1
|
Jafari F, Javdansirat S, Sanaie S, Naseri A, Shamekh A, Rostamzadeh D, Dolati S. Osteosarcoma: A comprehensive review of management and treatment strategies. Ann Diagn Pathol 2020; 49:151654. [PMID: 33130384 DOI: 10.1016/j.anndiagpath.2020.151654] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 07/09/2020] [Accepted: 10/20/2020] [Indexed: 01/25/2023]
Abstract
Osteosarcoma, a bone cancer usually seen in children and young adults, is generally a high-grade malignancy presented by extreme metastases to the lungs. Osteosarcoma has a tendency for appearing in bones with rapid growth rate. The etiology of osteosarcoma is multifaceted and poorly understood. A molecular consideration of this disease will lead to a directed tumor treatment. The present treatment for osteosarcoma comprises of an arrangement of systemic chemotherapy and wide surgical resection. Survival rate is increased by the progress of destructive systemic chemotherapies. So, the development of new treatment approaches for metastatic osteosarcoma is essential. Immunomodulation has been used in clinical settings. Through targeting surface antigens expressed on tumor cells, particular antibodies and exploitation of cellular immunotherapy against sarcomas have been confirmed to be effective as cancer therapeutics. In this article, we have reviewed epidemiology, etiology, pathogenesis, diagnosis, and treatment of osteosarcoma and we have focused on different methods of immunotherapy including vaccines, cell-based immunotherapy, cytokines, and monoclonal antibodies.
Collapse
Affiliation(s)
- Farzaneh Jafari
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeed Javdansirat
- Clinical Research development unit Center, Beheshti Hospital, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Sarvin Sanaie
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amirreza Naseri
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Shamekh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Davood Rostamzadeh
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Sanam Dolati
- Physical Medicine and Rehabilitation Research Center, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
2
|
Wedekind MF, Cripe TP. Oncolytic Viruses and Their Potential as a Therapeutic Opportunity in Osteosarcoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1258:77-89. [PMID: 32767235 DOI: 10.1007/978-3-030-43085-6_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Osteosarcoma remains an unmet medical need. Oncolytic viruses are gaining traction as novel cancer therapeutics. These viruses are either naturally nonpathogenic or engineered to be safe by specific genetic deletions yet retain the ability to infect and kill human cancer cells and elicit anticancer immunity. Some versions are being specifically designed and tested in patients with osteosarcoma, though due to their generalized mechanism of action most are being tested in patients across a broad range of cancer types. The activity of these viruses is impacted not only by the susceptibility of tumor cells to infection but also by the tumor microenvironment (TME) and by tumor immunogenicity. Here we review the field of oncolytic viruses with a particular emphasis on highlighting any available data in preclinical osteosarcoma models or in patients with osteosarcoma. While in general the viruses have been shown safe to administer to patients by a variety of routes, their therapeutic efficacy to date has been limited. Given the low rate of adverse events and the likely absence of long-term side effects, the utility of oncolytic viruses will most likely be realized when used in combination with other agents.
Collapse
Affiliation(s)
| | - Timothy P Cripe
- Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
3
|
Recombinant viruses with other anti-cancer therapeutics: a step towards advancement of oncolytic virotherapy. Cancer Gene Ther 2018; 25:216-226. [PMID: 29735993 DOI: 10.1038/s41417-018-0018-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 02/23/2018] [Accepted: 03/02/2018] [Indexed: 01/15/2023]
Abstract
Cancer as a disease is a multifaceted foe which sometimes succumbs to the prescribed treatment and sometimes develops resistance against various therapies. Conventional cancer therapies suffer from many limitations, the least of which is their specificity and systemic side effects. In a majority of cases, acquired mutations render the cancer cells resistant to therapy and lower the prognostic outcome. In the constant effort to devise a therapeutic moiety which can comprehensively eliminate cancer cells, oncolytic viruses provide an attractive avenue as they selectively infect and lyse cancer cells sparing normal cells from their effects. Viruses can be engineered for their host specificity and toxicity as a promising anti-cancer tool. As it is essential to devise a strategy to address all targets involved in cancer development and progression, the idea of using oncolytic viruses with enhanced anti-cancer activity through arming with foreign genes gained merit and is showing promising advent in clinical studies. The use of oncolytic viruses as an agent of combination therapy for cancer treatment also gained much attention in the recent past. This review focuses on the emerging role of oncolytic viruses as vital components of anti-cancer regimen presenting a new dimension in an ever-changing cancer therapy scenario.
Collapse
|
4
|
Oncolytic vaccinia virus combined with radiotherapy induces apoptotic cell death in sarcoma cells by down-regulating the inhibitors of apoptosis. Oncotarget 2018; 7:81208-81222. [PMID: 27783991 PMCID: PMC5348387 DOI: 10.18632/oncotarget.12820] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 10/11/2016] [Indexed: 12/13/2022] Open
Abstract
Advanced extremity melanoma and sarcoma present a significant therapeutic challenge, requiring multimodality therapy to treat or even palliate disease. These aggressive tumours are relatively chemo-resistant, therefore new treatment approaches are urgently required. We have previously reported on the efficacy of oncolytic virotherapy (OV) delivered by isolated limb perfusion. In this report, we have improved therapeutic outcomes by combining OV with radiotherapy. In vitro, the combination of oncolytic vaccinia virus (GLV-1h68) and radiotherapy demonstrated synergistic cytotoxicity. This effect was not due to increased viral replication, but mediated through induction of intrinsic apoptosis. GLV-1h68 therapy downregulated the anti-apoptotic BCL-2 proteins (MCL-1 and BCL-XL) and the downstream inhibitors of apoptosis, resulting in cleavage of effector caspases 3 and 7. In an in vivo ILP model, the combination of OV and radiotherapy significantly delayed tumour growth and prolonged survival compared to single agent therapy. These data suggest that the virally-mediated down-regulation of anti-apoptotic proteins may increase the sensitivity of tumour cells to the cytotoxic effects of ionizing radiation. Oncolytic virotherapy represents an exciting candidate for clinical development when delivered by ILP. Its ability to overcome anti-apoptotic signals within tumour cells points the way to further development in combination with conventional anti-cancer therapies.
Collapse
|
5
|
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
|
6
|
Waters AM, Stafman LL, Garner EF, Mruthyunjayappa S, Stewart JE, Friedman GK, Coleman JM, Markert JM, Gillespie GY, Beierle EA. Effect of Repeat Dosing of Engineered Oncolytic Herpes Simplex Virus on Preclinical Models of Rhabdomyosarcoma. Transl Oncol 2016; 9:419-430. [PMID: 27751346 PMCID: PMC5067929 DOI: 10.1016/j.tranon.2016.07.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 07/14/2016] [Accepted: 07/18/2016] [Indexed: 12/16/2022] Open
Abstract
Rhabdomyosarcoma (RMS), a tumor of skeletal muscle origin, is the most common sarcoma of childhood. Despite multidrug chemotherapy regimens, surgical intervention, and radiation treatment, outcomes remain poor, especially in advanced disease, and novel therapies are needed for the treatment of these aggressive malignancies. Genetically engineered oncolytic viruses, such as herpes simplex virus-1 (HSV), are currently being explored as treatments for pediatric tumors. M002, an oncolytic HSV, has both copies of the γ134.5 gene deleted, enabling replication in tumor cells but thwarting infection of normal, postmitotic cells. We hypothesized that M002 would infect human RMS tumor cells and lead to decreased tumor cell survival in vitro and impede tumor growth in vivo. In the current study, we demonstrated that M002 could infect, replicate in, and decrease cell survival in both embryonal (ERMS) and alveolar rhabdomyosarcoma (ARMS) cells. Additionally, M002 reduced xenograft tumor growth and increased animal survival in both ARMS and ERMS. Most importantly, we showed for the first time that repeated dosing of oncolytic virus coupled with low-dose radiation provided improved tumor response in RMS. These findings provide support for the clinical investigation of oncolytic HSV in pediatric RMS.
Collapse
Affiliation(s)
- Alicia M Waters
- Department of Surgery, Division of Pediatric Surgery, University of Alabama, Birmingham, Birmingham, AL, USA 35233
| | - Laura L Stafman
- Department of Surgery, Division of Pediatric Surgery, University of Alabama, Birmingham, Birmingham, AL, USA 35233
| | - Evan F Garner
- Department of Surgery, Division of Pediatric Surgery, University of Alabama, Birmingham, Birmingham, AL, USA 35233
| | - Smitha Mruthyunjayappa
- Department of Surgery, Division of Pediatric Surgery, University of Alabama, Birmingham, Birmingham, AL, USA 35233
| | - Jerry E Stewart
- Department of Surgery, Division of Pediatric Surgery, University of Alabama, Birmingham, Birmingham, AL, USA 35233
| | - Gregory K Friedman
- Department of Pediatrics, Division of Hematology/Oncology, University of Alabama, Birmingham, Birmingham, AL, USA 35233
| | - Jennifer M Coleman
- Department of Surgery, Division of Neurosurgery, University of Alabama, Birmingham, Birmingham, AL, USA 35233
| | - James M Markert
- Department of Surgery, Division of Neurosurgery, University of Alabama, Birmingham, Birmingham, AL, USA 35233
| | - G Yancey Gillespie
- Department of Surgery, Division of Neurosurgery, University of Alabama, Birmingham, Birmingham, AL, USA 35233
| | - Elizabeth A Beierle
- Department of Surgery, Division of Pediatric Surgery, University of Alabama, Birmingham, Birmingham, AL, USA 35233.
| |
Collapse
|
7
|
Wang B, Yan X, Guo Q, Li Y, Zhang H, Xie JS, Meng X. Deficiency of caspase 3 in tumor xenograft impairs therapeutic effect of measles virus Edmoston strain. Oncotarget 2016; 6:16019-30. [PMID: 25909216 PMCID: PMC4599253 DOI: 10.18632/oncotarget.3496] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 03/07/2015] [Indexed: 11/25/2022] Open
Abstract
The oncolytic measles virus Edmonston (MV-Edm) strain shows considerable oncolytic activity against a variety of human tumors. In this study, we report MV-Edm is able to trigger apoptosis pathways in infected tumor cells and elucidate the roles of cellular apoptosis in the whole oncolytic process. We also show that activated caspase 3, a key executioner of apoptosis, plays key roles in the oncolytic virotherapy. Activated caspase 3 can accelerate viral replication in cervical cancer cells and enhance the killing effects of the virus. Deficiency of caspase 3 either in tumor cells or in tumor xenograft significantly desensitized tumor to oncolysis with MV-Edm. In the infected cells, caspase 3 regulates interferon α release, which can inhibit viral replication in neighboring tumor cells. We propose that caspase-3 activation enhances the oncolytic effects of MV-Edm, thus inhibiting tumor growth in mice.
Collapse
Affiliation(s)
- Biao Wang
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences of China Medical University Shenyang, P.R. China
| | - Xu Yan
- Department of Prosthodontics, School of Stomatology, China Medical University, Shenyang, P.R. China
| | - Qingguo Guo
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences of China Medical University Shenyang, P.R. China
| | - Yan Li
- Department of Oncology, Tumour Angiogenesis and Microenvironment Laboratory (TAML), First Affiliated Hospital, Liaoning Medical College, Jinzhou, P.R. China
| | - Haiyan Zhang
- Department of Geriatrics, The First Affiliated Hospital, China Medical University, Shenyang, P.R. China
| | - Ji Sheng Xie
- Department of Ecsomatics, Youjiang Medical College for Nationalities, Baise City, P.R. China
| | - Xin Meng
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences of China Medical University Shenyang, P.R. China
| |
Collapse
|
8
|
Zonov E, Kochneva G, Yunusova A, Grazhdantseva A, Richter V, Ryabchikova E. Features of the Antitumor Effect of Vaccinia Virus Lister Strain. Viruses 2016; 8:E20. [PMID: 26771631 PMCID: PMC4728580 DOI: 10.3390/v8010020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 12/30/2015] [Accepted: 01/06/2016] [Indexed: 02/06/2023] Open
Abstract
Oncolytic abilities of vaccinia virus (VACV) served as a basis for the development of various recombinants for treating cancer; however, "natural" oncolytic properties of the virus are not examined in detail. Our study was conducted to know how the genetically unmodified L-IVP strain of VACV produces its antitumor effect. Human A431 carcinoma xenografts in nude mice and murine Ehrlich carcinoma in C57Bl mice were used as targets for VACV, which was injected intratumorally. A set of virological methods, immunohistochemistry, light and electron microscopy was used in the study. We found that in mice bearing A431 carcinoma, the L-IVP strain was observed in visceral organs within two weeks, but rapidly disappeared from the blood. The L-IVP strain caused decrease of sizes in both tumors, however, in different ways. Direct cell destruction by replicating virus plays a main role in regression of A431 carcinoma xenografts, while in Ehrlich carcinoma, which poorly supported VACV replication, the virus induced decrease of mitoses by pushing tumor cells into S-phase of cell cycle. Our study showed that genetically unmodified VACV possesses at least two mechanisms of antitumor effect: direct destruction of tumor cells and suppression of mitoses in tumor cells.
Collapse
Affiliation(s)
- Evgeniy Zonov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences (ICBFM SB RAS), 8 Lavrentiev Avenue, Novosibirsk 630090, Russia.
| | - Galina Kochneva
- State Research Center of Virology and Biotechnology "Vector", Koltsovo 630559, Russia.
| | - Anastasiya Yunusova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences (ICBFM SB RAS), 8 Lavrentiev Avenue, Novosibirsk 630090, Russia.
| | | | - Vladimir Richter
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences (ICBFM SB RAS), 8 Lavrentiev Avenue, Novosibirsk 630090, Russia.
| | - Elena Ryabchikova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences (ICBFM SB RAS), 8 Lavrentiev Avenue, Novosibirsk 630090, Russia.
| |
Collapse
|
9
|
Wan J, Zhang X, Liu T, Zhang X. Strategies and developments of immunotherapies in osteosarcoma. Oncol Lett 2015; 11:511-520. [PMID: 26834853 DOI: 10.3892/ol.2015.3962] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 10/27/2015] [Indexed: 12/14/2022] Open
Abstract
Osteosarcoma (OS) is a frequently observed primary malignant tumor. Current therapy for osteosarcoma consists of comprehensive treatment. The long-term survival rate of patients exhibiting nonmetastatic OS varies between 65-70%. However, a number of OS cases have been observed to be resistant to currently used therapies, leading to disease recurrence and lung metastases, which are the primary reasons leading to patient mortality. In the present review, a number of pieces of evidence provide support for the potential uses of immunotherapy, including immunomodulation and vaccine therapy, for the eradication of tumors via upregulation of the immune response. Adoptive T-cell therapy and oncolytic virotherapy have been used to treat OS and resulted in objective responses. Immunologic checkpoint blockade and targeted therapy are also potentially promising therapeutic tools. Immunotherapy demonstrates significant promise with regard to improving the outcomes for patients exhibiting OS.
Collapse
Affiliation(s)
- Jia Wan
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Xianghong Zhang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Tang Liu
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Xiangsheng Zhang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
10
|
Abstract
Current standard treatments of cancer can prolong survival of many cancer patients but usually do not effectively cure the disease. Oncolytic virotherapy is an emerging therapeutic for the treatment of cancer that exploits replication-competent viruses to selectively infect and destroy cancerous cells while sparing normal cells and tissues. Clinical and/or preclinical studies on oncolytic viruses have revealed that the candidate viruses being tested in trials are remarkably safe and offer potential for treating many classes of currently incurable cancers. Among these candidates are vaccinia and myxoma viruses, which belong to the family Poxviridae and possess promising oncolytic features. This article describes poxviruses that are being developed for oncolytic virotherapy and summarizes the outcomes of both clinical and preclinical studies. Additionally, studies demonstrating superior efficacy when poxvirus oncolytic virotherapy is combined with conventional therapies are described.
Collapse
Affiliation(s)
- Winnie M. Chan
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, Florida 32610
| | - Grant McFadden
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, Florida 32610
| |
Collapse
|
11
|
Pencavel TD, Wilkinson MJ, Mansfield DC, Khan AA, Seth R, Karapanagiotou EM, Roulstone V, Aguilar RJ, Chen NG, Szalay AA, Hayes AJ, Harrington KJ. Isolated limb perfusion with melphalan, tumour necrosis factor-alpha and oncolytic vaccinia virus improves tumour targeting and prolongs survival in a rat model of advanced extremity sarcoma. Int J Cancer 2014; 136:965-76. [DOI: 10.1002/ijc.29059] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 05/08/2014] [Indexed: 11/06/2022]
Affiliation(s)
- Tim D. Pencavel
- Targeted Therapy Team, The Institute of Cancer Research; London United Kingdom
- Department of Academic Surgery; Sarcoma/Melanoma Unit, Royal Marsden Hospital NHS Foundation Trust; London United Kingdom
| | - Michelle J. Wilkinson
- Targeted Therapy Team, The Institute of Cancer Research; London United Kingdom
- Department of Academic Surgery; Sarcoma/Melanoma Unit, Royal Marsden Hospital NHS Foundation Trust; London United Kingdom
| | - David C. Mansfield
- Targeted Therapy Team, The Institute of Cancer Research; London United Kingdom
| | - Aadil A. Khan
- Targeted Therapy Team, The Institute of Cancer Research; London United Kingdom
| | - Rohit Seth
- Targeted Therapy Team, The Institute of Cancer Research; London United Kingdom
| | | | - Victoria Roulstone
- Targeted Therapy Team, The Institute of Cancer Research; London United Kingdom
| | | | - Nanhai G. Chen
- Genelux Corporation, San Diego Science Center; San Diego CA
- Department of Radiation Oncology, Rebecca and John Moores Comprehensive Cancer Center; University of California; San Diego CA
| | - Aladar A. Szalay
- Genelux Corporation, San Diego Science Center; San Diego CA
- Department of Radiation Oncology, Rebecca and John Moores Comprehensive Cancer Center; University of California; San Diego CA
- Department of Biochemistry, Rudolf Virchow Center for Experimental Biomedicine; University of Würzberg; Am Hubland Germany
- Institute for Molecular Infection Biology; University of Würzberg; Am Hubland Germany
| | - Andrew J. Hayes
- Department of Academic Surgery; Sarcoma/Melanoma Unit, Royal Marsden Hospital NHS Foundation Trust; London United Kingdom
| | - Kevin J. Harrington
- Targeted Therapy Team, The Institute of Cancer Research; London United Kingdom
| |
Collapse
|
12
|
Hofmann E, Weibel S, Szalay AA. Combination treatment with oncolytic Vaccinia virus and cyclophosphamide results in synergistic antitumor effects in human lung adenocarcinoma bearing mice. J Transl Med 2014; 12:197. [PMID: 25030093 PMCID: PMC4105246 DOI: 10.1186/1479-5876-12-197] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 07/10/2014] [Indexed: 12/27/2022] Open
Abstract
Background The capacity of the recombinant Vaccinia virus GLV-1h68 as a single agent to efficiently treat different human or canine cancers has been shown in several preclinical studies. Currently, its human safety and efficacy are investigated in phase I/II clinical trials. In this study we set out to evaluate the oncolytic activity of GLV-1h68 in the human lung adenocarcinoma cell line PC14PE6-RFP in cell cultures and analyzed the antitumor potency of a combined treatment strategy consisting of GLV-1h68 and cyclophosphamide (CPA) in a mouse model of PC14PE6-RFP lung adenocarcinoma. Methods PC14PE6-RFP cells were treated in cell culture with GLV-1h68. Viral replication and cell survival were determined by plaque assays and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays, respectively. Subcutaneously implanted PC14PE6-RFP xenografts were treated by systemic injection of GLV-1h68, CPA or a combination of both. Tumor growth and viral biodistribution were monitored and immune-related antigen profiling of tumor lysates was performed. Results GLV-1h68 efficiently infected, replicated in and lysed human PC14PE6-RFP cells in cell cultures. PC14PE6-RFP tumors were efficiently colonized by GLV-1h68 leading to much delayed tumor growth in PC14PE6-RFP tumor-bearing nude mice. Combination treatment with GLV-1h68 and CPA significantly improved the antitumor efficacy of GLV-1h68 and led to an increased viral distribution within the tumors. Pro-inflammatory cytokines and chemokines were distinctly elevated in tumors of GLV-1h68-treated mice. Factors expressed by endothelial cells or present in the blood were decreased after combination treatment. A complete loss in the hemorrhagic phenotype of the PC14PE6-RFP tumors and a decrease in the number of blood vessels after combination treatment could be observed. Conclusions CPA and GLV-1h68 have synergistic antitumor effects on PC14PE6-RFP xenografts. We strongly suppose that in the PC14PE6-RFP model the enhanced tumor growth inhibition achieved by combining GLV-1h68 with CPA is due to an effect on the vasculature rather than an immunosuppressive action of CPA. These results provide evidence to support further preclinical studies of combining GLV-1h68 and CPA in other highly angiogenic tumor models. Moreover, data presented here demonstrate that CPA can be combined successfully with GLV-1h68 based oncolytic virus therapy and therefore might be promising as combination therapy in human clinical trials.
Collapse
Affiliation(s)
| | | | - Aladar A Szalay
- Department of Biochemistry, Biocenter, University of Wuerzburg, D-97074 Wuerzburg, Germany.
| |
Collapse
|
13
|
Liu YP, Wang J, Avanzato VA, Bakkum-Gamez JN, Russell SJ, Bell JC, Peng KW. Oncolytic vaccinia virotherapy for endometrial cancer. Gynecol Oncol 2014; 132:722-9. [PMID: 24434058 PMCID: PMC3977925 DOI: 10.1016/j.ygyno.2014.01.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Revised: 01/08/2014] [Accepted: 01/09/2014] [Indexed: 01/13/2023]
Abstract
OBJECTIVE Oncolytic virotherapy is a promising modality in endometrial cancer (EC) therapy. In this study, we compared the efficacy of the Copenhagen and Wyeth strains of oncolytic vaccinia virus (VV) incorporating the human thyroidal sodium iodide symporter (hNIS) as a reporter gene (VVNIS-C and VVNIS-W) in EC. METHODS Infectivity of VVNIS-C and VVNIS-W in type I (HEC1A, Ishikawa, KLE, RL95-2, and AN3 CA) and type II (ARK-1, ARK-2, and SPEC-2) human EC cell lines was evaluated. Athymic mice with ARK-2 or AN3 CA xenografts were treated with one intravenous dose of VVNIS-C or VVNIS-W. Tumor regression and in vivo infectivity were monitored via NIS expression using SPECT-CT imaging. RESULTS All EC cell lines except KLE were susceptible to infection and killing by VVNIS-C and VVNIS-W in vitro. VVNIS-C had higher infectivity and oncolytic activity than VVNIS-W in all cell lines, most notably in AN3 CA. Intravenous VVNIS-C was more effective at controlling AN3 CA xenograft growth than VVNIS-W, while both VVNIS-C and VVNIS-W ceased tumor growth and induced tumor regression in 100% of mice bearing ARK-2 xenografts. CONCLUSION Overall, VVNIS-C has more potent oncolytic viral activity than VVSIN-W in EC. VV appears to be most active in type II EC. Novel therapies are needed for the highly lethal type II EC histologies and further development of a VV clinical trial in type II EC is warranted.
Collapse
Affiliation(s)
- Yu-Ping Liu
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Jiahu Wang
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON KlY 4E9, Canada
| | - Victoria A Avanzato
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA; Pennsylvania State University, State College, PA, USA
| | | | - Stephen J Russell
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA; Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - John C Bell
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON KlY 4E9, Canada
| | - Kah-Whye Peng
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA; Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
14
|
Radons J. Inflammatory stress and sarcomagenesis: a vicious interplay. Cell Stress Chaperones 2014; 19:1-13. [PMID: 24046208 PMCID: PMC3857425 DOI: 10.1007/s12192-013-0449-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 07/15/2013] [Accepted: 07/17/2013] [Indexed: 12/15/2022] Open
Abstract
Chronic inflammation represents one of the hallmarks of cancer, but its role in sarcomagenesis has long been overlooked. Sarcomas are a rare and heterogeneous group of tumors of mesenchymal origin accounting for less than 1 % of cancers in adults but 21 % of cancers in the pediatric population. Sarcomas are associated with bad prognosis, and their management requires a multidisciplinary team approach. Several lines of evidence indicate that inflammation has been implicated in sarcomagenesis leading to the activation of the key transcription factors HIF-1, NF- κB, and STAT-3 involved in a complex inflammatory network. In the past years, an increasing number of new targets have been identified in the treatment of sarcomas leading to the development of new drugs that aim to interrupt the vicious connection between inflammation and sarcomagenesis. This article makes a brief overview of preclinical and clinical evidence of the molecular pathways involved in the inflammatory stress response in sarcomagenesis and the most targeted therapies.
Collapse
Affiliation(s)
- Jürgen Radons
- multimmune GmbH c/o Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Straße 22, 81675, Munich, Germany,
| |
Collapse
|
15
|
Oncolytic Viruses for Potential Osteosarcoma Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 804:259-83. [DOI: 10.1007/978-3-319-04843-7_14] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
16
|
Radons J. The role of inflammation in sarcoma. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 816:259-313. [PMID: 24818727 DOI: 10.1007/978-3-0348-0837-8_11] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Sarcomas encompass a heterogenous group of tumors with diverse pathologically and clinically overlapping features. It is a rarely curable disease, and their management requires a multidisciplinary team approach. Chronic inflammation has emerged as one of the hallmarks of tumors including sarcomas. Classical inflammation-associated sarcomas comprise the inflammatory malignant fibrous histiocytoma and Kaposi sarcoma. The identification of specific chromosomal translocations and important intracellular signaling pathways such as Ras/Raf/MAPK, insulin-like growth factor, PI3K/AKT/mTOR, sonic hedgehog and Notch together with the increasing knowledge of angiogenesis has led to development of targeted therapies that aim to interrupt these pathways. Innovative agents like oncolytic viruses opened the way to design new therapeutic options with encouraging findings. Preclinical evidence also highlights the therapeutic potential of anti-inflammatory nutraceuticals as they can inhibit multiple pathways while being less toxic. This chapter gives an overview of actual therapeutic standards, newest evidence-based studies and exciting options for targeted therapies in sarcomas.
Collapse
Affiliation(s)
- Jürgen Radons
- Department of Radiotherapy and Radiation Oncology, Klinikum rechts der Isar, Technische Universität München, Ismaninger Straße 22, 81675, Munich, Germany,
| |
Collapse
|
17
|
Lun X, Ruan Y, Jayanthan A, Liu DJ, Singh A, Trippett T, Bell J, Forsyth P, Johnston RN, Narendran A. Double-deleted vaccinia virus in virotherapy for refractory and metastatic pediatric solid tumors. Mol Oncol 2013; 7:944-54. [PMID: 23816608 DOI: 10.1016/j.molonc.2013.05.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 05/27/2013] [Accepted: 05/28/2013] [Indexed: 10/26/2022] Open
Abstract
BACKGROUND Previous studies have shown successful antitumor effects of systemically delivered double-deleted vaccinia virus (vvDD) against a number of adult tumor models, including glioma, colon and ovarian cancers. The purpose of this study was to investigate the oncolytic potential of vvDD against a panel of cell lines representative of pediatric solid tumors that are currently difficult to cure. METHODS Cell lines derived from central nervous system atypical teratoid rhabdoid tumor (AT/RT) (BT12, BT16 and KCCF1), sarcoma (143B, HOS, RD and RH30), and neuroblastoma (SKNAS, SKNBE2, IMR-5 and IMR-32) were examined for vvDD mediated cytotoxicity defined by virus expansion followed by loss of tumor cell viability. The normal human fibroblast cell line HS68 was used as a control. Next, relevant orthotopic, subcutaneous and lung metastasis xenograft models were treated with intravenous doses of live vvDD or killed virus controls (DV). Tumor growth inhibition and viral replication were quantified and survival outcomes of these animals were assessed. RESULTS vvDD was able to infect and kill nine of eleven of the pediatric tumor cells (81.8%) in vitro. In xenograft models, intravenous administration of a single dose of vvDD significantly inhibited the growth of tumors and prolonged the survival of intracranial and metastatic tumors. CONCLUSIONS Oncolytic vvDD administered i.v. shows activity in preclinical models of pediatric malignancies that are resistant to many currently available treatments. Our data support further evaluation of vvDD virotherapy for refractory pediatric solid tumors.
Collapse
Affiliation(s)
- Xueqing Lun
- Pediatric Oncology Experimental Therapeutics Investigators Consortium (POETIC) Laboratory for Pre-Clinical and Drug Discovery Studies, University of Calgary, Canada and Division of Pediatric Oncology, Alberta Children's Hospital, Calgary, Alberta, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Growth inhibition of different human colorectal cancer xenografts after a single intravenous injection of oncolytic vaccinia virus GLV-1h68. J Transl Med 2013; 11:79. [PMID: 23531320 PMCID: PMC3621142 DOI: 10.1186/1479-5876-11-79] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Accepted: 03/20/2013] [Indexed: 12/18/2022] Open
Abstract
Background Despite availability of efficient treatment regimens for early stage colorectal cancer, treatment regimens for late stage colorectal cancer are generally not effective and thus need improvement. Oncolytic virotherapy using replication-competent vaccinia virus (VACV) strains is a promising new strategy for therapy of a variety of human cancers. Methods Oncolytic efficacy of replication-competent vaccinia virus GLV-1h68 was analyzed in both, cell cultures and subcutaneous xenograft tumor models. Results In this study we demonstrated for the first time that the replication-competent recombinant VACV GLV-1h68 efficiently infected, replicated in, and subsequently lysed various human colorectal cancer lines (Colo 205, HCT-15, HCT-116, HT-29, and SW-620) derived from patients at all four stages of disease. Additionally, in tumor xenograft models in athymic nude mice, a single injection of intravenously administered GLV-1h68 significantly inhibited tumor growth of two different human colorectal cell line tumors (Duke’s type A-stage HCT-116 and Duke’s type C-stage SW-620), significantly improving survival compared to untreated mice. Expression of the viral marker gene ruc-gfp allowed for real-time analysis of the virus infection in cell cultures and in mice. GLV-1h68 treatment was well-tolerated in all animals and viral replication was confined to the tumor. GLV-1h68 treatment elicited a significant up-regulation of murine immune-related antigens like IFN-γ, IP-10, MCP-1, MCP-3, MCP-5, RANTES and TNF-γ and a greater infiltration of macrophages and NK cells in tumors as compared to untreated controls. Conclusion The anti-tumor activity observed against colorectal cancer cells in these studies was a result of direct viral oncolysis by GLV-1h68 and inflammation-mediated innate immune responses. The therapeutic effects occurred in tumors regardless of the stage of disease from which the cells were derived. Thus, the recombinant vaccinia virus GLV-1h68 has the potential to treat colorectal cancers independently of the stage of progression.
Collapse
|
19
|
Gholami S, Chen CH, Belin LJ, Lou E, Fujisawa S, Antonacci C, Carew A, Chen NG, De Brot M, Zanzonico PB, Szalay AA, Fong Y. Vaccinia virus GLV-1h153 is a novel agent for detection and effective local control of positive surgical margins for breast cancer. Breast Cancer Res 2013; 15:R26. [PMID: 23506710 PMCID: PMC3672815 DOI: 10.1186/bcr3404] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 02/13/2013] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Surgery is currently the definitive treatment for early-stage breast cancer. However, the rate of positive surgical margins remains unacceptably high. The human sodium iodide symporter (hNIS) is a naturally occurring protein in human thyroid tissue, which enables cells to concentrate radionuclides. The hNIS has been exploited to image and treat thyroid cancer. We therefore investigated the potential of a novel oncolytic vaccinia virus GLV1h-153 engineered to express the hNIS gene for identifying positive surgical margins after tumor resection via positron emission tomography (PET). Furthermore, we studied its role as an adjuvant therapeutic agent in achieving local control of remaining tumors in an orthotopic breast cancer model. METHODS GLV-1h153, a replication-competent vaccinia virus, was tested against breast cancer cell lines at various multiplicities of infection (MOIs). Cytotoxicity and viral replication were determined. Mammary fat pad tumors were generated in athymic nude mice. To determine the utility of GLV-1h153 in identifying positive surgical margins, 90% of the mammary fat pad tumors were surgically resected and subsequently injected with GLV-1h153 or phosphate buffered saline (PBS) in the surgical wound. Serial Focus 120 microPET images were obtained six hours post-tail vein injection of approximately 600 μCi of 124I-iodide. RESULTS Viral infectivity, measured by green fluorescent protein (GFP) expression, was time- and concentration-dependent. All cell lines showed less than 10% of cell survival five days after treatment at an MOI of 5. GLV-1h153 replicated efficiently in all cell lines with a peak titer of 27 million viral plaque forming units (PFU) ( <10,000-fold increase from the initial viral dose ) by Day 4. Administration of GLV-1h153 into the surgical wound allowed positive surgical margins to be identified via PET scanning. In vivo, mean volume of infected surgically resected residual tumors four weeks after treatment was 14 mm3 versus 168 mm3 in untreated controls (P < 0.05). CONCLUSIONS This is the first study to our knowledge to demonstrate a novel vaccinia virus carrying hNIS as an imaging tool in identifying positive surgical margins of breast cancers in an orthotopic murine model. Moreover, our results suggest that GLV-1h153 is a promising therapeutic agent in achieving local control for positive surgical margins in resected breast tumors.
Collapse
|
20
|
Innate immune defense defines susceptibility of sarcoma cells to measles vaccine virus-based oncolysis. J Virol 2013; 87:3484-501. [PMID: 23302892 DOI: 10.1128/jvi.02106-12] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The oncolytic potential of measles vaccine virus (MeV) has been demonstrated in several tumor entities. Here, we investigated the susceptibility of eight sarcoma cell lines to MeV-mediated oncolysis and found five to be susceptible, whereas three proved to be resistant. In the MeV-resistant cell lines, we often observed an inhibition of viral replication along with a strong upregulation of the intracellular virus-sensing molecule RIG-I and of the interferon (IFN)-stimulated gene IFIT1. Not only expression of IFIT1 but also phosphorylation of IFN-stimulated Stat1 took place rapidly and were found to be persistent over time. In contrast, susceptible cell lines showed a much weaker, delayed, or completely missing expression of IFIT1 as well as a delayed or only transient phosphorylation of Stat1, whereas exogenic stimulation with beta interferon (IFN-β) resulted in a comparable profound activation of Stat1 and expression of IFIT1 in all cell lines. Pretreatment with IFN-β rendered three of the susceptible cell lines more resistant to MeV-mediated oncolysis. These data suggest that differences in the innate immune defense often account for different degrees of susceptibility of sarcoma cell lines to MeV-mediated oncolysis. From a therapeutic perspective, we were able to overcome resistance to MeV by increasing the multiplicity of infection (MOI) and by addition of the prodrug 5-fluorocytosine (FC), thereby exploiting the suicide gene function of virotherapeutic vector MeV-SCD armed with the SCD fusion protein, which consists of yeast cytosine deaminase and yeast uracil phosphoribosyltransferase.
Collapse
|