Meeting Product Development Challenges in Manufacturing Clinical Grade Oncolytic Adenoviruses

Total Page:16

File Type:pdf, Size:1020Kb

Meeting Product Development Challenges in Manufacturing Clinical Grade Oncolytic Adenoviruses Oncogene (2005) 24, 7792–7801 & 2005 Nature Publishing Group All rights reserved 0950-9232/05 $30.00 www.nature.com/onc Meeting product development challenges in manufacturing clinical grade oncolytic adenoviruses Peter K Working*,1, Andy Lin1 and Flavia Borellini1 1Cell Genesys, Inc., 500 Forbes Boulevard, South San Francisco, CA 94080, USA Oncolytic adenoviruses have been considered for use as issue, is possible because the expression of the adeno- anticancer therapy for decades, and numerous means of virus genome is a tightly regulated cascade, which can be conferring tumor selectivity have been developed. As with considered as divided into early (E) and late (L) phases. any new therapy, the trip from the laboratory bench to the The E1 region gene products are critical for efficient clinic has revealed a number of significant development expression of essentially all the other regions of the hurdles. Viral therapies are subject to specific regulations adenovirus genome, such that controlling the expression and must meet a variety of well-defined criteria for purity, of the E1 region will effectively control adenoviral potency, stability, and product characterization prior to replication. Oncolytic adenoviruses that utilize this their use in the clinic. Published regulatory guidelines, approach include CG7870 (Yu et al., 2001), which although developed specifically for biotechnology-derived utilizes prostate-specific transcription regulatory ele- products, are applicable to the production of oncolytic ments to control E1 and target prostate cancer, and adenoviruses and other cell-based products, and they CG0070 (Bristol et al., 2003), which utilizes the human should be consulted early during development. Most E2F-1 promoter cloned in place of the endogenous E1a importantly, both the manufacturing process and the promoter to target the diverse group of cancers that development of characterization and release assays should have a defective retinoblastoma (Rb) pathway. CG0070 be science-driven, use the best available science and also encodes the cDNA for human granulocyte-macro- technology, and must consider the unique nature of the phage colony-stimulating factor (GM-CSF) in the E3 product: a living, and mutatable, virus. Potentially region. Both of these oncolytic adenoviruses are significant impacts on product quality and safety stem currently being evaluated in the clinic. from the possibility of genetic instability related to over- The second approach for engineering tumor specifi- engineering the viruses (as evidenced by their recombina- city entails the deletion of viral gene function(s) that are tion and/or occasional reversion to wild-type virus during critical for efficient viral replication in normal cells, but manufacturing). This report provides examples of some of dispensable in tumor cells. O’Shea and McCormick the critical components affecting the development and discuss this method further in this issue. Examples production of clinical grade material and summarizes the include Rb-inactivated E1a-mutated variants such as significant progress made in recent years. Addl922–947 and Ad5-D24RGD, and p53-defective Oncogene (2005) 24, 7792–7801. doi:10.1038/sj.onc.1209045 E1b-deleted adenovirus variants like Addl1520 (Onyx- 015) and Ad5-CD/Tkrep (Bischoff et al., 1996; Freytag Keywords: oncolytic adenovirus; product development; et al., 1998; Heise et al., 2000; Johnson, 2002). These manufacturing mutants have deletions of the E1a-CR2 and the E1b-55 kD gene regions that are responsible for binding and inactivating Rb family members and p53, respectively. Such viruses were originally believed to target cancer The idea that viruses could be useful as selective cells with genetic defects in the Rb and p53 pathways, anticancer agents is not a new one, dating back to which are defective in most human tumors (Biederer almost a century. In recent years, a variety of tumor- et al., 2002), but more recent work has shown that selective viruses, including over 50 tumor-selective differential late viral RNA export in tumor vs normal oncolytic adenovirus variants have been tested for cells, rather than p53 activation, appears to be the major antitumor activity in animals and humans. There are determinant of the selectivity of Onyx-015 (O’Shea three broad approaches to imbuing adenoviruses with et al., 2004). tumor selectivity. First, the viral genome can be The third approach is based on receptor-mediated engineered to limit the expression of essential viral targeting of tumor cells through genetic modification of genes to tumor tissues via the use of tumor- or tissue- the adenoviral capsid and can be termed transductional specific promoters, that is, by transcriptional targeting. targeting; Mathis et al. discuss it in detail in this issue. This approach, discussed in detail by Ko et al. in this To successfully create a virotherapy agent using this approach requires ablation of the natural viral uptake *Correspondence: PK Working; mechanism (deselection), identification of tumor-specific E-mail: [email protected] receptor targets on cancer cells, and the engineering of Product development of oncolytic adenoviruses PK Working et al 7793 new ligands into the viral coat (reselection), all without replication-competent and replication-deficient, and disrupting viral integrity (Douglas et al., 1999; Alemany the detection of wild-type adenovirus contaminants or and Curiel, 2001; Hemminki et al., 2001; Suzuki et al., wild-type virus produced during manufacturing. These 2001). unique components of product quality assessment of an The objective of this review is to discuss challenges in adenovirus can be evaluated using a bioamplification manufacturing selectively replicating oncolytic adeno- assay, that is, an assay that amplifies either pre-existing viruses for use in clinical trials and, eventually, recombinant replication-competent adenovirus (RCA) commercial use. It focuses on the developmental hurdles variants or wild-type adenovirus and that provides for transcriptionally targeted adenovirus variants made selective pressure to force the expression of genetic with the first strategy, such as CG7870 and CG0070, but instability. most of the hurdles discussed are relevant to the development of any type oncolytic adenovirus and, to Engineering a stable virus a lesser extent, any type of viral-based therapeutic. For more detailed discussions of the basis of adenoviral An unintended effect of engineering tumor-selective replication and strategies for its control, the interested adenoviruses is the potential introduction of genomic reader is directed to Ko et al. and O’Shea et al. (this instability. Wild-type adenoviruses have evolved over volume) and several other recently published reviews the millennia to do their job admirably well: they (Alemany et al., 1999; Biederer et al., 2002; Kirn, 2001; replicate efficiently in host cells and generate thousands Reid et al., 2002; Yu et al., 2002; Wakimoto et al., 2003). of progeny virions to maintain the infection. The adenoviral genome is stable, yet malleable enough to adapt to new circumstances, and changes that attenuate Development of oncolytic adenoviruses its replicative ability may result in genomic instability as the adenovirus attempts to revert to wild type. As with any drug or biologic product destined for use in Engineered oncolytic adenoviruses must retain a stable humans, oncolytic adenovirus products are subject to genome for consistent production and biological activ- broad, yet detailed regulations and must meet a variety ity, but engineering an adenovirus to direct its replica- of well-defined criteria for purity, potency, stability, and tion preferentially to tumor cells, effectively attenuating product characterization, among other things. Guidance its ability to replicate, may introduce significant genomic can be found in the US and EU regulatory documenta- instability reflecting the effort of the engineered virus to tion and policies for Good Manufacturing Practices ‘recover’ from that attenuation. (GMP) for making therapeutic products for human use. Overengineering, for example, the introduction of Particularly relevant are the guidelines for products large variations into the adenoviral genome, may result derived from biotechnology, the latest changes to which in significant genetic instability that prevents the clinical have been described in a recent review article (Jones and use of a virus and may lead to recombinants of differing Levine, 2004). Although developed specifically for specificity and/or potency, potentially including in- biotechnology-derived products, these guidelines are creased potency beyond that of wild-type adenovirus. applicable to the production of oncolytic adenoviruses Examples of genomic instability are reported in the and other cell-based products, and it is critical to consult literature, including dual-promoter-controlled viruses in them during product development, that is, they are which the same promoter is used twice in a single relevant and, perhaps more to the point, they are adenoviral backbone to improve tumor selectivity. In required. However, since oncolytic adenoviruses the Calydon/Cell Genesys, Inc. (CGI) virus CV716, E1a are living and, therefore, more variable, less stable and and E1b are independently controlled by separate PSE even mutatable, there are consequent unique and elements (Henderson et al., 2001; Henderson and Yu, significant challenges for their production, a major one 2002). Tumor selectivity was improved; compared to
Recommended publications
  • POTENTIAL ROLE of MUC1 by Dahlia M. Besmer a Dissertatio
    THE DEVELOPMENT OF NOVEL THERAPEUTICS IN PANCREATIC AND BREAST CANCERS: POTENTIAL ROLE OF MUC1 by Dahlia M. Besmer A dissertation submitted to the faculty of The University of North Carolina at Charlotte in partial fulfillment of the requirements for the degree of Doctor of Philosophy in Biology Charlotte 2013 Approved by: _____________________________ Dr. Pinku Mukherjee _____________________________ Dr. Valery Grdzelishvili _____________________________ Dr. Mark Clemens _____________________________ Dr. Didier Dréau _____________________________ Dr. Craig Ogle ii © 2013 Dahlia M. Besmer ALL RIGHTS RESERVED iii ABSTRACT DAHLIA MARIE BESMER. Development of novel therapeutics in pancreatic and breast cancers: potential role of MUC1. (Under the direction of DR. PINKU MUKHERJEE) Pancreatic ductal adenocarcinoma (PDA) is the 4th leading cause of cancer-related deaths in the US, and breast cancer (BC) contributes to ~40,000 deaths annually. The development of novel therapeutic agents for improving patient outcome is of paramount importance. Importantly, MUC1 is a mucin glycoprotein expressed on the apical surface of normal glandular epithelia but is over expressed and aberrantly glycosylated in >80% of human PDA and in >90% of BC. In the present study, we first utilize a model of PDA that is Muc1-null in order to elucidate the oncogenic role of MUC1. We show that lack of Muc1 significantly decreased proliferation, invasion, and mitotic rates both in vivo and in vitro. Next, we evaluated the anticancer efficacy of oncolytic virus (OV) therapy that utilizes viruses to kill tumor cells. The oncolytic potential of vesicular stomatitis virus (VSV) was analyzed in a panel of human PDA cell lines in vitro and in vivo in immune compromised mice.
    [Show full text]
  • (12) United States Patent (10) Patent No.: US 8.450,106 B2 Kaur Et Al
    USOO845O106B2 (12) United States Patent (10) Patent No.: US 8.450,106 B2 Kaur et al. (45) Date of Patent: May 28, 2013 (54) ONCOLYTIC VIRUS 2009.0117644 A1 5/2009 Yazaki et al. 2009/0215147 A1 8/2009 Zhang et al. (75) Inventors: Balveen Kaur, Dublin, OH (US); 2009, 0220460 A1 9, 2009 Coffin Antonio Chiocca, Powell, OH (US); 2010.0086522 A1 4/2010 Bell et al. Yoshinaga Saeki, Toyama (JP) FOREIGN PATENT DOCUMENTS WO 2008/O12695 A2 1, 2008 (73) Assignee: The Ohio State University Research Foundation, Columbus, OH (US) OTHER PUBLICATIONS (*) Notice: Subject to any disclaimer, the term of this Abdallah, B.M. etal...TheUse of Mesenchymal (Skeletal) StemCells patent is extended or adjusted under 35 for Treatment of Degenerative Diseases: Current Status and Future U.S.C. 154(b) by 410 days. Prespectives, Journal of Cellular Physiology, 2009, pp. 9-12, 218. Aboody, K.S. et al., Neural stem cells display extensive tropism for (21) Appl. No.: 12/697,891 pathology in adult brain: Evidence from intracranial gliomas, PNAS, Nov. 7, 2000, pp. 12846-12851,97(23). Aboody, K.S. et al., Stem and progenitor cell-mediated tumor selec (22) Filed: Feb. 1, 2010 tive gene therapy, Gene Therapy, 2008, pp. 739-752, 15. (65) Prior Publication Data Abordo-Adesida, E. et al., Stability of Lentiviral Vector-Mediated Transgene Expression in the Brain in the Presence of Systemic US 2010/0272686 A1 Oct. 28, 2010 Antivector Immune Responses, Hum Gene Ther. 2005, pp. 741-751, 16(6). Related U.S. Application Data Abremski, K. et al., Bacteriophage P1 Site-specific Recombination, The Journal of Biological Chemistry, Feb.
    [Show full text]
  • Advances and Potential Pitfalls of Oncolytic Viruses Expressing Immunomodulatory Transgene Therapy for Malignant Gliomas Qing Zhang1,2,3 and Fusheng Liu1,2,3
    Zhang and Liu Cell Death and Disease (2020) 11:485 https://doi.org/10.1038/s41419-020-2696-5 Cell Death & Disease REVIEW ARTICLE Open Access Advances and potential pitfalls of oncolytic viruses expressing immunomodulatory transgene therapy for malignant gliomas Qing Zhang1,2,3 and Fusheng Liu1,2,3 Abstract Glioblastoma (GBM) is an immunosuppressive, lethal brain tumor. Despite advances in molecular understanding and therapies, the clinical benefits have remained limited, and the life expectancy of patients with GBM has only been extended to ~15 months. Currently, genetically modified oncolytic viruses (OV) that express immunomodulatory transgenes constitute a research hot spot in the field of glioma treatment. An oncolytic virus is designed to selectively target, infect, and replicate in tumor cells while sparing normal tissues. Moreover, many studies have shown therapeutic advantages, and recent clinical trials have demonstrated the safety and efficacy of their usage. However, the therapeutic efficacy of oncolytic viruses alone is limited, while oncolytic viruses expressing immunomodulatory transgenes are more potent inducers of immunity and enhance immune cell-mediated antitumor immune responses in GBM. An increasing number of basic studies on oncolytic viruses encoding immunomodulatory transgene therapy for malignant gliomas have yielded beneficial outcomes. Oncolytic viruses that are armed with immunomodulatory transgenes remain promising as a therapy against malignant gliomas and will undoubtedly provide new insights into possible clinical uses or strategies. In this review, we summarize the research advances related to oncolytic viruses that express immunomodulatory transgenes, as well as potential treatment pitfalls in patients with malignant gliomas. 1234567890():,; 1234567890():,; 1234567890():,; 1234567890():,; Facts Open questions 1.
    [Show full text]
  • Use of Replicating Oncolytic Adenoviruses in Combination Therapy for Cancer
    Vol. 10, 5299–5312, August 15, 2004 Clinical Cancer Research 5299 Review Use of Replicating Oncolytic Adenoviruses in Combination Therapy for Cancer Roland L. Chu,1,2 Dawn E. Post,1 therapy and radiation damage to their DNA. This confers resist- Fadlo R. Khuri,1 and Erwin G. Van Meir1 ance to treatment through clonal expansion of genetically re- sistant tumor cells. Much effort has been directed toward finding 1Laboratory of Molecular Neuro-Oncology, Departments of Neurosurgery, Hematology/Oncology, and Winship Cancer Institute, alternate pathways that would complement therapeutic induc- and 2AFLAC Cancer and Blood Disorders Center, Children’s tion of apoptosis, overcome multidrug resistance, and ultimately Healthcare of Atlanta, Emory University, Atlanta, Georgia improve overall cure rates. Although the dream of developing a single curative drug per cancer type drives the field, the reality is that combination therapy will be the most effective way of ABSTRACT improving survival. Oncolytic virotherapy is the use of genetically engi- The use of viruses that specifically kill tumor cells while neered viruses that specifically target and destroy tumor sparing normal cells, known as oncolytic virotherapy, has re- cells via their cytolytic replication cycle. Viral-mediated tu- emerged over the past 7 years (1, 2). Viruses have evolved to mor destruction is propagated through infection of nearby maximize their ability to enter cells, use the machinery of the tumor cells by the newly released progeny. Each cycle host cell to replicate and package their own genome and lyse the should amplify the number of oncolytic viruses available for cells to release their progeny and propagate the viral replicative infection.
    [Show full text]
  • Oncolytic Herpes Simplex Virus-Based Therapies for Cancer
    cells Review Oncolytic Herpes Simplex Virus-Based Therapies for Cancer Norah Aldrak 1 , Sarah Alsaab 1,2, Aliyah Algethami 1, Deepak Bhere 3,4,5 , Hiroaki Wakimoto 3,4,5,6 , Khalid Shah 3,4,5,7, Mohammad N. Alomary 1,2,* and Nada Zaidan 1,2,* 1 Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology, P.O. Box 6086, Riyadh 11451, Saudi Arabia; [email protected] (N.A.); [email protected] (S.A.); [email protected] (A.A.) 2 National Center for Biotechnology, Life Science and Environmental Research Institute, King Abdulaziz City for Science and Technology, P.O. Box 6086, Riyadh 11451, Saudi Arabia 3 Center for Stem Cell Therapeutics and Imaging (CSTI), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA; [email protected] (D.B.); [email protected] (H.W.); [email protected] (K.S.) 4 Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA 5 BWH Center of Excellence for Biomedicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA 6 Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA 7 Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA * Correspondence: [email protected] (M.N.A.); [email protected] (N.Z.) Abstract: With the increased worldwide burden of cancer, including aggressive and resistant cancers, oncolytic virotherapy has emerged as a viable therapeutic option. Oncolytic herpes simplex virus (oHSV) can be genetically engineered to target cancer cells while sparing normal cells.
    [Show full text]
  • Genomic DNA Damage and ATR-Chk1 Signaling Determine Oncolytic Adenoviral Efficacy in Human Ovarian Cancer Cells Claire M
    Research article Genomic DNA damage and ATR-Chk1 signaling determine oncolytic adenoviral efficacy in human ovarian cancer cells Claire M. Connell,1,2 Atsushi Shibata,2 Laura A. Tookman,1 Kyra M. Archibald,1 Magdalena B. Flak,1 Katrina J. Pirlo,1 Michelle Lockley,1 Sally P. Wheatley,2 and Iain A. McNeish1 1Centre for Molecular Oncology and Imaging, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom. 2Genome Damage and Stability Centre, University of Sussex, Brighton, United Kingdom. Oncolytic adenoviruses replicate selectively within and lyse malignant cells. As such, they are being devel- oped as anticancer therapeutics. However, the sensitivity of ovarian cancers to adenovirus cytotoxicity varies greatly, even in cells of similar infectivity. Using both the adenovirus E1A-CR2 deletion mutant dl922-947 and WT adenovirus serotype 5 in a panel of human ovarian cancer cell lines that cover a 3-log range of sen- sitivity, we observed profound overreplication of genomic DNA only in highly sensitive cell lines. This was associated with the presence of extensive genomic DNA damage. Inhibition of ataxia telangiectasia and Rad3- related checkpoint kinase 1 (ATR-Chk1), but not ataxia telangiectasia mutated (ATM), promoted genomic DNA damage and overreplication in resistant and partially sensitive cells. This was accompanied by increased adenovirus cytotoxicity both in vitro and in vivo in tumor-bearing mice. We also demonstrated that Cdc25A was upregulated in highly sensitive ovarian cancer cell lines after adenovirus infection and was stabilized after loss of Chk1 activity. Knockdown of Cdc25A inhibited virus-induced DNA damage in highly sensitive cells and blocked the effects of Chk1 inhibition in resistant cells.
    [Show full text]
  • Combining Oncolytic Virotherapy with P53 Tumor Suppressor Gene Therapy
    Review Combining Oncolytic Virotherapy with p53 Tumor Suppressor Gene Therapy Christian Bressy,1 Eric Hastie,2 and Valery Z. Grdzelishvili1 1Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC 28223, USA; 2Department of Biology, Duke University, Durham, NC 27708, USA Oncolytic virus (OV) therapy utilizes replication-competent ing and killing cancer cells (oncotoxicity); (3) able to stimulate an viruses to kill cancer cells, leaving non-malignant cells un- adaptive immune response against cancer cells; and (4) resistant to harmed. With the first U.S. Food and Drug Administration- premature clearance by the immune system during treatment. approved OV, dozens of clinical trials ongoing, and an abun- Despite encouraging results, OV monotherapy based exclusively on dance of translational research in the field, OV therapy is virus replication-induced oncolysis often does not demonstrate all poised to be one of the leading treatments for cancer. A number of these desired qualities, especially when tested against virus-resis- of recombinant OVs expressing a transgene for p53 (TP53) or tant malignancies. Today’s hurdles facing OV therapies remain the – another p53 family member (TP63 or TP73) were engineered same as those described in early6 8 and recent reviews.9,10 Several with the goal of generating more potent OVs that function methods have been developed to increase the anti-cancer activities synergistically with host immunity and/or other therapies to of OVs. Most commonly, OVs are engineered to express an exoge- reduce or eliminate tumor burden. Such transgenes have nous transgene with anti-tumor activity and/or combined with proven effective at improving OV therapies, and basic research standard treatments like radiotherapy or chemotherapy, or with has shown mechanisms of p53-mediated enhancement of OV small-molecule inhibitors of virus-host interactions.
    [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 Tumor Targeting Oncolytic Adenovirus Can Improve Therapeutic Outcomes
    www.nature.com/scientificreports OPEN A tumor targeting oncolytic adenovirus can improve therapeutic outcomes in chemotherapy Received: 21 December 2018 Accepted: 17 April 2019 resistant metastatic human breast Published: xx xx xxxx carcinoma Ali Sakhawat, Ling Ma, Tahir Muhammad, Aamir Ali Khan, Xuechai Chen & Yinghui Huang Breast cancer is the most prevalent malignancy in women, which remains untreatable once metastatic. The treatment of advanced breast cancer is restricted due to chemotherapy resistance. We previously investigated anti-cancer potential of a tumor selective oncolytic adenovirus along with cisplatin in three lung cancer cells; A549, H292, and H661, and found it very efcient. To our surprise, this virotherapy showed remarkable cytotoxicity to chemo-resistant cancer cells. Here, we extended our investigation by using two breast cancer cells and their resistant sublines to further validate CRAd’s anti-resistance properties. Results of in vitro and in vivo analyses recapitulated the similar anti-tumor potential of CRAd. Based on the molecular analysis through qPCR and western blotting, we suggest upregulation of coxsackievirus-adenovirus receptor (CAR) as a selective vulnerability of chemotherapy-resistant tumors. CAR knockdown and overexpression experiments established its important involvement in the success of CRAd-induced tumor inhibition. Additionally, through transwell migration assay we demonstrate that CRAd might have anti-metastatic properties. Mechanistic analysis show that CRAd pre-treatment could reverse epithelial to mesenchymal transition in breast cancer cells, which needs further verifcation. These insights may prove to be a timely opportunity for the application of CRAd in recurrent drug-resistant cancers. Breast cancer is the most prevalent malignancy in women, which remains untreatable once metastatic1.
    [Show full text]
  • UNIVERSITY of CALIFORNIA, SAN DIEGO Engineering Tumor-Specific
    UNIVERSITY OF CALIFORNIA, SAN DIEGO Engineering tumor-specific oncolytic adenoviruses with small molecule- controlled expanded tropisms A dissertation submitted in partial satisfaction of the requirements for the degree Doctor of Philosophy in Biology by Shigeki Joseph Miyake-Stoner Committee in charge: Professor Clodagh O’Shea, Chair Professor Jeff Hasty Professor Martin Hetzer Professor Elizabeth Komives Professor Stephen Mayfield The Dissertation of Shigeki Miyake-Stoner is approved, and it is acceptable in quality and form for publication on microfilm and electronically: ______________________________________________________________ ______________________________________________________________ ______________________________________________________________ ______________________________________________________________ ______________________________________________________________ Chair University of California, San Diego 2017 Signature Page iii Table of Contents Signature Page ................................................................................................. iii Table of Contents ............................................................................................ iv List of Figures .................................................................................................. vii List of Tables ................................................................................................... ix Acknowledgements .......................................................................................... x Vita
    [Show full text]
  • Strategies and Challenges to Arming Oncolytic Viruses with Therapeutic Genes Terry W Hermiston and Irene Kuhn Berlex Biosciences, Richmond, California 94804-0099, USA
    Cancer Gene Therapy (2002) 9, 1022 – 1035 D 2002 Nature Publishing Group All rights reserved 0929-1903/02 $25.00 www.nature.com/cgt Review Armed therapeutic viruses: Strategies and challenges to arming oncolytic viruses with therapeutic genes Terry W Hermiston and Irene Kuhn Berlex Biosciences, Richmond, California 94804-0099, USA. Oncolytic viruses are attractive therapeutics for cancer because they selectively amplify, through replication and spread, the input dose of virus in the target tumor. To date, clinical trials have demonstrated marked safety but have not realized their theoretical efficacy potential. In this review, we consider the potential of armed therapeutic viruses, whose lytic potential is enhanced by genetically engineered therapeutic transgene expression from the virus, as potential vehicles to increase the potency of these agents. Several classes of therapeutic genes are outlined, and potential synergies and hurdles to their delivery from replicating viruses are discussed. Cancer Gene Therapy (2002) 9, 1022–1035 doi:10.1038/sj.cgt.7700542 Keywords: oncolytic virus; armed therapeutic virus; gene therapy; cancer umor-selective, replication-competent oncolytic vi- Three strategies are being pursued to overcome this weak- Truses offer several unique features as cancer therapeu- ness. One is to create less attenuated (more potent) viruses tics. First, the input dose is amplified in a tumor-dependent either through use of alternative viruses or by employing fashion. Consequently, even if only a small proportion of the alternative, less attenuating, mechanisms for restricting input dose infects some of the target tumor cells, this replication to tumor cells.1–3 The second is to employ infective dose should be capable of replicating in and additional cytotoxic mechanisms, beyond the direct lytic eliminating neoplastic cells, using successive waves of functions of the virus, by arming these viruses with replication and lysis until the tumor mass is completely therapeutic genes.4 Particularly attractive in this context are destroyed.
    [Show full text]
  • Intratumoral OH2, an Oncolytic Herpes Simplex Virus 2, in Patients with Advanced Solid Tumors: a Multicenter, Phase I/II Clinical Trial
    Open access Original research J Immunother Cancer: first published as 10.1136/jitc-2020-002224 on 9 April 2021. Downloaded from Intratumoral OH2, an oncolytic herpes simplex virus 2, in patients with advanced solid tumors: a multicenter, phase I/II clinical trial 1 1,2 1 3 4 5 Bo Zhang, Jing Huang , Jialin Tang, Sheng Hu, Suxia Luo, Zhiguo Luo, Fuxiang Zhou,6 Shiyun Tan,7 Jieer Ying,8 Qing Chang,9 Rui Zhang,9 Chengyun Geng,9 Dawei Wu,10 Xiangyong Gu,11 Binlei Liu11,12 To cite: Zhang B, Huang J, ABSTRACT with immune checkpoint inhibitors in selected tumor types Tang J, et al. Intratumoral Background OH2 is a genetically engineered oncolytic is warranted. OH2, an oncolytic herpes herpes simplex virus type 2 designed to selectively amplify simplex virus 2, in patients in tumor cells and express granulocyte- macrophage with advanced solid tumors: a colony- stimulating factor to enhance antitumor immune BACKGROUND multicenter, phase I/II clinical Oncolytic virotherapy represents a unique trial. Journal for ImmunoTherapy responses. We investigated the safety, tolerability and of Cancer 2021;9:e002224. antitumor activity of OH2 as single agent or in combination antitumor strategy using natural or geneti- doi:10.1136/jitc-2020-002224 with HX008, an anti- programmed cell death protein 1 cally engineered viruses to infect and repli- antibody, in patients with advanced solid tumors. cate in tumor cells. The mechanisms of the ► Additional supplemental Methods In this multicenter, phase I/II trial, we enrolled tumoricidal effect include direct tumor cell material is published online only. patients with standard treatment- refractory advanced solid lysis caused by selective infection, and the To view, please visit the journal tumors who have injectable lesions.
    [Show full text]