Viroimmunotherapy for Breast Cancer: Promises, Problems and Future Directions

Total Page:16

File Type:pdf, Size:1020Kb

Viroimmunotherapy for Breast Cancer: Promises, Problems and Future Directions Cancer Gene Therapy (2021) 28:757–768 https://doi.org/10.1038/s41417-020-00265-6 REVIEW ARTICLE Viroimmunotherapy for breast cancer: promises, problems and future directions 1 1 Shyambabu Chaurasiya ● Yuman Fong Received: 18 May 2020 / Revised: 26 August 2020 / Accepted: 12 November 2020 / Published online: 2 December 2020 © The Author(s), under exclusive licence to Springer Nature America, Inc. 2020 Abstract Virotherapy, a strategy to use live viruses as therapeutics, is a relatively novel field in the treatment of cancer. With the advancements in molecular biology and virology, there has been a huge increase in research on cancer virotherapy. For the treatment of cancer, viruses could be used either as vectors in gene therapy or as oncolytic agents. A variety of viruses have been studied for their potential usage in gene therapy or oncolytic therapy. In this review, we discuss virotherapy with a special focus on breast cancer. Breast cancer is the most common cancer and the leading cause of cancer-related deaths in women worldwide. Current treatments are insufficient to cure metastatic breast cancer and are often associated with severe side effects that further deteriorates patients’ quality of life. Therefore, novel therapeutic approaches such as virotherapy 1234567890();,: 1234567890();,: need to be developed for the treatment of breast cancer. Here we summarize the current treatments for breast cancer and the potential use of virotherapy in the treatment of the disease. Furthermore, we discuss the use of oncolytic viruses as immunotherapeutics and the rational combination of oncolytic viruses with other therapeutics for optimal treatment of breast cancer. Finally, we outline the progress made in virotherapy for breast cancer and the shortcomings that need to be addressed for this novel therapy to move to the clinic for better treatment of breast cancer. Breast cancer ever-evolving. Traditional classifications of BrCa were based on the histology and biology of the tumors, whereas Breast cancer (BrCa) is the second most commonly diag- recent classification schemes are mainly based on molecular nosed cancer in the world and according to the last Global differences [5, 6]. Comprehensive gene expression profiling Cancer Statistics (GLOBOCON 2018), BrCa represented by different independent groups using a large set of breast 11.6% of all cancers and caused 6.6% of all cancer-related tumors has led researchers to categorize BrCa into five deaths [1]. Major advancements in the diagnosis and treat- major molecular sub-types, which vary in the expression ment of BrCa made over the last three decades have levels of the estrogen receptor (ER) and human epidermal improved the quality of life and overall survival of patients. growth factor receptor 2 (HER2). The five molecular sub- However, despite improvements in diagnostic methods, types are normal breast-like, luminal A, luminal B, HER2 20–50% of patients develop metastatic disease which +/ER−, and basal-like or triple-negative [7, 8]. Treatments remains difficult to treat and accounts for the majority of for BrCa are dictated to some extent by the molecular sub- deaths [2, 3]. BrCa is a highly heterogeneous disease, and types and prognosis varies among the sub-types, but in the extensive intra- and inter-tumoral heterogeneity are general, patients with basal-like (or triple-negative) tumors thought to be the result of the distinct cells of origin and have the worst prognosis whereas patients with luminal A specific alterations at genetic and epigenetic levels [4]. BrCa tumors have the best prognosis [4, 9]. has been classified in many ways and the classification is Current therapies * Shyambabu Chaurasiya Surgery is commonly used to remove primary breast tumors [email protected] while radiation and chemotherapies are used as adjuvants in 1 Department of Surgery, City of Hope National Medical Center, the treatment of BrCa [2, 10]. For most of the twentieth Duarte, CA, USA century, the Halsted radical mastectomy, an aggressive form 758 S. Chaurasiya, Y. Fong of surgery that includes removal of the breast together with chemotherapy are initially observed in TNBC patients, they muscles of the chest wall, was the standard form of surgery quickly develop resistance to the drugs, and disease relapse for the treatment of BrCa [11, 12]. This approach was based is very common with the median survival of only 13 months on the premise that metastasis in BrCa occurs through the [29]. Taken together, with the advancements in knowledge locoregional spread and thus could be cured by aggressive about BrCa many therapeutic options have been developed, surgery. However, studies in the 1970s showed that radical which have substantially increased the overall survival of mastectomy has no benefit over less aggressive surgeries in patients, however, there is still a need for better treatment terms of either disease recurrence or overall survival options to further improve patients’ quality of life as well as [11, 13]. Later, less aggressive breast conservation surgery overall survival. became more common for local therapy combined with a wide variety of systemic adjuvant therapies [14]. Adjuvant therapies given systemically are aimed at eradicating dis- Virotherapy for breast cancer seminated cancer cells in order to minimize the probability of metastatic growth [15]. Like many other types of cancer, During the course of evolution, cancer cells accumulate metastases are the main cause of death in BrCa patients, many mutations that confer the cells with the ability to resist accounting for more than 90% of total mortality [16]. Stu- cell death, acquire replicative immortality, and render them dies have shown that the use of adjuvant radiation-, chemo-, insensitive to immune surveillance [30]. Interestingly, many endocrine and HER2-directed therapies significantly redu- of the same mutations that allow cancer cells to thrive, also ces the risk of disease recurrence and improves the overall make them better hosts for viruses. Virotherapy, a strategy survival of patients [17–19]. Radiation is usually applied to to use live viruses as therapeutics, exploits these mutated the tumor bed and regional lymph nodes after the resection loopholes to target cancer cells. Cancer virotherapy could of the primary tumor [20]. The most common chemotherapy be broadly categorized into two sub-classes: (i) use of non- adjuvant regimens include the combination of cyclopho- replicating viruses as vectors in cancer gene therapy and (ii) sphamide with doxorubicin and/or docetaxel or methotrex- use of replicating viruses as oncolytic agents. In this review ate with 5-fluorouracil (an excellent review on this topic is article, we will discuss both aspects of virotherapy with provided by Anampa et al. [13]). However, the use of respect to BrCa. radiation- and chemotherapies is limited by off-target toxi- cities and development of resistance to these therapies [21, 22]. Off-target toxicities resulting from chemotherapies Non-replicating viruses for breast cancer substantially affect the quality of patients’ lives and ~40% therapy of patients suffer from disease recurrence and die of metastases [23]. More recent therapies for BrCa have been Many preclinical studies as well as clinical trials have developed to target the expression of estrogen receptor examined the feasibility of gene therapy for the treatment of (ER), progesterone receptor (PR) and human epidermal BrCa. Unlike gene therapy for monogenic disorders where the growth factor receptor 2 (HER2) expression. About 75% of goal is to restore the function of the defective gene, the goal of human BrCa express ER, and endocrine therapy is used for cancer gene therapy is to directly or indirectly kill cancer cells the treatment of such subtype [24]. Tamoxifen, an ER [31]. For direct killing of cancer cells, the strategy of suicide modulator, is the most commonly used endocrine therapy gene therapy is employed, which involves the delivery of a for ER/PR+ BrCa. Patients with HER2+ BrCa are treated toxic gene or a gene that could convert an inactive prodrug with trastuzumab, a monoclonal antibody against HER2. into a cytotoxic drug specifically in cancer cells causing cell Trastuzumab is usually given in combination with adjuvant death [32]. Two most studied strategies for suicide gene chemotherapy either concurrently or sequentially [25]. therapy are (i) herpes simplex virus (HSV)-thymidine kinase However, trastuzumab has been shown to induce some (TK) gene in combination with prodrug ganciclovir and (ii) degree of cardiac toxicity, and approximately half of the cytosine deaminase gene in combination with the prodrug treated patients experience disease relapse within 3 years 5-fluorocytosine [32]. [26, 27]. Majumdar et al. [33] studied adenovirus-mediated HSV- Basal-like or triple-negative breast cancer (TNBC) is the TK (Ad-HSV-TK) expression plus ganciclovir as suicide most aggressive subtype with the highest relapse rate and gene therapy in BrCa models . The authors found that HSV- high incidence of brain metastasis [28]. TNBCs are not TK-ganciclovir therapy had a minimal effect against the amenable to currently available targeted therapies as they murine 4T1 tumors, however, a combination of HSV-TK- lack these molecular targets. Anthracyclines and taxanes are ganciclovir therapy with adenoviruses expressing inter- usually used in the treatment of TNBC, however with very leukin(IL)-2 and granulocyte-macrophage colony-stimulat- limited success [28]. Although favorable responses to ing factor
Recommended publications
  • 1 Systemic Combination Virotherapy for Melanoma with Tumor Antigen-Expressing Vesicular Stomatitis Virus and Adoptive T Cell
    Author Manuscript Published OnlineFirst on July 26, 2012; DOI: 10.1158/0008-5472.CAN-12-0600 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Systemic Combination Virotherapy For Melanoma With Tumor Antigen-Expressing Vesicular Stomatitis Virus And Adoptive T Cell Transfer Diana M. Rommelfanger1,2, Phonphimon Wongthida1, Rosa M. Diaz1,3, Karen M. Kaluza1,3, Jill M. Thompson1, Timothy J. Kottke1, and Richard G. Vile1,3 1Department of Molecular Medicine, 2Department of Molecular Pharmacology and Experimental Therapeutics, 3Department of Immunology, Mayo Clinic, Rochester, MN 55905 Running title: Antitumor effects of systemic combination viro-immunotherapy Keywords: vesicular stomatitis virus, adoptive cell transfer, melanoma, virotherapy, tumor- associated antigen This work was supported by the Richard M. Schulze Family Foundation, the Mayo Foundation, and by NIH grants CA107082, CA130878, and CA132734. Correspondence: Richard Vile, Ph.D. / Mayo Clinic / 200 First St SW / Rochester, MN 55905 Phone: 507-284-9941 / Fax: 507-266-2122 / Email: [email protected] The authors declare no conflict of interest. 1 Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2012 American Association for Cancer Research. Author Manuscript Published OnlineFirst on July 26, 2012; DOI: 10.1158/0008-5472.CAN-12-0600 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Abstract Oncolytic virotherapy offers the potential to treat tumors both as a single agent and in combination with traditional modalities such as chemotherapy and radiotherapy. Here we describe an effective, fully systemic treatment regimen, which combines virotherapy, acting essentially as an adjuvant immunotherapy, with adoptive cell transfer (ACT).
    [Show full text]
  • Adjuvant Oncolytic Virotherapy for Personalized Anti-Cancer Vaccination
    ARTICLE https://doi.org/10.1038/s41467-021-22929-z OPEN Adjuvant oncolytic virotherapy for personalized anti-cancer vaccination D. G. Roy 1,2, K. Geoffroy 3,4,5, M. Marguerie1,2, S. T. Khan1,2, N. T. Martin1, J. Kmiecik6,7, D. Bobbala6, A. S. Aitken1,2, C. T. de Souza1, K. B. Stephenson6, B. D. Lichty6,8, R. C. Auer1,2, D. F. Stojdl2,6,7, J. C. Bell 1,2 & ✉ M.-C. Bourgeois-Daigneault 3,4,5 By conferring systemic protection and durable benefits, cancer immunotherapies are emer- 1234567890():,; ging as long-term solutions for cancer treatment. One such approach that is currently undergoing clinical testing is a therapeutic anti-cancer vaccine that uses two different viruses expressing the same tumor antigen to prime and boost anti-tumor immunity. By providing the additional advantage of directly killing cancer cells, oncolytic viruses (OVs) constitute ideal platforms for such treatment strategy. However, given that the targeted tumor antigen is encoded into the viral genomes, its production requires robust infection and therefore, the vaccination efficiency partially depends on the unpredictable and highly variable intrinsic sensitivity of each tumor to OV infection. In this study, we demonstrate that anti-cancer vaccination using OVs (Adenovirus (Ad), Maraba virus (MRB), Vesicular stomatitis virus (VSV) and Vaccinia virus (VV)) co-administered with antigenic peptides is as efficient as antigen-engineered OVs and does not depend on viral replication. Our strategy is particularly attractive for personalized anti-cancer vaccines targeting patient-specific mutations. We suggest that the use of OVs as adjuvant platforms for therapeutic anti-cancer vaccination warrants testing for cancer treatment.
    [Show full text]
  • Review the Oncolytic Virotherapy Treatment Platform for Cancer
    Cancer Gene Therapy (2002) 9, 1062 – 1067 D 2002 Nature Publishing Group All rights reserved 0929-1903/02 $25.00 www.nature.com/cgt Review The oncolytic virotherapy treatment platform for cancer: Unique biological and biosafety points to consider Richard Vile,1 Dale Ando,2 and David Kirn3,4 1Molecular Medicine Program, Mayo Clinic, Rochester, Minnesota, USA; 2Cell Genesys Corp., Foster City, California, USA; 3Department of Pharmacology, Oxford University Medical School, Oxford, UK; and 4Kirn Oncology Consulting, San Francisco, California, USA. The field of replication-selective oncolytic viruses (virotherapy) has exploded over the last 10 years. As with many novel therapeutic approaches, initial overexuberance has been tempered by clinical trial results with first-generation agents. Although a number of significant hurdles to this approach have now been identified, novel solutions have been proposed and improvements are being made at a furious rate. This article seeks to initiate a discussion of these hurdles, approaches to overcome them, and unique safety and regulatory issues to consider. Cancer Gene Therapy (2002) 9, 1062 – 1067 doi:10.1038/sj.cgt.7700548 Keywords: oncolytic; virotherapy; experimental therapeutics; cancer ew cancer treatments are needed. These agents must limitation of levels of delivery to cancer cells in a solid tumor Nhave novel mechanisms of action and thereby lack mass. Over 10 different virotherapy agents have entered, or cross-resistance with currently available treatments. Viruses will soon be entering, clinical trials; one such adenovirus have evolved to infect, replicate in, and kill human cells (dl1520) has entered a Phase III clinical trial in recurrent through diverse mechanisms. Clinicians treated hundreds of head and neck carcinoma.
    [Show full text]
  • A Bibliometric Review of Oncolytic Virus Research As a Novel Approach For
    Mozafari Nejad et al. Virol J (2021) 18:98 https://doi.org/10.1186/s12985-021-01571-7 REVIEW Open Access A bibliometric review of oncolytic virus research as a novel approach for cancer therapy Amir Sasan Mozafari Nejad1 , Tehjeeb Noor2, Ziaul Haque Munim3, Mohammad Yousef Alikhani4* and Amir Ghaemi5* Abstract Background: In recent years, oncolytic viruses (OVs) have drawn attention as a novel therapy to various types of can- cers, both in clinical and preclinical cancer studies all around the world. Consequently, researchers have been actively working on enhancing cancer therapy since the early twentieth century. This study presents a systematic review of the literature on OVs, discusses underlying research clusters and, presents future directions of OVs research. Methods: A total of 1626 published articles related to OVs as cancer therapy were obtained from the Web of Science (WoS) database published between January 2000 and March 2020. Various aspects of OVs research, including the countries/territories, institutions, journals, authors, citations, research areas, and content analysis to fnd trending and emerging topics, were analysed using the bibliometrix package in the R-software. Results: In terms of the number of publications, the USA based researchers were the most productive (n 611) followed by Chinese (n 197), and Canadian (n 153) researchers. The Molecular Therapy journal ranked frst= both in terms of the number= of publications (n 133)= and local citations (n 1384). The most prominent institution was Mayo Clinic from the USA (n 117) followed= by the University of Ottawa= from Canada (n 72), and the University of Helsinki from Finland (n 63).= The most impactful author was Bell J.C with the highest number= of articles (n 67) and total local citations (n =885).
    [Show full text]
  • CRISPR-Cas9: a New and Promising Player in Gene Therapy
    Downloaded from http://jmg.bmj.com/ on October 22, 2015 - Published by group.bmj.com Methods CRISPR-Cas9: a new and promising player in gene therapy 1 2 3 4,1 5 Editor’s choice Lu Xiao-Jie, Xue Hui-Ying, Ke Zun-Ping, Chen Jin-Lian, Ji Li-Juan Scan to access more free content 1Department of ABSTRACT as insertional mutations and non-physical expres- Gastroenterology, Shanghai First introduced into mammalian organisms in 2013, the sion of proteins, the programmable nucleases use a East Hospital, Tongji University ‘ ’ School of Medicine, Shanghai, RNA-guided genome editing tool CRISPR-Cas9 (clustered cut-and-paste strategy, that is, remove the defect China regularly interspaced short palindromic repeats/CRISPR- and install the correct, thus representing an prefer- 2The Reproductive Center, associated nuclease 9) offers several advantages over able tool for gene therapy. Recently, a RNA-guided Jiangsu Huai’an Maternity and conventional ones, such as simple-to-design, easy-to-use genome editing tool termed CRISPR-Cas9 (clus- ’ Children Hospital, Huai an, and multiplexing (capable of editing multiple genes tered regularly interspaced short palindromic China 3Department of Cardiology, simultaneously). Consequently, it has become a cost- repeats/CRISPR-associated nuclease 9) added to the The Fifth People’s Hospital effective and convenient tool for various genome editing list of programmable nucleases, offers several of Shanghai, Fudan University, purposes including gene therapy studies. In cell lines or advantages over its counterparts and shows thera- Shanghai, China animal models, CRISPR-Cas9 can be applied for peutic potentials. Herein, we introduce the basic 4Department of Gastroenterology, Shanghai therapeutic purposes in several ways.
    [Show full text]
  • Intraarterial Delivery of Virotherapy for Glioblastoma
    NEUROSURGICAL FOCUS Neurosurg Focus 50 (2):E7, 2021 Intraarterial delivery of virotherapy for glioblastoma Visish M. Srinivasan, MD,1 Frederick F. Lang, MD,2 and Peter Kan, MD3 1Department of Neurosurgery, Barrow Neurological Institute, Phoenix, Arizona; 2Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas; and 3Department of Neurosurgery, University of Texas Medical Branch, Galveston, Texas Oncolytic viruses (OVs) have been used in the treatment of cancer, in a focused manner, since the 1990s. These OVs have become popular in the treatment of several cancers but are only now gaining interest in the treatment of glioblas- toma (GBM) in recent clinical trials. In this review, the authors discuss the unique applications of intraarterial (IA) delivery of OVs, starting with concepts of OV, how they apply to IA delivery, and concluding with discussion of the current ongo- ing trials. Several OVs have been used in the treatment of GBM, including specifically several modified adenoviruses. IA delivery of OVs has been performed in the hepatic circulation and is now being studied in the cerebral circulation to help enhance delivery and specificity. There are some interesting synergies with immunotherapy and IA delivery of OVs. Some of the shortcomings are discussed, specifically the systemic response to OVs and feasibility of treatment. Future studies can be performed in the preclinical setting to identify the ideal candidates for translation into clinical trials, as well as the nuances of this novel delivery method. https://thejns.org/doi/abs/10.3171/2020.11.FOCUS20845 KEYWORDS endovascular; intraarterial; glioblastoma; virotherapy; oncolytic; stem cell NCOLYTIC viruses (OVs) have been tested since the Intraarterial (IA) delivery of OVs has been established in 1950s;1–3 however, it was not until the 1990s1 that other cancers and has had early applications in GBM as viral genomic engineering resulted in the first gen- well.
    [Show full text]
  • Synergistic Combination of Oncolytic Virotherapy and Immunotherapy for Glioma Bingtao Tang1, Zong Sheng Guo2, David L
    Published OnlineFirst February 4, 2020; DOI: 10.1158/1078-0432.CCR-18-3626 CLINICAL CANCER RESEARCH | TRANSLATIONAL CANCER MECHANISMS AND THERAPY Synergistic Combination of Oncolytic Virotherapy and Immunotherapy for Glioma Bingtao Tang1, Zong Sheng Guo2, David L. Bartlett2, David Z. Yan1, Claire P. Schane1, Diana L. Thomas3, Jia Liu4, Grant McFadden5, Joanna L. Shisler6, and Edward J. Roy1 ABSTRACT ◥ Purpose: We hypothesized that the combination of a local Results: vvDD-IL15Ra-YFP and vMyx-IL15Ra-tdTr each þ stimulus for activating tumor-specific T cells and an anti- infected and killed GL261 cells in vitro. In vivo, NK cells and CD8 immunosuppressant would improve treatment of gliomas. Virally T cells were increased in the tumor due to the expression of IL15Ra- encoded IL15Ra-IL15 as the T-cell activating stimulus and a IL15. Each component of a combination treatment contributed to prostaglandin synthesis inhibitor as the anti-immunosuppressant prolonging survival: an oncolytic virus, the IL15Ra-IL15 expressed were combined with adoptive transfer of tumor-specific T cells. by the virus, a source of T cells (whether by prevaccination or Experimental Design: Two oncolytic poxviruses, vvDD vaccinia adoptive transfer), and prostaglandin inhibition all synergized virus and myxoma virus, were each engineered to express the fusion to produce elimination of gliomas in a majority of mice. vvDD- protein IL15Ra-IL15 and a fluorescent protein. Viral gene expres- IL15Ra-YFP occasionally caused ventriculitis-meningitis, but sion (YFP or tdTomato Red) was confirmed in the murine glioma vMyx-IL15Ra-tdTr was safe and effective, causing a strong infil- GL261 in vitro and in vivo.
    [Show full text]
  • Combination Immunotherapy Using Oncolytic Virus for the Treatment of Advanced Solid Tumors
    International Journal of Molecular Sciences Review Combination Immunotherapy Using Oncolytic Virus for the Treatment of Advanced Solid Tumors Chang-Myung Oh 1, Hong Jae Chon 2,* and Chan Kim 2,* 1 Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Korea; [email protected] 2 Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam 13497, Korea * Correspondence: [email protected] (H.J.C.); [email protected] (C.K.) Received: 24 September 2020; Accepted: 16 October 2020; Published: 19 October 2020 Abstract: Oncolytic virus (OV) is a new therapeutic strategy for cancer treatment. OVs can selectively infect and destroy cancer cells, and therefore act as an in situ cancer vaccine by releasing tumor-specific antigens. Moreover, they can remodel the tumor microenvironment toward a T cell-inflamed phenotype by stimulating widespread host immune responses against the tumor. Recent evidence suggests several possible applications of OVs against cancer, especially in combination with immune checkpoint inhibitors. In this review, we describe the molecular mechanisms of oncolytic virotherapy and OV-induced immune responses, provide a brief summary of recent preclinical and clinical updates on this rapidly evolving field, and discuss a combinational strategy that is able to overcome the limitations of OV-based monotherapy. Keywords: oncolytic virus; combination immunotherapy; immune checkpoint inhibitor; tumor microenvironment 1. Introduction Viruses are continuously co-evolving with our immune systems and have developed sophisticated mechanisms to manipulate host immune responses [1]. With scientific advances in the understanding of the molecular interplay between viruses and host immune systems, we can develop a new therapeutic modality against cancers using finely tuned viruses, called viroceuticals [2,3].
    [Show full text]
  • Review RNA Viruses As Virotherapy Agents Stephen J Russell Molecular Medicine Program, Mayo Clinic, Rochester, Minnesota 55905, USA
    Cancer Gene Therapy (2002) 9, 961 – 966 D 2002 Nature Publishing Group All rights reserved 0929-1903/02 $25.00 www.nature.com/cgt Review RNA viruses as virotherapy agents Stephen J Russell Molecular Medicine Program, Mayo Clinic, Rochester, Minnesota 55905, USA. RNA viruses are rapidly emerging as extraordinarily promising agents for oncolytic virotherapy. Integral to the lifecycles of all RNA viruses is the formation of double-stranded RNA, which activates a spectrum of cellular defense mechanisms including the activation of PKR and the release of interferon. Tumors are frequently defective in their PKR signaling and interferon response pathways, and therefore provide a relatively permissive substrate for the propagation of RNA viruses. For most of the oncolytic RNA viruses currently under study, tumor specificity is either a natural characteristic of the virus, or a serendipitous consequence of adapting the virus to propagate in human tumor cell lines. Further refinement and optimization of these oncolytic agents can be achieved through virus engineering. This article provides a summary of the current status of oncolytic virotherapy efforts for seven different RNA viruses, namely, mumps, Newcastle disease virus, measles virus, vesicular stomatitis virus, influenza, reovirus, and poliovirus. Cancer Gene Therapy (2002) 9, 961–966 doi:10.1038/sj.cgt.7700535 he majority of significant human and animal pathogenic double-stranded RNA is to stimulate release of interferons, Tviruses have RNA genomes. Influenza, measles, which activate PKR in adjacent uninfected cells, thereby mumps, rubella, polio, rabies, yellow fever, dengue, and protecting them from virus infection. Tumors are frequently Ebola hemorrhagic fever are among the better known human defective in their PKR signaling pathway, and therefore examples.
    [Show full text]
  • Development of Group B Coxsackievirus As an Oncolytic Virus: Opportunities and Challenges
    viruses Review Development of Group B Coxsackievirus as an Oncolytic Virus: Opportunities and Challenges Huitao Liu 1,2 and Honglin Luo 1,3,* 1 Centre for Heart Lung Innovation, St. Paul’s Hospital—University of British Columbia, Vancouver, BC V6Z 1Y6, Canada; [email protected] 2 Department of Experimental Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada 3 Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada * Correspondence: [email protected] Abstract: Oncolytic viruses have emerged as a promising strategy for cancer therapy due to their dual ability to selectively infect and lyse tumor cells and to induce systemic anti-tumor immunity. Among various candidate viruses, coxsackievirus group B (CVBs) have attracted increasing attention in recent years. CVBs are a group of small, non-enveloped, single-stranded, positive-sense RNA viruses, belonging to species human Enterovirus B in the genus Enterovirus of the family Picornaviridae. Preclinical studies have demonstrated potent anti-tumor activities for CVBs, particularly type 3, against multiple cancer types, including lung, breast, and colorectal cancer. Various approaches have been proposed or applied to enhance the safety and specificity of CVBs towards tumor cells and to further increase their anti-tumor efficacy. This review summarizes current knowledge and strategies for developing CVBs as oncolytic viruses for cancer virotherapy. The challenges arising from these studies and future prospects are also discussed in this review. Citation: Liu, H.; Luo, H. Keywords: coxsackievirus B; oncolytic virus; cancer; virotherapy; cancer; toxicity; anti-tumor immunity Development of Group B Coxsackievirus as an Oncolytic Virus: Opportunities and Challenges.
    [Show full text]
  • Interferon Signaling Predicts Response to Oncolytic Virotherapy
    www.oncotarget.com Oncotarget, 2019, Vol. 10, (No. 16), pp: 1544-1545 Editorial Interferon signaling predicts response to oncolytic virotherapy Cheyne Kurokawa and Evanthia Galanis Oncolytic virotherapy is emerging as a promising the observed difference in replication among our patients therapeutic approach for cancer treatment and involves [7]. To our surprise analysis of expression levels of the the use of viruses that selectively replicate and kill tumor three MV receptors revealed comparable levels among cells. Results from clinical trials clearly indicate that not study patients, thus suggesting that a post-entry restriction all patients achieve a favorable therapeutic response, mechanism rather than an entry related mechanism was however [1]. Talimogene laherparepvec (TVEC), an responsible for the observed differences in replication HSV-1 strain expressing GMCSF was approved in 2015 [6]. In order to investigate this further we studied gene by FDA and later the European Medicines Agency for the expression differences in primary GBM patient-derived treatment of metastatic melanoma marking a significant xenografts (PDXs) that were permissive or resistant to MV breakthrough in the field of oncolytic virotherapy. In infection and cell killing. A comparison of differentially the phase III trial that led to FDA approval, a durable activated pathways between MV resistant and permissive objective response was observed in 16% of patients treated cells revealed a pre-existing antiviral state in resistant cells, with TVEC as compared to 2% of patients treated with characterized by the constitutive activation of the antiviral GM-CSF alone [2]. Although this study met its primary interferon (IFN) pathway. This allowed us to develop a endpoint of durable objective response leading to the diagonal linear analysis algorithm (DLDA), a weighted approval of TVEC for this indication, the therapeutic gene signature consisting of 22 interferon stimulated genes benefit from TVEC was still only achieved in a subset of (ISG).
    [Show full text]
  • The Oncolytic Newcastle Disease Virus As an Effective Immunotherapeutic Strategy Against Glioblastoma
    NEUROSURGICAL FOCUS Neurosurg Focus 50 (2):E8, 2021 The oncolytic Newcastle disease virus as an effective immunotherapeutic strategy against glioblastoma Joshua A. Cuoco, DO, MS,1–3 Cara M. Rogers, DO,1–3 and Sandeep Mittal, MD, FRCSC1–4 1Carilion Clinic Neurosurgery, Roanoke; 2Fralin Biomedical Research Institute at Virginia Tech Carilion School of Medicine, Roanoke; 3School of Neuroscience, Virginia Tech, Blacksburg; and 4Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, Virginia Glioblastoma is the most frequent primary brain tumor in adults, with a dismal prognosis despite aggressive resec- tion, chemotherapeutics, and radiotherapy. Although understanding of the molecular pathogenesis of glioblastoma has progressed in recent years, therapeutic options have failed to significantly change overall survival or progression-free survival. Thus, researchers have begun to explore immunomodulation as a potential strategy to improve clinical out- comes. The application of oncolytic virotherapy as a novel biological to target pathogenic signaling in glioblastoma has brought new hope to the field of neuro-oncology. This class of immunotherapeutics combines selective cancer cell lysis prompted by virus induction while promoting a strong inflammatory antitumor response, thereby acting as an effective in situ tumor vaccine. Several investigators have reported the efficacy of experimental oncolytic viruses as demonstrated by improved long-term survival in cancer patients with advanced disease. Newcastle disease virus (NDV) is one of the most well-researched oncolytic viruses known to affect a multitude of human cancers, including glioblastoma. Preclinical in vitro and in vivo studies as well as human clinical trials have demonstrated that NDV exhibits oncolytic activity against glioblastoma, providing a promising avenue of potential treatment.
    [Show full text]