strain Guang9 (VG9) is an attenuated Chinese vaccinia virus and its oncolytic efficacy has been evaluated in our previous study. To improve the tumor selectivity and oncolytic efficacy, we here developed a thymidine kinase (TK)-deleted vaccinia virus based on Guang9 strain. The viral replication, marker gene expression and cytotoxicity in various cell lines were evaluated; antitumor effects in vivo were assessed in multiple tumor models. In vitro, the TK-deleted vaccinia virus replicated rapidly, but the cytotoxicity varied in different cell lines. It was notably attenuated in normal cells and resting cells in vitro, while tumor-selectively replicated in vivo. Significant antitumor effects were observed both in murine melanoma tumor model and human hepatoma tumor model. It significantly inhibited the growth of subcutaneously implanted tumors and prolonged the survival of tumor-bearing mice. Collectively, TK-deleted vaccinia strain Guang9 is a promising constructive virus vector for tumor-directed gene therapy and will be a potential therapeutic strategy in cancer treatment.
Collapse
Affiliation(s)
- Lili Deng
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Jun Fan
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Yuedi Ding
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Jue Zhang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Bin Zhou
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Yi Zhang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | - Biao Huang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| |
Collapse
|
18
|
Lundstrom K. New frontiers in oncolytic viruses: optimizing and selecting for virus strains with improved efficacy. Biologics 2018; 12:43-60. [PMID: 29445265 PMCID: PMC5810530 DOI: 10.2147/btt.s140114] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Oncolytic viruses have demonstrated selective replication and killing of tumor cells. Different types of oncolytic viruses – adenoviruses, alphaviruses, herpes simplex viruses, Newcastle disease viruses, rhabdoviruses, Coxsackie viruses, and vaccinia viruses – have been applied as either naturally occurring or engineered vectors. Numerous studies in animal-tumor models have demonstrated substantial tumor regression and prolonged survival rates. Moreover, clinical trials have confirmed good safety profiles and therapeutic efficacy for oncolytic viruses. Most encouragingly, the first cancer gene-therapy drug – Gendicine, based on oncolytic adenovirus type 5 – was approved in China. Likewise, a second-generation oncolytic herpes simplex virus-based drug for the treatment of melanoma has been registered in the US and Europe as talimogene laherparepvec.
Collapse
|
19
|
Abstract
Gene therapy based on viral vectors has demonstrated steady progress recently, not only in the area of cancers. A multitude of viral vectors has been engineered for both preventive and therapeutic applications. Two main approaches comprise of viral vector-based delivery of toxic or anticancer genes or immunization with anticancer antigens. Tumor growth inhibition and tumor regression have been observed, providing improved survival rates in animal tumor models. Furthermore, vaccine-based cancer immunotherapy has demonstrated both tumor regression and protection against challenges with lethal doses of tumor cells. Several clinical trials with viral vectors have also been conducted. Additionally, viral vector-based cancer drugs have been approved. This review gives an overview of different viral vector systems and their applications in cancer gene therapy.
Collapse
|
20
|
Zhang T, Suryawanshi YR, Kordish DH, Woyczesczyk HM, Jeng D, Essani K. Tanapoxvirus lacking a neuregulin-like gene regresses human melanoma tumors in nude mice. Virus Genes 2017; 53:52-62. [PMID: 27738905 PMCID: PMC5300959 DOI: 10.1007/s11262-016-1402-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 10/06/2016] [Indexed: 10/25/2022]
Abstract
Neuregulin (NRG), an epidermal growth factor is known to promote the growth of various cell types, including human melanoma cells through ErbB family of tyrosine kinases receptors. Tanapoxvirus (TPV)-encoded protein TPV-15L, a functional mimic of NRG, also acts through ErbB receptors. Here, we show that the TPV-15L protein promotes melanoma proliferation. TPV recombinant generated by deleting the 15L gene (TPVΔ15L) showed replication ability similar to that of wild-type TPV (wtTPV) in owl monkey kidney cells, human lung fibroblast (WI-38) cells, and human melanoma (SK-MEL-3) cells. However, a TPV recombinant with both 15L and the thymidine kinase (TK) gene 66R ablated (TPVΔ15LΔ66R) replicated less efficiently compared to TPVΔ15L and the parental virus. TPVΔ15L exhibited more robust tumor regression in the melanoma-bearing nude mice compared to other TPV recombinants. Our results indicate that deletion of TPV-15L gene product which facilitates the growth of human melanoma cells can be an effective strategy to enhance the oncolytic potential of TPV for the treatment of melanoma.
Collapse
Affiliation(s)
- Tiantian Zhang
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, USA
| | - Yogesh R Suryawanshi
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, USA
| | - Dennis H Kordish
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, USA
| | - Helene M Woyczesczyk
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, USA
| | - David Jeng
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, USA
| | - Karim Essani
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, USA.
| |
Collapse
|
21
|
Toward a world of theranostic medication: Programming biological sentinel systems for therapeutic intervention. Adv Drug Deliv Rev 2016; 105:66-76. [PMID: 27189230 DOI: 10.1016/j.addr.2016.05.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 03/02/2016] [Accepted: 05/05/2016] [Indexed: 12/21/2022]
Abstract
Theranostic systems support diagnostic and therapeutic functions in a single integrated entity and enable precise spatiotemporal control of the generation of therapeutic molecules according to the individual patient's disease state, thereby maximizing the therapeutic outcome and minimizing side effects. These systems can also incorporate reporter systems equipped with a disease-sensing module that can be used to estimate the efficacy of treatment in vivo. Among these reporter systems, biological sentinel systems, such as viruses, bacteria, and mammalian cells, have great potential for use in the development of novel theranostic systems because of their ability to sense a variety of disease markers and secrete various therapeutic molecules. Furthermore, recent advances in biotechnology and synthetic biology have made it possible to treat these biological systems as true programmable entities capable of conducting complex operations, to accurately identify each individual patient's disease state. In this review, we introduce the basic design principles of these rapidly expanding classes of biological sentinel system-based theranostic agents, with a focus on recent advances, and we also discuss potential enabling technologies that can further improve these systems and provide more sophisticated therapeutic interventions in the near future. In addition, we consider the possibility of synergistic use of theranostic agents that use different modalities and discuss the prospects for next-generation theranostic agents.
Collapse
|
22
|
Abstract
Oncolytic viruses (OVs) are being extensively studied for their potential roles in the development of cancer therapy regimens. In addition to their direct lytic effects, OVs can initiate and drive systemic antitumor immunity indirectly via release of tumor antigen, as well as by encoding and delivering immunostimulatory molecules. This combination makes them an effective platform for the development of immunotherapeutic strategies beyond their primary lytic function. Engineering the viruses to also express tumor-associated antigens (TAAs) allows them to simultaneously serve as therapeutic vaccines, targeting and amplifying an immune response to TAAs. Our group and others have shown that vaccinating intratumorally with a poxvirus that encodes TAAs, in addition to immune stimulatory molecules, can modulate the tumor microenvironment, overcome immune inhibitory pathways, and drive both local and systemic tumor specific immune responses.
Collapse
|
23
|
Abstract
Viral vectors have frequently been applied in gene therapy with the final goal of treating various diseases in the areas of neurology, neurodegeneration, metabolic disease, and cancer. Vectors have been engineered based on AAV, adenoviruses, alphaviruses, herpes simplex viruses, lentiviruses, and retroviruses. Some vectors are suitable for short-term episomal transgene expression, whereas others are integrated into the host cell genome to provide long-term expression. Additionally, hybrid vectors with favorable features from different viruses have been developed. Therapeutic genes of choice have typically been toxic genes such as thymidine kinase, pro-apoptotic genes like Bax, and immunostimulatory genes (for instance, interleukin-12). A large number of animal studies have demonstrated proof of concept of viral gene therapy. Many types of viral vectors have been employed in more than 700 clinical trials that have been carried out or are currently in progress.
Collapse
Affiliation(s)
- Kenneth Lundstrom
- Regulon Inc., Chemin des Croisettes 22, CH-1066 Epalinges, Switzerland.
| |
Collapse
|
24
|
Hirvinen M, Capasso C, Guse K, Garofalo M, Vitale A, Ahonen M, Kuryk L, Vähä-Koskela M, Hemminki A, Fortino V, Greco D, Cerullo V. Expression of DAI by an oncolytic vaccinia virus boosts the immunogenicity of the virus and enhances antitumor immunity. MOLECULAR THERAPY-ONCOLYTICS 2016; 3:16002. [PMID: 27626058 PMCID: PMC5008257 DOI: 10.1038/mto.2016.2] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 12/18/2015] [Indexed: 12/17/2022]
Abstract
In oncolytic virotherapy, the ability of the virus to activate the immune system is a key attribute with regard to long-term antitumor effects. Vaccinia viruses bear one of the strongest oncolytic activities among all oncolytic viruses. However, its capacity for stimulation of antitumor immunity is not optimal, mainly due to its immunosuppressive nature. To overcome this problem, we developed an oncolytic VV that expresses intracellular pattern recognition receptor DNA-dependent activator of IFN-regulatory factors (DAI) to boost the innate immune system and to activate adaptive immune cells in the tumor. We showed that infection with DAI-expressing VV increases expression of several genes related to important immunological pathways. Treatment with DAI-armed VV resulted in significant reduction in the size of syngeneic melanoma tumors in mice. When the mice were rechallenged with the same tumor, DAI-VV-treated mice completely rejected growth of the new tumor, which indicates immunity established against the tumor. We also showed enhanced control of growth of human melanoma tumors and elevated levels of human T-cells in DAI-VV-treated mice humanized with human peripheral blood mononuclear cells. We conclude that expression of DAI by an oncolytic VV is a promising way to amplify the vaccine potency of an oncolytic vaccinia virus to trigger the innate-and eventually the long-lasting adaptive immunity against cancer.
Collapse
Affiliation(s)
- Mari Hirvinen
- Laboratory of ImmunoViroTherapy, Centre for Drug Research (CDR), Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki , Helsinki, Finland
| | - Cristian Capasso
- Laboratory of ImmunoViroTherapy, Centre for Drug Research (CDR), Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki , Helsinki, Finland
| | - Kilian Guse
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki , Helsinki, Finland
| | - Mariangela Garofalo
- Laboratory of ImmunoViroTherapy, Centre for Drug Research (CDR), Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki , Helsinki, Finland
| | - Andrea Vitale
- Department of Movement Sciences and Wellness (DiSMEB), University of Naples Parthenope and CEINGE-Biotecnologie Avanzate , Naples, Italy
| | - Marko Ahonen
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki , Helsinki, Finland
| | - Lukasz Kuryk
- Laboratory of ImmunoViroTherapy, Centre for Drug Research (CDR), Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland; Oncos Therapeutics Ltd., Helsinki, Finland
| | | | - Akseli Hemminki
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland; TILT Biotherapeutics, Ltd., Helsinki, Finland; Department of Oncology, Helsinki University Central Hospital, Helsinki, Finland
| | | | - Dario Greco
- Finnish Institute of Occupational Health , Helsinki, Finland
| | - Vincenzo Cerullo
- Laboratory of ImmunoViroTherapy, Centre for Drug Research (CDR), Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki , Helsinki, Finland
| |
Collapse
|
25
|
Yaghchi CA, Zhang Z, Alusi G, Lemoine NR, Wang Y. Vaccinia virus, a promising new therapeutic agent for pancreatic cancer. Immunotherapy 2015; 7:1249-58. [PMID: 26595180 PMCID: PMC4976866 DOI: 10.2217/imt.15.90] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The poor prognosis of pancreatic cancer patients signifies a need for radically new therapeutic strategies. Tumor-targeted oncolytic viruses have emerged as attractive therapeutic candidates for cancer treatment due to their inherent ability to specifically target and lyse tumor cells as well as induce antitumor effects by multiple action mechanisms. Vaccinia virus has several inherent features that make it particularly suitable for use as an oncolytic agent. In this review, we will discuss the potential of vaccinia virus in the management of pancreatic cancer in light of our increased understanding of cellular and immunological mechanisms involved in the disease process as well as our extending knowledge in the biology of vaccinia virus.
Collapse
Affiliation(s)
- Chadwan Al Yaghchi
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, UK
| | - Zhongxian Zhang
- National Centre for International Research in Cell & Gene Therapy, Sino-British Research Centre for Molecular Oncology, Zhengzhou University, China
| | - Ghassan Alusi
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, UK
| | - Nicholas R Lemoine
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, UK
- National Centre for International Research in Cell & Gene Therapy, Sino-British Research Centre for Molecular Oncology, Zhengzhou University, China
| | - Yaohe Wang
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, UK
- National Centre for International Research in Cell & Gene Therapy, Sino-British Research Centre for Molecular Oncology, Zhengzhou University, China
| |
Collapse
|
26
|
Conrad SJ, El-Aswad M, Kurban E, Jeng D, Tripp BC, Nutting C, Eversole R, Mackenzie C, Essani K. Oncolytic tanapoxvirus expressing FliC causes regression of human colorectal cancer xenografts in nude mice. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:19. [PMID: 25887490 PMCID: PMC4337313 DOI: 10.1186/s13046-015-0131-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 01/29/2015] [Indexed: 12/12/2022]
Abstract
Colorectal cancers are significant causes of morbidity and mortality and existing therapies often perform poorly for individuals afflicted with advanced disease. Oncolytic virotherapy is an emerging therapeutic modality with great promise for addressing this medical need. Herein we describe the in vivo testing of recombinant variants of the tanapoxvirus (TPV). Recombinant viruses were made ablated for either the 66R gene (encoding a thymidine kinase), the 2L gene (encoding a TNF-binding protein), or both. Some of the recombinants were armed to express mouse chemotactic protein 1 (mCCL2/mMCP-1), mouse granulocyte-monocyte colony stimulating factor (mGM-CSF), or bacterial flagellin (FliC). Tumors were induced in athymic nude mice by implantation of HCT 116 cells and subsequently treated by a single intratumoral injection of one of the recombinant TPVs. Histological examination showed a common neoplastic cell type and a range of immune cell infiltration, necrosis, and tumor cell organization. Significant regression was seen in tumors treated with virus TPV/Δ2L/Δ66R/fliC, and to a lesser extent the recombinants TPV/Δ2L and TPV/Δ66R. Our results suggest that oncolytic recombinants of the TPV armed with activators of the innate immune response may be effective virotherapeutic agents for colorectal cancers in humans and should be explored further to fully realize their potential.
Collapse
Affiliation(s)
- Steven J Conrad
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, USA.
| | - Mohamed El-Aswad
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, USA.
| | - Esaw Kurban
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, USA.
| | - David Jeng
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, USA.
| | - Brian C Tripp
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, USA.
| | - Charles Nutting
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, USA.
| | - Robert Eversole
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, USA.
| | - Charles Mackenzie
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan, USA.
| | - Karim Essani
- Laboratory of Virology, Department of Biological Sciences, Western Michigan University, Kalamazoo, MI, 49008, USA.
| |
Collapse
|
27
|
Progress in oncolytic virotherapy for the treatment of thyroid malignant neoplasm. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2014; 33:91. [PMID: 25366264 PMCID: PMC4242545 DOI: 10.1186/s13046-014-0091-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 10/22/2014] [Indexed: 01/05/2023]
Abstract
Thyroid malignant neoplasm develops from follicular or parafollicular thyroid cells. A higher proportion of anaplastic thyroid cancer has an adverse prognosis. New drugs are being used in clinical treatment. However, for advanced thyroid malignant neoplasm such as anaplastic thyroid carcinoma, the major impediment to successful control of the disease is the absence of effective therapies. Oncolytic virotherapy has significantly progressed as therapeutics in recent years. The advance is that oncolytic viruses can be designed with biological specificity to infect, replicate and lyse tumor cells. Significant advances in virotherapy have being achieved to improve the accessibility, safety and efficacy of the treatment. Therefore, it is necessary to summarize and bring together the main areas covered by these investigations for the virotherapy of thyroid malignant neoplasm. We provide an overview of the progress in virotherapy research and clinical trials, which employ virotherapy for thyroid malignant neoplasm as well as the future prospect for virotherapy of thyroid malignant neoplasms.
Collapse
|
28
|
Lamb SA, Rahman MM, McFadden G. Recombinant myxoma virus lacking all poxvirus ankyrin-repeat proteins stimulates multiple cellular anti-viral pathways and exhibits a severe decrease in virulence. Virology 2014; 464-465:134-145. [PMID: 25068401 DOI: 10.1016/j.virol.2014.06.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 05/01/2014] [Accepted: 06/17/2014] [Indexed: 12/31/2022]
Abstract
Although the production of single gene knockout viruses is a useful strategy to study viral gene functions, the redundancy of many host interactive genes within a complex viral genome can obscure their collective functions. In this study, a rabbit-specific poxvirus, myxoma virus (MYXV), was genetically altered to disrupt multiple members of the poxviral ankyrin-repeat (ANK-R) protein superfamily, M-T5, M148, M149 and M150. A particularly robust activation of the NF-κB pathway was observed in A549 cells following infection with the complete ANK-R knockout (vMyx-ANKsKO). Also, an increased release of IL-6 was only observed upon infection with vMyx-ANKsKO. In virus-infected rabbit studies, vMyx-ANKsKO was the most extensively attenuated and produced the smallest primary lesion of all ANK-R mutant constructs. This study provides the first insights into the shared functions of the poxviral ANK-R protein superfamily in vitro and in vivo.
Collapse
Affiliation(s)
- Stephanie A Lamb
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, 1600 SW Archer Road, PO Box 100266, Gainesville FL 32610, USA
| | - Masmudur M Rahman
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, 1600 SW Archer Road, PO Box 100266, Gainesville FL 32610, USA
| | - Grant McFadden
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, 1600 SW Archer Road, PO Box 100266, Gainesville FL 32610, USA
| |
Collapse
|
29
|
Parviainen S, Ahonen M, Diaconu I, Kipar A, Siurala M, Vähä-Koskela M, Kanerva A, Cerullo V, Hemminki A. GMCSF-armed vaccinia virus induces an antitumor immune response. Int J Cancer 2014; 136:1065-72. [PMID: 25042001 DOI: 10.1002/ijc.29068] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 06/23/2014] [Indexed: 12/14/2022]
Abstract
Oncolytic Western Reserve strain vaccinia virus selective for epidermal growth factor receptor pathway mutations and tumor-associated hypermetabolism was armed with human granulocyte-macrophage colony-stimulating factor (GMCSF) and a tdTomato fluorophore. As the assessment of immunological responses to human transgenes is challenging in the most commonly used animal models, we used immunocompetent Syrian golden hamsters, known to be sensitive to human GMCSF and semipermissive to vaccinia virus. Efficacy was initially tested in vitro on various human and hamster cell lines and oncolytic potency of transgene-carrying viruses was similar to unarmed virus. The hGMCSF-encoding virus was able to completely eradicate subcutaneous pancreatic tumors in hamsters, and to fully protect the animals from subsequent rechallenge with the same tumor. Induction of specific antitumor immunity was also shown by ex vivo co-culture experiments with hamster splenocytes. In addition, histological examination revealed increased infiltration of neutrophils and macrophages in GMCSF-virus-treated tumors. These findings help clarify the mechanism of action of GMCSF-armed vaccinia viruses undergoing clinical trials.
Collapse
Affiliation(s)
- Suvi Parviainen
- Cancer Gene Therapy Group, Department of Pathology and Transplantation Laboratory, Haartman Institute, University of Helsinki, Helsinki, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Hollenbaugh JA, Gee P, Baker J, Daly MB, Amie SM, Tate J, Kasai N, Kanemura Y, Kim DH, Ward BM, Koyanagi Y, Kim B. Host factor SAMHD1 restricts DNA viruses in non-dividing myeloid cells. PLoS Pathog 2013; 9:e1003481. [PMID: 23825958 PMCID: PMC3694861 DOI: 10.1371/journal.ppat.1003481] [Citation(s) in RCA: 140] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 05/22/2013] [Indexed: 01/01/2023] Open
Abstract
SAMHD1 is a newly identified anti-HIV host factor that has a dNTP triphosphohydrolase activity and depletes intracellular dNTP pools in non-dividing myeloid cells. Since DNA viruses utilize cellular dNTPs, we investigated whether SAMHD1 limits the replication of DNA viruses in non-dividing myeloid target cells. Indeed, two double stranded DNA viruses, vaccinia and herpes simplex virus type 1, are subject to SAMHD1 restriction in non-dividing target cells in a dNTP dependent manner. Using a thymidine kinase deficient strain of vaccinia virus, we demonstrate a greater restriction of viral replication in non-dividing cells expressing SAMHD1. Therefore, this study suggests that SAMHD1 is a potential innate anti-viral player that suppresses the replication of a wide range of DNA viruses, as well as retroviruses, which infect non-dividing myeloid cells. Various viral pathogens such as HIV-1, herpes simplex virus (HSV) and vaccinia virus infect terminally-differentiated/non-dividing macrophages during the course of viral pathogenesis. Unlike dividing cells, non-dividing cells lack chromosomal DNA replication, do not enter the cell cycle, and harbor very low levels of cellular dNTPs, which are substrates of viral DNA polymerases. A series of recent studies revealed that the host protein SAMHD1 is dNTP triphosphohydrolase, which contributes to the poor dNTP abundance in non-dividing myeloid cells, and restricts proviral DNA synthesis of HIV-1 and other lentiviruses in macrophages, dendritic cells, and resting T cells. In this report, we demonstrate that SAMHD1 also controls the replication of large dsDNA viruses: vaccinia virus and HSV-1, in primary human monocyte-derived macrophages. SAMHD1 suppresses the replication of these DNA viruses to an even greater extent in the absence of viral genes that are involved in dNTP metabolism such as thymidine kinase. Therefore, this study supports that dsDNA viruses evolved to express enzymes necessary to increase the levels of dNTPs as a mechanism to overcome the restriction induced by SAMHD1 in myeloid cells.
Collapse
Affiliation(s)
- Joseph A. Hollenbaugh
- Department of Microbiology and Immunology, University of Rochester, Rochester, New York, United States of America
- Center for Drug Discovery, The Department of Pediatrics, Emory University, Atlanta, Georgia, United States of America
| | - Peter Gee
- Laboratory of Viral Pathogenesis, Institute for Virus Research, Kyoto University, Kyoto, Japan
| | - Jonathon Baker
- Department of Microbiology and Immunology, University of Rochester, Rochester, New York, United States of America
| | - Michele B. Daly
- Department of Microbiology and Immunology, University of Rochester, Rochester, New York, United States of America
- Center for Drug Discovery, The Department of Pediatrics, Emory University, Atlanta, Georgia, United States of America
| | - Sarah M. Amie
- Department of Microbiology and Immunology, University of Rochester, Rochester, New York, United States of America
| | - Jessica Tate
- Department of Microbiology and Immunology, University of Rochester, Rochester, New York, United States of America
| | - Natsumi Kasai
- Laboratory of Viral Pathogenesis, Institute for Virus Research, Kyoto University, Kyoto, Japan
| | - Yuka Kanemura
- Laboratory of Viral Pathogenesis, Institute for Virus Research, Kyoto University, Kyoto, Japan
| | - Dong-Hyun Kim
- Department of Pharmacy, Kyung-Hee University, Seoul, South Korea
| | - Brian M. Ward
- Department of Microbiology and Immunology, University of Rochester, Rochester, New York, United States of America
- * E-mail: (BW); (YK); (BK)
| | - Yoshio Koyanagi
- Laboratory of Viral Pathogenesis, Institute for Virus Research, Kyoto University, Kyoto, Japan
- * E-mail: (BW); (YK); (BK)
| | - Baek Kim
- Department of Microbiology and Immunology, University of Rochester, Rochester, New York, United States of America
- Center for Drug Discovery, The Department of Pediatrics, Emory University, Atlanta, Georgia, United States of America
- Department of Pharmacy, Kyung-Hee University, Seoul, South Korea
- * E-mail: (BW); (YK); (BK)
| |
Collapse
|
31
|
Abstract
Genetically engineered tumor-selective vaccinia virus (VV) has been demonstrated to be a highly effective oncolytic agent, but immune clearance may limit its therapeutic potential. As previously demonstrated, immunosuppression can lead to significant enhancement of viral recovery and therapeutic effect, but the magnitude of complement-mediated viral inactivation has not been fully elucidated and warrants further investigation. Using fluorescent microscopy and quantitative plaque assays, we have determined complement's key role in viral clearance and its multi-faceted means to pathogen destruction. Complement can lead to direct viral destruction and inhibition of viral uptake into cells, even in the absence of anti-vaccinia antibodies. Our data demonstrate C5 to be integral to the clearance pathway, and its inhibition by Staphylococcal superantigen-like protein leads to a 90-fold and 150-fold enhancement of VV infectivity in both the presence and absence of anti-VV antibodies, respectively. This study suggests that complement inhibition may reduce vaccinia viral neutralization and may be critical to future in vivo work.
Collapse
|
32
|
Reinboth J, Ascierto ML, Chen NG, Zhang Q, Yu YA, Aguilar RJ, Carretero R, Worschech A, Zhao Y, Wang E, Marincola FM, Szalay AA. Correlates between host and viral transcriptional program associated with different oncolytic vaccinia virus isolates. Hum Gene Ther Methods 2012; 23:285-96. [PMID: 23131031 DOI: 10.1089/hgtb.2012.057] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Vaccinia virus (VACV) has emerged as an attractive tool in oncolytic virotherapy. VACV replication efficiency plays a crucial role in the therapeutic outcome. However, little is known about the influence of host factors on viral replication efficiency and permissiveness of a host cell line to infection and oncolysis. In this study, replication of the attenuated VACV GLV-1h68 strain and three wild-type VACV isolates was determined in two autologous human melanoma cell lines (888-MEL and 1936-MEL). Host gene expression and viral gene expression in infected cells were evaluated via respective expression array platforms. Microarray analyses followed by sequential statistical approaches characterized human genes that change specifically due to virus infection. Viral gene transcription correlated with viral replication in a time-dependent manner. A set of human genes revealed strong correlations with the respective viral gene expression. Finally we identified a set of human genes with possible predictive value for viral replication in an independent dataset. The results demonstrate a probable correlation between viral replication, early gene expression, and the respective host response, and thus a possible involvement of human host factors in viral early replication. The characterization of human target genes that influence viral replication could help answer the question of host cell permissiveness to oncolytic virotherapy and provide important information for the development of novel recombinant vaccinia viruses with improved features to enhance replication rate and hence trigger therapeutic outcome.
Collapse
Affiliation(s)
- Jennifer Reinboth
- Genelux Corporation, San Diego Science Center, San Diego, CA 92109, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Reinboth J, Ascierto ML, Chen NG, Zhang Q, Yu YA, Aguilar RJ, Carretero R, Worschech A, Zhao Y, Wang E, Marincola FM, Szalay AA. Correlates between host and viral transcriptional program associated with different oncolytic vaccinia virus isolates. Hum Gene Ther Methods 2012. [DOI: 10.1089/hum.2012.057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
34
|
Zeyaullah M, Patro M, Ahmad I, Ibraheem K, Sultan P, Nehal M, Ali A. Oncolytic viruses in the treatment of cancer: a review of current strategies. Pathol Oncol Res 2012; 18:771-81. [PMID: 22714538 DOI: 10.1007/s12253-012-9548-2] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 05/30/2012] [Indexed: 12/18/2022]
Abstract
Oncolytic viruses are live, replication-competent viruses that replicate selectively in tumor cells leading to the destruction of the tumor cells. Tumor-selective replicating viruses offer appealing advantages over conventional cancer therapy and are promising a new approach for the treatment of human cancer. The development of virotherapeutics is based on several strategies. Virotherapy is not a new concept, but recent technical advances in the genetic modification of oncolytic viruses have improved their tumor specificity, leading to the development of new weapons for the war against cancer. Clinical trials with oncolytic viruses demonstrate the safety and feasibility of an effective virotherapeutic approach. Strategies to overcome potential obstacles and challenges to virotherapy are currently being explored. Systemic administrations of oncolytic viruses will successfully extend novel treatment against a range of tumors. Combination therapy has shown some encouraging antitumor responses by eliciting strong immunity against established cancer.
Collapse
Affiliation(s)
- Md Zeyaullah
- Department of Microbiology, Faculty of Medicine, Omar Al-Mukhtar University, Al-Baida, Libya.
| | | | | | | | | | | | | |
Collapse
|
35
|
|
36
|
Guse K, Cerullo V, Hemminki A. Oncolytic vaccinia virus for the treatment of cancer. Expert Opin Biol Ther 2011; 11:595-608. [DOI: 10.1517/14712598.2011.558838] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
37
|
Abstract
Vaccinia virus (VACV) is arguably the most successful live biotherapeutic agent because of its critical role in the eradication of smallpox, one of the most deadly diseases in human history. VACV has been exploited as an oncolytic therapeutic agent for cancer since 1922. This virus selectively infects and destroys tumor cells, while sparing normal cells, both in cell cultures and in animal models. A certain degree of therapeutic efficacy also has been demonstrated in patients with different types of cancer. In recent years, several strategies have been successfully developed to further improve its tumor selectivity and antitumor efficacy. Oncolytic VACVs carrying imaging genes represent a new treatment strategy that combines tumor site-specific therapeutics with diagnostics (theranostics).
Collapse
Affiliation(s)
- Nanhai G Chen
- Genelux Corporation, San Diego Science Center, San Diego, CA 92109, USA; Genelux Corporation, San Diego Science Center, 3030 Bunker Hill Street, Suite 310, San Diego, CA 92109, USA
| | - Aladar A Szalay
- Rudolf Virchow Center for Experimental Biomedicine, Institute for Biochemistry & Institute for Molecular Infection Biology, University of Würzburg, Am Hubland, D-97074 Würzburg, Germany
- Department of Radiation Oncology, Rebecca & John Moores Comprehensive Cancer Center, University of California, San Diego, CA 92093, USA
| |
Collapse
|
38
|
|
39
|
Antiangiogenic arming of an oncolytic vaccinia virus enhances antitumor efficacy in renal cell cancer models. J Virol 2009; 84:856-66. [PMID: 19906926 DOI: 10.1128/jvi.00692-09] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Oncolytic vaccinia viruses have shown compelling results in preclinical cancer models and promising preliminary safety and antitumor activity in early clinical trials. However, to facilitate systemic application it would be useful to improve tumor targeting and antitumor efficacy further. Here we report the generation of vvdd-VEGFR-1-Ig, a targeted and armed oncolytic vaccinia virus. Tumor targeting was achieved by deletion of genes for thymidine kinase and vaccinia virus growth factor, which are necessary for replication in normal but not in cancer cells. Given the high vascularization typical of kidney cancers, we armed the virus with the soluble vascular endothelial growth factor (VEGF) receptor 1 protein for an antiangiogenic effect. Systemic application of high doses of vvdd-VEGFR-1-Ig resulted in cytokine induction in an immunocompromised mouse model. Upon histopathological analysis, splenic extramedullary hematopoiesis was seen in all virus-injected mice and was more pronounced in the vvdd-VEGFR-1-Ig group. Analysis of the innate immune response after intravenous virus injection revealed high transient and dose-dependent cytokine elevations. When medium and low doses were used for intratumoral or intravenous injection, vvdd-VEGFR-1-Ig exhibited a stronger antitumor effect than the unarmed control. Furthermore, expression of VEGFR-1-Ig was confirmed, and a concurrent antiangiogenic effect was seen. In an immunocompetent model, systemic vvdd-VEGFR-1-Ig exhibited superior antitumor efficacy compared to the unarmed control virus. In conclusion, the targeted and armed vvdd-VEGFR-1-Ig has promising anticancer activity in renal cell cancer models. Extramedullary hematopoiesis may be a sensitive indicator of vaccinia virus effects in mice.
Collapse
|
40
|
Sverdlov ED. Not gene therapy, but genetic surgery-the right strategy to attack cancer. MOLECULAR GENETICS, MICROBIOLOGY AND VIROLOGY : MOLEKULYARNAYA GENETIKA, MIKROBIOLOGIYA I VIRUSOLOGIYA 2009; 24:93-113. [PMID: 32214647 PMCID: PMC7089455 DOI: 10.3103/s089141680903001x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In this review, I will suggest to divide all the approaches united now under common term "gene therapy" into two broad strategies of which the first one uses the methodology of targeted therapy with all its characteristics, but with genes in the role of agents targeted at a certain molecular component(s) presumably crucial for cancer maintenance. In contrast, the techniques of the other strategy are aimed at the destruction of tumors as a whole using the features shared by all cancers, for example relatively fast mitotic cell division or active angiogenesis. While the first strategy is "true" gene therapy, the second one is more like genetic surgery when a surgeon just cuts off a tumor with his scalpel and has no interest in knowing delicate mechanisms of cancer emergence and progression. I will try to substantiate the idea that the last strategy is the only right one, and its simplicity is paradoxically adequate to the super-complexity of tumors that originates from general complexity of cell regulation, strongly disturbed in tumor cells, and especially from the complexity of tumors as evolving cell populations, affecting also their ecological niche formed by neighboring normal cells and tissues. An analysis of the most widely used for such a "surgery" suicide gene/prodrug combinations will be presented in some more details.
Collapse
Affiliation(s)
- E D Sverdlov
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry RAN, Moscow, Russia
| |
Collapse
|
41
|
Zhang H, Takayama K, Zhang L, Uchino J, Harada A, Harada T, Hisasue J, Nakagaki N, Zhou C, Nakanishi Y. Tetracycline-inducible promoter-based conditionally replicative adenoviruses for the control of viral replication. Cancer Gene Ther 2009; 16:415-22. [DOI: 10.1038/cgt.2008.101] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
42
|
Abstract
BACKGROUND Current therapies for multiple myeloma (MM) are not curative, thus novel targeted therapeutics are being developed. One such targeted therapy is oncolytic virotherapy, wherein viruses specifically infect and kill the malignant plasma cells, leaving normal cells intact. OBJECTIVE This review provides an overview of the mechanisms and results of the oncolytic viruses being used to date and discusses the recent advances in the field of virotherapy for MM. METHODS All papers using viruses to treat MM were identified and screened. Only papers describing replicating, oncolytic viruses were reviewed. RESULTS/CONCLUSIONS Several viruses are currently being developed preclinically and clinically to treat MM, including measles virus, vesicular stomatitis virus, coxsackievirus A21 and vaccinia virus. Other viruses are being used preclinically to purge myeloma cells from autologous bone marrow transplants. Efforts to improve myeloma-specific targeting, avoid the antiviral immune response and evaluate combination therapies for MM are ongoing.
Collapse
Affiliation(s)
- Amaalia E Stief
- Experimental Therapeutics, Toronto General Research Institute, 67 College Street, Room 4-408, Toronto, ON, M5G 2M1, Canada
| | | |
Collapse
|
43
|
Prestwich RJ, Errington F, Harrington KJ, Pandha HS, Selby P, Melcher A. Oncolytic viruses: do they have a role in anti-cancer therapy? Clin Med Oncol 2008; 2:83-96. [PMID: 21892269 PMCID: PMC3161683 DOI: 10.4137/cmo.s416] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Oncolytic viruses are replication competent, tumor selective and lyse cancer cells. Their potential for anti-cancer therapy is based upon the concept that selective intratumoral replication will produce a potent anti-tumor effect and possibly bystander or remote cell killing, whilst minimizing normal tissue toxicity. Viruses may be naturally oncolytic or be engineered for oncolytic activity, and possess a host of different mechanisms to provide tumor selectivity. Clinical use of live replicating viruses is associated with a unique set of safety issues. Clinical experience has so far provided evidence of limited efficacy and a favourable toxicity profile. The interaction with the host immune system is complex. An anti-viral immune response may limit efficacy by rapidly clearing the virus. However, virally-induced cell lysis releases tumor associated antigens in a 'dangerous' context, and limited evidence suggests that this can lead to the generation of a specific anti-tumor immune response. Combination therapy with chemotherapy or radiotherapy represents a promising avenue for ongoing translation of oncolytic viruses into clinical practice. Obstacles to therapy include highly effective non-specific host mechanisms to clear virus following systemic delivery, immune-mediated clearance, and intratumoral barriers limiting virus spread. A number of novel strategies are now under investigation to overcome these barriers. This review provides an overview of the potential role of oncolytic viruses, highlighting recent progress towards developing effective therapy and asks if they are a realistic therapeutic option at this stage.
Collapse
Affiliation(s)
- Robin J Prestwich
- Cancer Research UK, St James's University Hospital, Beckett Street, Leeds, LS9 7TF, UK
| | | | | | | | | | | |
Collapse
|
44
|
Chalikonda S, Kivlen MH, O’Malley ME, Dong XD(E, McCart JA, Gorry MC, Yin XY, Brown CK, Zeh HJ, Guo ZS, Bartlett DL. Oncolytic virotherapy for ovarian carcinomatosis using a replication-selective vaccinia virus armed with a yeast cytosine deaminase gene. Cancer Gene Ther 2008; 15:115-25. [PMID: 18084242 PMCID: PMC2975702 DOI: 10.1038/sj.cgt.7701110] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2007] [Revised: 10/09/2007] [Accepted: 11/10/2007] [Indexed: 01/03/2023]
Abstract
In this study, we assessed the ability of a highly tumor-selective oncolytic vaccinia virus armed with a yeast cytosine deaminase gene to infect and lyse human and murine ovarian tumors both in vitro and in vivo. The virus vvDD-CD could infect, replicate in and effectively lyse both human and mouse ovarian cancer cells in vitro. In two different treatment schedules involving either murine MOSEC or human A2780 ovarian carcinomatosis models, regional delivery of vvDD-CD selectively targeted tumor cells and ovarian tissue, effectively delaying the development of either tumor or ascites and leading to significant survival advantages. Oncolytic virotherapy using vvDD-CD in combination with the prodrug 5-fluorocytosine conferred an additional long-term survival advantage upon tumor-bearing immunocompetent mice. These findings demonstrate that a tumor-selective oncolytic vaccinia combined with gene-directed enzyme prodrug therapy is a highly effective strategy for treating advanced ovarian cancers in both syngeneic mouse and human xenograft models. Given the biological safety, tumor selectivity and oncolytic potency of this armed oncolytic virus, this dual therapy merits further investigation as a promising new treatment for metastatic ovarian cancer.
Collapse
Affiliation(s)
- Sricharan Chalikonda
- Department of Surgery, University of Pittsburgh School of Medicine, and the University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, USA
| | - Maryann H. Kivlen
- Surgery Branch, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Mark E. O’Malley
- Department of Surgery, University of Pittsburgh School of Medicine, and the University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, USA
| | - Xiang Da (Eric) Dong
- Department of Surgery, University of Pittsburgh School of Medicine, and the University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, USA
| | - J. Andrea McCart
- Surgery Branch, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Michael C. Gorry
- Department of Surgery, University of Pittsburgh School of Medicine, and the University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, USA
| | - Xiao-Yu Yin
- Department of Surgery, University of Pittsburgh School of Medicine, and the University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, USA
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Charles K. Brown
- Department of Surgery, University of Pittsburgh School of Medicine, and the University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, USA
| | - Herbert J. Zeh
- Department of Surgery, University of Pittsburgh School of Medicine, and the University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, USA
| | - Z. Sheng Guo
- Department of Surgery, University of Pittsburgh School of Medicine, and the University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, USA
| | - David L. Bartlett
- Department of Surgery, University of Pittsburgh School of Medicine, and the University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, USA
- Surgery Branch, National Cancer Institute, NIH, Bethesda, Maryland, USA
| |
Collapse
|
45
|
Hedley D, Ogilvie L, Springer C. Carboxypeptidase-G2-based gene-directed enzyme-prodrug therapy: a new weapon in the GDEPT armoury. Nat Rev Cancer 2007; 7:870-9. [PMID: 17943135 DOI: 10.1038/nrc2247] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Gene-directed enzyme-prodrug therapy (GDEPT) aims to improve the therapeutic ratio (benefit versus toxic side-effects) of cancer chemotherapy. A gene encoding a 'suicide' enzyme is introduced into the tumour to convert a subsequently administered non-toxic prodrug into an active drug selectively in the tumour, but not in normal tissues. Significant effects can now be achieved in vitro and in targeted experimental models, and GDEPT therapies are entering the clinic. Our group has developed a GDEPT system that uses the bacterial enzyme carboxypeptidase G2 to convert nitrogen mustard prodrugs into potent DNA crosslinking agents, and a clinical trial of this system is pending.
Collapse
Affiliation(s)
- Douglas Hedley
- Institute of Cancer Research Haddow Laboratories, 15, Cotswold Road, Sutton, Surrey, UK
| | | | | |
Collapse
|
46
|
Stanford MM, McFadden G. Myxoma virus and oncolytic virotherapy: a new biologic weapon in the war against cancer. Expert Opin Biol Ther 2007; 7:1415-25. [PMID: 17727330 DOI: 10.1517/14712598.7.9.1415] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Oncolytic virotherapy is an innovative alternative to more conventional cancer therapies. The ability of some viruses to specifically target and kill malignant cancerous cells while leaving normal tissue unscathed has opened a large repertoire of new and selective cancer killing therapeutic candidates. Poxviruses, such as vaccinia virus, have a long history of use in humans as live vaccines and have more recently been studied as potential platforms for delivery of immunotherapeutics and attenuated variants of vaccinia have been explored as oncolytic candidates. In contrast, the poxvirus myxoma virus is a novel oncolytic candidate that has no history of use in humans directly, as it has a distinct and absolute host species tropism to lagomorphs (rabbits). Myxoma virus has been recently shown to be able to also selectively infect and kill human tumor cells, a unique tropism that is linked to dysregulated intracellular signalling pathways found in the majority of human cancers. This review outlines the existing knowledge on the tropism of myxoma virus for human cancer cells, as well as preclinical data exhibiting its ability to infect and clear tumors in animal models of cancer. This is an exciting new therapeutic option for treating cancer, and myxoma virus joins a growing group of oncolytic virus candidates that are being developed as a new class of cancer therapies in man.
Collapse
Affiliation(s)
- Marianne M Stanford
- University of Western Ontario, Biotherapeutics Research Group, Robarts Research Institute, Department of Microbiology and Immunology, London, Ontario, N6G 2V4, Canada
| | | |
Collapse
|
47
|
Yang S, Guo ZS, O'Malley ME, Yin X, Zeh HJ, Bartlett DL. A new recombinant vaccinia with targeted deletion of three viral genes: its safety and efficacy as an oncolytic virus. Gene Ther 2007; 14:638-47. [PMID: 17268533 DOI: 10.1038/sj.gt.3302914] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
To enhance further the safety and efficacy of oncolytic vaccinia virus, we have developed a new virus with targeted deletions of three viral genes encoding thymidine kinase and antiapoptotic/host range proteins SPI-1 and SPI-2 (vSPT). Infection of human and murine tumor cell lines yielded nearly equivalent or a log lower virus recovery in comparison to parental viruses. Viral infection activated multiple caspases in cancer cells but not in normal cells, suggesting infected cells may die via different pathways. In tumor-bearing mice, vSPT recovery from MC38 tumor was slightly reduced in comparison to two parental viruses. However, no virus was recovered from the brains and livers of mice injected with vSPT in contrast to control viruses. vSPT demonstrated significantly lower pathogenicity in nude mice. Systemic delivery of vSPT showed significant tumor inhibition in subcutaneous MC38 tumor, human ovarian A2780 and murine ovarian MOSEC carcinomatosis models; however, the tumor inhibition by vSPT was reduced compared with parental viruses. These results demonstrated that although deletion of these three viral genes further enhanced tumor selectivity, it also weakened the oncolytic potency. This study illustrates the complexity of creating a tumor-selective oncolytic virus by deleting multiple viral genes involved in multiple cellular pathways.
Collapse
Affiliation(s)
- S Yang
- University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | | | | | | | | |
Collapse
|
48
|
|
49
|
Thorne SH. Strategies to achieve systemic delivery of therapeutic cells and microbes to tumors. Expert Opin Biol Ther 2007; 7:41-51. [PMID: 17150018 DOI: 10.1517/14712598.7.1.41] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In order to more effectively treat cancer, targeted delivery of therapeutic agents will be needed. The creation of delivery vehicles capable of locating and entering tumors before delivering a therapeutic payload will, therefore, enable the design of more beneficial and less toxic treatment platforms. Although nanoparticles, microbubbles and liposomes may also partially address these issues, the use of biological agents as delivery vehicles presently holds much promise. Through the hijacking of natural pathogen or cell trafficking pathways it is possible to actively target such agents to the tumor; they are then capable of selective replication (multiplying their therapeutic potential) and may be directly cytolytic themselves and/or may be utilized to deliver therapeutic genes. These agents, such as oncolytic viruses, attenuated bacteria and eukaryotic cells (cellular immunotherapeutics and progenitor and stem cells) will be discussed along with the mechanisms employed to deliver them systemically to tumors, including disseminated disease and micrometsastases.
Collapse
Affiliation(s)
- Steve H Thorne
- Stanford University, Bio-X Programme and Department of Pediatrics, Clark Center, California 94305, USA.
| |
Collapse
|
50
|
Stanford MM, Barrett JW, Nazarian SH, Werden S, McFadden G. Oncolytic virotherapy synergism with signaling inhibitors: Rapamycin increases myxoma virus tropism for human tumor cells. J Virol 2006; 81:1251-60. [PMID: 17108021 PMCID: PMC1797522 DOI: 10.1128/jvi.01408-06] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Myxoma virus is a rabbit-specific poxvirus pathogen that also exhibits a unique tropism for human tumor cells and is dramatically oncolytic for human cancer xenografts. Most tumor cell lines tested are permissive for myxoma infection in a fashion intimately tied to the activation state of Akt kinase. A host range factor of myxoma virus, M-T5, directly interacts with Akt and mediates myxoma virus tumor cell tropism. mTOR is a regulator of cell growth and metabolism downstream of Akt and is specifically inhibited by rapamycin. We report that treatment of nonpermissive human tumor cell lines, which normally restrict myxoma virus replication, with rapamycin dramatically increased virus tropism and spread in vitro. This increased myxoma replication is concomitant with global effects on mTOR signaling, specifically, an increase in Akt kinase. In contrast to the effects on human cancer cells, rapamycin does not increase myxoma virus replication in rabbit cell lines or permissive human tumor cell lines with constitutively active Akt. This indicates that rapamycin increases the oncolytic capacity of myxoma virus for human cancer cells by reconfiguring the internal cell signaling environment to one that is optimal for productive virus replication and suggests the possibility of a potentially therapeutic synergism between kinase signaling inhibitors and oncolytic poxviruses for cancer treatment.
Collapse
Affiliation(s)
- Marianne M Stanford
- Biotherapeutics Research Group, Robarts Research Institute, Department of Microbiology and Immunology, University of Western Ontario, London, Ontario N6G 2V4, Canada
| | | | | | | | | |
Collapse
|