51
|
Virotherapy in Germany-Recent Activities in Virus Engineering, Preclinical Development, and Clinical Studies. Viruses 2021; 13:v13081420. [PMID: 34452286 PMCID: PMC8402873 DOI: 10.3390/v13081420] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/16/2021] [Accepted: 07/16/2021] [Indexed: 02/06/2023] Open
Abstract
Virotherapy research involves the development, exploration, and application of oncolytic viruses that combine direct killing of cancer cells by viral infection, replication, and spread (oncolysis) with indirect killing by induction of anti-tumor immune responses. Oncolytic viruses can also be engineered to genetically deliver therapeutic proteins for direct or indirect cancer cell killing. In this review—as part of the special edition on “State-of-the-Art Viral Vector Gene Therapy in Germany”—the German community of virotherapists provides an overview of their recent research activities that cover endeavors from screening and engineering viruses as oncolytic cancer therapeutics to their clinical translation in investigator-initiated and sponsored multi-center trials. Preclinical research explores multiple viral platforms, including new isolates, serotypes, or fitness mutants, and pursues unique approaches to engineer them towards increased safety, shielded or targeted delivery, selective or enhanced replication, improved immune activation, delivery of therapeutic proteins or RNA, and redirecting antiviral immunity for cancer cell killing. Moreover, several oncolytic virus-based combination therapies are under investigation. Clinical trials in Germany explore the safety and potency of virotherapeutics based on parvo-, vaccinia, herpes, measles, reo-, adeno-, vesicular stomatitis, and coxsackie viruses, including viruses encoding therapeutic proteins or combinations with immune checkpoint inhibitors. These research advances represent exciting vantage points for future endeavors of the German virotherapy community collectively aimed at the implementation of effective virotherapeutics in clinical oncology.
Collapse
|
52
|
Spiesschaert B, Angerer K, Park J, Wollmann G. Combining Oncolytic Viruses and Small Molecule Therapeutics: Mutual Benefits. Cancers (Basel) 2021; 13:3386. [PMID: 34298601 PMCID: PMC8306439 DOI: 10.3390/cancers13143386] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 02/07/2023] Open
Abstract
The focus of treating cancer with oncolytic viruses (OVs) has increasingly shifted towards achieving efficacy through the induction and augmentation of an antitumor immune response. However, innate antiviral responses can limit the activity of many OVs within the tumor and several immunosuppressive factors can hamper any subsequent antitumor immune responses. In recent decades, numerous small molecule compounds that either inhibit the immunosuppressive features of tumor cells or antagonize antiviral immunity have been developed and tested for. Here we comprehensively review small molecule compounds that can achieve therapeutic synergy with OVs. We also elaborate on the mechanisms by which these treatments elicit anti-tumor effects as monotherapies and how these complement OV treatment.
Collapse
Affiliation(s)
- Bart Spiesschaert
- Christian Doppler Laboratory for Viral Immunotherapy of Cancer, Medical University Innsbruck, 6020 Innsbruck, Austria; (B.S.); (K.A.)
- Institute of Virology, Medical University Innsbruck, 6020 Innsbruck, Austria
- ViraTherapeutics GmbH, 6063 Rum, Austria
- Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach a.d. Riss, Germany;
| | - Katharina Angerer
- Christian Doppler Laboratory for Viral Immunotherapy of Cancer, Medical University Innsbruck, 6020 Innsbruck, Austria; (B.S.); (K.A.)
- Institute of Virology, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - John Park
- Boehringer Ingelheim Pharma GmbH & Co. KG, 88397 Biberach a.d. Riss, Germany;
| | - Guido Wollmann
- Christian Doppler Laboratory for Viral Immunotherapy of Cancer, Medical University Innsbruck, 6020 Innsbruck, Austria; (B.S.); (K.A.)
- Institute of Virology, Medical University Innsbruck, 6020 Innsbruck, Austria
| |
Collapse
|
53
|
Oncolytic Viruses for Malignant Glioma: On the Verge of Success? Viruses 2021; 13:v13071294. [PMID: 34372501 PMCID: PMC8310195 DOI: 10.3390/v13071294] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/26/2021] [Accepted: 06/28/2021] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma is one of the most difficult tumor types to treat with conventional therapy options like tumor debulking and chemo- and radiotherapy. Immunotherapeutic agents like oncolytic viruses, immune checkpoint inhibitors, and chimeric antigen receptor T cells have revolutionized cancer therapy, but their success in glioblastoma remains limited and further optimization of immunotherapies is needed. Several oncolytic viruses have demonstrated the ability to infect tumors and trigger anti-tumor immune responses in malignant glioma patients. Leading the pack, oncolytic herpesvirus, first in its class, awaits an approval for treating malignant glioma from MHLW, the federal authority of Japan. Nevertheless, some major hurdles like the blood–brain barrier, the immunosuppressive tumor microenvironment, and tumor heterogeneity can engender suboptimal efficacy in malignant glioma. In this review, we discuss the current status of malignant glioma therapies with a focus on oncolytic viruses in clinical trials. Furthermore, we discuss the obstacles faced by oncolytic viruses in malignant glioma patients and strategies that are being used to overcome these limitations to (1) optimize delivery of oncolytic viruses beyond the blood–brain barrier; (2) trigger inflammatory immune responses in and around tumors; and (3) use multimodal therapies in combination to tackle tumor heterogeneity, with an end goal of optimizing the therapeutic outcome of oncolytic virotherapy.
Collapse
|
54
|
Varudkar N, Oyer JL, Copik A, Parks GD. Oncolytic parainfluenza virus combines with NK cells to mediate killing of infected and non-infected lung cancer cells within 3D spheroids: role of type I and type III interferon signaling. J Immunother Cancer 2021; 9:jitc-2021-002373. [PMID: 34172515 PMCID: PMC8237729 DOI: 10.1136/jitc-2021-002373] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2021] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND There is intense interest in developing novel oncolytic viruses, which can be used in cancer therapies along with immune cells such as natural killer (NK) cells. We have previously developed a particle-based method for in vitro expansion of highly cytotoxic human NK cells (PM21-NK cells). Here, we have tested the hypothesis that oncolytic parainfluenza virus 5 (P/V virus) can combine with PM21-NK cells for targeted killing of lung cancer cells. METHODS PM21-NK cells were assayed for killing of P/V virus-infected A549, H1299 and Calu-1 lung cancer cells in two-dimensional (2D) and three-dimensional (3D) cultures using flow cytometry, luminescence and kinetic imaging-based methods. Blocking antibodies were used to evaluate NK cell activating receptors involved in PM21-NK cell killing of infected target cells. Media transfer experiments tested soluble factors that increase PM21-NK cell killing of both P/V virus-infected and uninfected tumor cells. RESULTS In 2D cultures, PM21-NK cells efficiently killed P/V virus-infected cancer cells compared with non-infected cells, through involvement of the viral glycoprotein and NK cell receptors NKp30, NKp46 and NKG2D. In 3D spheroid cultures, P/V virus infection was restricted to the outer layer of the spheroid. However, PM21-NK cells were able to more efficiently kill both the outer layer of infected cells in the spheroid and progressing further to kill the uninfected interior cells. Media transfer experiments demonstrated that P/V virus infection produced both type I and type III interferons, which decreased cell growth, which contributed to a reduction in the overall number of uninfected tumor cells in conjunction with PM21-NK cells. Across five cancer cell lines, the contribution of P/V virus infection on PM21-NK cell killing of target cells correlated with interferon induction. CONCLUSION Our data support the potential of combining oncolytic parainfluenza virus with PM21-NK cell adoptive therapy against lung cancer.
Collapse
Affiliation(s)
- Namita Varudkar
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Jeremiah L Oyer
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Alicja Copik
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Griffith D Parks
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| |
Collapse
|
55
|
Maulana TI, Kromidas E, Wallstabe L, Cipriano M, Alb M, Zaupa C, Hudecek M, Fogal B, Loskill P. Immunocompetent cancer-on-chip models to assess immuno-oncology therapy. Adv Drug Deliv Rev 2021; 173:281-305. [PMID: 33798643 DOI: 10.1016/j.addr.2021.03.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 03/08/2021] [Accepted: 03/17/2021] [Indexed: 12/12/2022]
Abstract
The advances in cancer immunotherapy come with several obstacles, limiting its widespread use and benefits so far only to a small subset of patients. One of the underlying challenges remains to be the lack of representative nonclinical models that translate to human immunity and are able to predict clinical efficacy and safety outcomes. In recent years, immunocompetent Cancer-on-Chip models emerge as an alternative human-based platform that enables the integration and manipulation of complex tumor microenvironment. In this review, we discuss novel opportunities offered by Cancer-on-Chip models to advance (mechanistic) immuno-oncology research, ranging from design flexibility to multimodal analysis approaches. We then exemplify their (potential) applications for the research and development of adoptive cell therapy, immune checkpoint therapy, cytokine therapy, oncolytic virus, and cancer vaccines.
Collapse
|
56
|
Wu T, Zhu J. Recent development and optimization of pseudomonas aeruginosa exotoxin immunotoxins in cancer therapeutic applications. Int Immunopharmacol 2021; 96:107759. [PMID: 34162138 DOI: 10.1016/j.intimp.2021.107759] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/30/2021] [Accepted: 05/01/2021] [Indexed: 12/12/2022]
Abstract
Recombinant immunotoxins are fusion proteins composed of a peptide toxin and a specific targeting domain through genetic recombination. They are engineered to recognize disease-specific target receptors and kill the cell upon internalization. Full-sized monoclonal antibodies, smaller antibody fragments and ligands, such as a cytokine or a growth factor, have been commonly used as the targeting domain, while bacterial Pseudomonas aeruginosa exotoxin (PE) is the usual toxin fusion partner, due to its natural cytotoxicity and other unique advantages. PE-based recombinant immunotoxins have shown remarkable efficacy in the treatment of tumors and autoimmune diseases. At the same time, efforts are underway to address major challenges, including immunogenicity, nonspecific cytotoxicity and poor penetration, which limit their clinical applications. Recent strategies for structural optimization of PE-based immunotoxins, combined with mutagenesis approaches, have reduced the immunogenicity and non-specific cytotoxicity, thus increasing both their safety and efficacy. This review highlights novel insights and design concepts that were used to advance immunotoxins for the treatment of hematological and solid tumors and also presents future development prospect of PE-based recombinant immunotoxins that are expected to play an important role in cancer therapy.
Collapse
Affiliation(s)
- Tong Wu
- Engineering Research Center of Cell and Therapeutic Antibody, MOE, China; School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Jianwei Zhu
- Engineering Research Center of Cell and Therapeutic Antibody, MOE, China; School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China; Jecho Laboratories, Inc., Frederick, MD 21704, USA.
| |
Collapse
|
57
|
Senekal NS, Mahasa KJ, Eladdadi A, de Pillis L, Ouifki R. Natural Killer Cells Recruitment in Oncolytic Virotherapy: A Mathematical Model. Bull Math Biol 2021; 83:75. [PMID: 34008149 DOI: 10.1007/s11538-021-00903-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 04/20/2021] [Indexed: 01/17/2023]
Abstract
In this paper, we investigate how natural killer (NK) cell recruitment to the tumor microenvironment (TME) affects oncolytic virotherapy. NK cells play a major role against viral infections. They are, however, known to induce early viral clearance of oncolytic viruses, which hinders the overall efficacy of oncolytic virotherapy. Here, we formulate and analyze a simple mathematical model of the dynamics of the tumor, OV and NK cells using currently available preclinical information. The aim of this study is to characterize conditions under which the synergistic balance between OV-induced NK responses and required viral cytopathicity may or may not result in a successful treatment. In this study, we found that NK cell recruitment to the TME must take place neither too early nor too late in the course of OV infection so that treatment will be successful. NK cell responses are most influential at either early (partly because of rapid response of NK cells to viral infections or antigens) or later (partly because of antitumoral ability of NK cells) stages of oncolytic virotherapy. The model also predicts that: (a) an NK cell response augments oncolytic virotherapy only if viral cytopathicity is weak; (b) the recruitment of NK cells modulates tumor growth; and (c) the depletion of activated NK cells within the TME enhances the probability of tumor escape in oncolytic virotherapy. Taken together, our model results demonstrate that OV infection is crucial, not just to cytoreduce tumor burden, but also to induce the stronger NK cell response necessary to achieve complete or at least partial tumor remission. Furthermore, our modeling framework supports combination therapies involving NK cells and OV which are currently used in oncolytic immunovirotherapy to treat several cancer types.
Collapse
Affiliation(s)
- Noma Susan Senekal
- Department of Mathematics and Computer Science, National University of Lesotho, Roma, Maseru, Lesotho.
| | - Khaphetsi Joseph Mahasa
- Department of Mathematics and Computer Science, National University of Lesotho, Roma, Maseru, Lesotho
| | | | | | - Rachid Ouifki
- Department of Mathematics and Applied Mathematics, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
58
|
Long S, Gu Y, An Y, Lin X, Chen X, Wang X, Liao C, Ouyang W, Wang N, He Z, Zhao X. Reovirus enhances cytotoxicity of natural killer cells against colorectal cancer via TLR3 pathway. J Transl Med 2021; 19:185. [PMID: 33933132 PMCID: PMC8088708 DOI: 10.1186/s12967-021-02853-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/21/2021] [Indexed: 02/08/2023] Open
Abstract
Background Cetuximab has been approved for use for first-line treatment of patients with wild-type KRAS metastatic colorectal cancer (CRC). However, treatment with cetuximab has shown limited efficacy as a CRC monotherapy. In addition, natural killer (NK) cell function is known to be severely attenuated in cancer patients. The goal of this study was to develop a new strategy to enhance antibody-dependent cell-mediated cytotoxicity (ADCC) mediated by NK cells, in combination with cetuximab against CRC cells. Methods Ex vivo expanded NK cells were stimulated with reovirus, and reovirus-activated NK cells mediated ADCC assay were performed on CRC cells in combination with cetuximab. The synergistic antitumor effects of reovirus-activated NK cells and cetuximab were tested on DLD-1 tumor-bearing mice. Finally, Toll-like receptor 3 (TLR3) knockdown in NK cells, along with chemical blockade of TLR3/dsRNA complex, and inhibition of the TLR3 downstream signaling pathway, were performed to explore the mechanisms by which reovirus enhances NK cell cytotoxicity. Results We first confirmed that exposure of NK cells to reovirus enhanced their cytotoxicity in a dose-dependent manner.We then investigated whether reovirus-activated NK cells exposed to cetuximab-bound CRC cells exhibited greater anti-tumor efficacy than either monotherapy. Co-culture of CRC cell lines with reovirus-activated NK cells indicated that NK cytotoxicity was significantly higher in combination with cetuximab, regardless of KRAS mutation status or EGFR expression level. We also found that reovirus activation of NK cells, in conjunction with cetuximab, resulted in significantly stronger anti-tumor efficacy.Finally, TLR3 knockdown, inhibition of TLR3/dsRNA complex or TBK1/IKKε demonstrated that activation of NK cells by reovirus was dependent on TLR3 and its downstream signaling pathway. Conclusions This study demonstrated that combination treatment of reovirus-activated NK cells with cetuximab synergistically enhances their anti-tumor cytotoxicity, suggesting a strong candidate strategy for clinical treatment of CRC. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-02853-y.
Collapse
Affiliation(s)
- Shiqi Long
- Center for Stem Cell and Tissue Engineering Research/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550004, Guizhou, China.,Key Laboratory of Adult Stem Cell Transformation Research, Guiyang, 550004, China
| | - Yangzhuo Gu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yuanyuan An
- Center for Stem Cell and Tissue Engineering Research/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550004, Guizhou, China.,Key Laboratory of Adult Stem Cell Transformation Research, Guiyang, 550004, China
| | - Xiaojin Lin
- Center for Stem Cell and Tissue Engineering Research/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550004, Guizhou, China.,Key Laboratory of Adult Stem Cell Transformation Research, Guiyang, 550004, China
| | - Xiaoqing Chen
- Center for Stem Cell and Tissue Engineering Research/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550004, Guizhou, China.,Key Laboratory of Adult Stem Cell Transformation Research, Guiyang, 550004, China
| | - Xianyao Wang
- Center for Stem Cell and Tissue Engineering Research/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550004, Guizhou, China.,Key Laboratory of Adult Stem Cell Transformation Research, Guiyang, 550004, China
| | - Chunxiang Liao
- Center for Stem Cell and Tissue Engineering Research/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550004, Guizhou, China.,Key Laboratory of Adult Stem Cell Transformation Research, Guiyang, 550004, China
| | - Weiwei Ouyang
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University and Guizhou Cancer Hospital, Guiyang, 550004, China
| | - Nianxue Wang
- Key Laboratory of Adult Stem Cell Transformation Research, Guiyang, 550004, China
| | - Zhixu He
- Center for Stem Cell and Tissue Engineering Research/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550004, Guizhou, China.,Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Xing Zhao
- Center for Stem Cell and Tissue Engineering Research/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550004, Guizhou, China. .,Key Laboratory of Adult Stem Cell Transformation Research, Guiyang, 550004, China. .,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
59
|
Seyed-Khorrami SM, Soleimanjahi H, Soudi S, Habibian A. MSCs loaded with oncolytic reovirus: migration and in vivo virus delivery potential for evaluating anti-cancer effect in tumor-bearing C57BL/6 mice. Cancer Cell Int 2021; 21:244. [PMID: 33933086 PMCID: PMC8088007 DOI: 10.1186/s12935-021-01848-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 02/20/2021] [Indexed: 02/07/2023] Open
Abstract
Background and aims Several oncolytic viruses applications have been approved in the clinic or in different phases of clinical trials. However, these methods have some rudimentary problems. Therefore, to enhance the delivery and quality of treatment, considering the advantage of cell carrier-based methods such as Mesenchymal Stem Cells (MSC) have been proposed. This study was designed to evaluate the performance and quality of cancer treatment based on MSCs loaded by oncolytic reovirus in the cancerous C57BL/6 mouse model. Also, we evaluated MSCs migration potency in vitro and in vivo following the oncolytic reovirus infection. Methods C57BL/6 mice were inoculated with TC-1 cell lines and tumors were established in the right flank. Mice were systemically treated with reovirus, MSCs-loaded with reovirus, MSCs, and PBS as a control in separated groups. Effects of infected AD-MSCs with reovirus on tumor growth and penetration in the tumor site were monitored. All groups of mice were monitored for two months in order to therapeutic and anticancer potential. After treatments, tumor size alteration and apoptosis rate, as well as cytokine release pattern was assessed. Results The results of the current study indicated that the effect of reovirus infection on AD-MSCs is not devastating the migration capacity especially in MOI 1 and 5 while intact cells remain. On the other hand, MSCs play an efficient role as a carrier to deliver oncolytic virus into the tumor site in comparison with systemic administration of reovirus alone. Apoptosis intensity relies on viral titration and passing time. Followed by systemic administration, treatment with oncolytic reovirus-infected AD-MSCs and MSCs alone had shown significant inhibition in tumor growth. Also, treatment by reovirus causes an increase in IFN-γ secretion. Conclusion The results of in vitro and in vivo study confirmed the tumor-homing properties of infected AD-MSCs and the significant antitumor activity of this platform. Hence, our results showed that the cell carrier strategy using oncolytic reovirus-loaded AD-MSCs enhanced virus delivery, infiltration, and antitumor activity can be effectively applied in most cancers.
Collapse
Affiliation(s)
| | - Hoorieh Soleimanjahi
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ala Habibian
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
60
|
Moaven O, Mangieri CW, Stauffer JA, Anastasiadis PZ, Borad MJ. Strategies to Develop Potent Oncolytic Viruses and Enhance Their Therapeutic Efficacy. JCO Precis Oncol 2021; 5:PO.21.00003. [PMID: 34250395 DOI: 10.1200/po.21.00003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/09/2021] [Accepted: 03/23/2021] [Indexed: 02/04/2023] Open
Abstract
Despite advancements in cancer therapy that have occurred over the past several decades, successful treatment of advanced malignancies remains elusive. Substantial resources and significant efforts have been directed toward the development of novel therapeutic modalities to improve patient outcomes. Oncolytic viruses (OVs) are emerging tools with unique characteristics that have attracted great interest in developing effective anticancer treatment. The original attraction was directed toward selective replication and cell-specific toxicity, two unique features that are either inherent to the virus or could be conferred by genetic engineering. However, recent advancements in the knowledge and understanding of OVs are shifting the therapeutic paradigm toward a greater focus on their immunomodulatory role. Nonetheless, there are still significant obstacles that remain to be overcome to enhance the efficiency of OVs as effective therapeutic modalities and potentially establish them as part of standard treatment regimens. In this review, we discuss advances in the design of OVs, strategies to enhance their therapeutic efficacy, functional translation into the clinical settings, and various obstacles that are still encountered in the efforts to establish them as effective anticancer treatments.
Collapse
Affiliation(s)
- Omeed Moaven
- Section of Surgical Oncology, Department of Surgery, Mayo Clinic Florida, Jacksonville, FL
| | - Christopher W Mangieri
- Section of Surgical Oncology, Department of Surgery, Wake Forest University, Winston-Salem, NC
| | - John A Stauffer
- Section of Surgical Oncology, Department of Surgery, Mayo Clinic Florida, Jacksonville, FL
| | | | - Mitesh J Borad
- Division of Medical Oncology, Department of Medicine, Mayo Clinic Arizona, Scottsdale, AZ
| |
Collapse
|
61
|
Combination Therapy of Novel Oncolytic Adenovirus with Anti-PD1 Resulted in Enhanced Anti-Cancer Effect in Syngeneic Immunocompetent Melanoma Mouse Model. Pharmaceutics 2021; 13:pharmaceutics13040547. [PMID: 33919827 PMCID: PMC8070801 DOI: 10.3390/pharmaceutics13040547] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/10/2021] [Accepted: 04/11/2021] [Indexed: 12/24/2022] Open
Abstract
Malignant melanoma, an aggressive form of skin cancer, has a low five-year survival rate in patients with advanced disease. Immunotherapy represents a promising approach to improve survival rates among patients at advanced stage. Herein, the aim of the study was to design and produce, by using engineering tools, a novel oncolytic adenovirus AdV-D24- inducible co-stimulator ligand (ICOSL)-CD40L expressing potent co-stimulatory molecules enhancing clinical efficacy through the modulation of anti-cancer immune responses. Firstly, we demonstrated the vector's identity and genetic stability by restriction enzyme assay and sequencing, then, by performing in vitro and in vivo pre-clinical studies we explored the anti-cancer efficacy of the virus alone or in combination with anti PD-1 inhibitor in human melanoma cell lines, i.e., MUG Mel-1 and MUG Mel-2, and in immunocompetent C57BL/6 melanoma B16V mouse model. We showed that both monotherapy and combination approaches exhibit enhanced anti-cancer ability and immunogenic cell death in in vitro settings. Furthermore, AdV-D24-ICOSL-CD40L combined with anti PD-1 revealed a fall in tumor volume and 100% survival in in vivo context, thus suggesting enhanced efficacy and survival via complementary anti-cancer properties of those agents in melanoma therapy. Collectively, the novel oncolytic vector AdV-D24-ICOSL-CD40L alone or in combination with anticancer drugs, such as check point inhibitors, may open novel therapeutic perspectives for the treatment of melanoma.
Collapse
|
62
|
Redirecting the Immune Microenvironment in Acute Myeloid Leukemia. Cancers (Basel) 2021; 13:cancers13061423. [PMID: 33804676 PMCID: PMC8003817 DOI: 10.3390/cancers13061423] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/13/2021] [Accepted: 03/17/2021] [Indexed: 12/28/2022] Open
Abstract
Simple Summary Despite remarkable progress in the outcome of childhood acute myeloid leukemia (AML), risk of relapse and refractory diseases remains high. Treatment of the chemo-refractory disease is restricted by dose-limiting therapy-related toxicities which necessitate alternative tolerable efficient therapeutic modalities. By disrupting its immune environment, leukemic blasts are known to gain the ability to evade immune surveillance and promote disease progression; therefore, many efforts have been made to redirect the immune system against malignant blasts. Deeper knowledge about immunologic alterations has paved the way to the discovery and development of novel targeted therapeutic concepts, which specifically override the immune evasion mechanisms to eradicate leukemic blasts. Herein, we review innovative immunotherapeutic strategies and their mechanisms of action in pediatric AML. Abstract Acute myeloid leukemia is a life-threatening malignant disorder arising in a complex and dysregulated microenvironment that, in part, promotes the leukemogenesis. Treatment of relapsed and refractory AML, despite the current overall success rates in management of pediatric AML, remains a challenge with limited options considering the heavy but unsuccessful pretreatments in these patients. For relapsed/refractory (R/R) patients, hematopoietic stem cell transplantation (HSCT) following ablative chemotherapy presents the only opportunity to cure AML. Even though in some cases immune-mediated graft-versus-leukemia (GvL) effect has been proven to efficiently eradicate leukemic blasts, the immune- and chemotherapy-related toxicities and adverse effects considerably restrict the feasibility and therapeutic power. Thus, immunotherapy presents a potent tool against acute leukemia but needs to be engineered to function more specifically and with decreased toxicity. To identify innovative immunotherapeutic approaches, sound knowledge concerning immune-evasive strategies of AML blasts and the clinical impact of an immune-privileged microenvironment is indispensable. Based on our knowledge to date, several promising immunotherapies are under clinical evaluation and further innovative approaches are on their way. In this review, we first focus on immunological dysregulations contributing to leukemogenesis and progression in AML. Second, we highlight the most promising therapeutic targets for redirecting the leukemic immunosuppressive microenvironment into a highly immunogenic environment again capable of anti-leukemic immune surveillance.
Collapse
|
63
|
Liu YT, Sun ZJ. Turning cold tumors into hot tumors by improving T-cell infiltration. Am J Cancer Res 2021; 11:5365-5386. [PMID: 33859752 PMCID: PMC8039952 DOI: 10.7150/thno.58390] [Citation(s) in RCA: 391] [Impact Index Per Article: 97.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 02/20/2021] [Indexed: 02/07/2023] Open
Abstract
Immunotherapy, represented by immune checkpoint inhibitors (ICIs), has greatly improved the clinical efficacy of malignant tumor therapy. ICI-mediated antitumor responses depend on the infiltration of T cells capable of recognizing and killing tumor cells. ICIs are not effective in "cold tumors", which are characterized by the lack of T-cell infiltration. To realize the full potential of immunotherapy and solve this obstacle, it is essential to understand the drivers of T-cell infiltration into tumors. We present a critical review of our understanding of the mechanisms underlying “cold tumors”, including impaired T-cell priming and deficient T-cell homing to tumor beds. “Hot tumors” with significant T-cell infiltration are associated with better ICI efficacy. In this review, we summarize multiple strategies that promote the transformation of "cold tumors" into “hot tumors” and discuss the mechanisms by which these strategies lead to increased T-cell infiltration. Finally, we discuss the application of nanomaterials to tumor immunotherapy and provide an outlook on the future of this emerging field. The combination of nanomedicines and immunotherapy enhances cross-presentation of tumor antigens and promotes T-cell priming and infiltration. A deeper understanding of these mechanisms opens new possibilities for the development of multiple T cell-based combination therapies to improve ICI effectiveness.
Collapse
|
64
|
Holbrook MC, Goad DW, Grdzelishvili VZ. Expanding the Spectrum of Pancreatic Cancers Responsive to Vesicular Stomatitis Virus-Based Oncolytic Virotherapy: Challenges and Solutions. Cancers (Basel) 2021; 13:1171. [PMID: 33803211 PMCID: PMC7963195 DOI: 10.3390/cancers13051171] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a devastating malignancy with poor prognosis and a dismal survival rate, expected to become the second leading cause of cancer-related deaths in the United States. Oncolytic virus (OV) is an anticancer approach that utilizes replication-competent viruses to preferentially infect and kill tumor cells. Vesicular stomatitis virus (VSV), one such OV, is already in several phase I clinical trials against different malignancies. VSV-based recombinant viruses are effective OVs against a majority of tested PDAC cell lines. However, some PDAC cell lines are resistant to VSV. Upregulated type I IFN signaling and constitutive expression of a subset of interferon-simulated genes (ISGs) play a major role in such resistance, while other mechanisms, such as inefficient viral attachment and resistance to VSV-mediated apoptosis, also play a role in some PDACs. Several alternative approaches have been shown to break the resistance of PDACs to VSV without compromising VSV oncoselectivity, including (i) combinations of VSV with JAK1/2 inhibitors (such as ruxolitinib); (ii) triple combinations of VSV with ruxolitinib and polycations improving both VSV replication and attachment; (iii) combinations of VSV with chemotherapeutic drugs (such as paclitaxel) arresting cells in the G2/M phase; (iv) arming VSV with p53 transgenes; (v) directed evolution approach producing more effective OVs. The latter study demonstrated impressive long-term genomic stability of complex VSV recombinants encoding large transgenes, supporting further clinical development of VSV as safe therapeutics for PDAC.
Collapse
Affiliation(s)
| | | | - Valery Z. Grdzelishvili
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC 28223, USA; (M.C.H.); (D.W.G.)
| |
Collapse
|
65
|
Jenner AL, Cassidy T, Belaid K, Bourgeois-Daigneault MC, Craig M. In silico trials predict that combination strategies for enhancing vesicular stomatitis oncolytic virus are determined by tumor aggressivity. J Immunother Cancer 2021; 9:jitc-2020-001387. [PMID: 33608375 PMCID: PMC7898884 DOI: 10.1136/jitc-2020-001387] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2020] [Indexed: 12/19/2022] Open
Abstract
Background Immunotherapies, driven by immune-mediated antitumorigenicity, offer the potential for significant improvements to the treatment of multiple cancer types. Identifying therapeutic strategies that bolster antitumor immunity while limiting immune suppression is critical to selecting treatment combinations and schedules that offer durable therapeutic benefits. Combination oncolytic virus (OV) therapy, wherein complementary OVs are administered in succession, offer such promise, yet their translation from preclinical studies to clinical implementation is a major challenge. Overcoming this obstacle requires answering fundamental questions about how to effectively design and tailor schedules to provide the most benefit to patients. Methods We developed a computational biology model of combined oncolytic vaccinia (an enhancer virus) and vesicular stomatitis virus (VSV) calibrated to and validated against multiple data sources. We then optimized protocols in a cohort of heterogeneous virtual individuals by leveraging this model and our previously established in silico clinical trial platform. Results Enhancer multiplicity was shown to have little to no impact on the average response to therapy. However, the duration of the VSV injection lag was found to be determinant for survival outcomes. Importantly, through treatment individualization, we found that optimal combination schedules are closely linked to tumor aggressivity. We predicted that patients with aggressively growing tumors required a single enhancer followed by a VSV injection 1 day later, whereas a small subset of patients with the slowest growing tumors needed multiple enhancers followed by a longer VSV delay of 15 days, suggesting that intrinsic tumor growth rates could inform the segregation of patients into clinical trials and ultimately determine patient survival. These results were validated in entirely new cohorts of virtual individuals with aggressive or non-aggressive subtypes. Conclusions Based on our results, improved therapeutic schedules for combinations with enhancer OVs can be studied and implemented. Our results further underline the impact of interdisciplinary approaches to preclinical planning and the importance of computational approaches to drug discovery and development.
Collapse
Affiliation(s)
- Adrianne L Jenner
- Sainte-Justine University Hospital Research Centre, Montreal, Quebec, Canada.,Department of Mathematics and Statistics, Université de Montréal, Montreal, Quebec, Canada
| | - Tyler Cassidy
- Department of Mathematics and Statistics, McGill University, Montreal, Quebec, Canada.,Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | - Katia Belaid
- Department of Mathematics and Statistics, Université de Montréal, Montreal, Quebec, Canada.,Statistique et Informatique Décisionnelle, Université Toulouse III Paul Sabatier, Toulouse, Occitanie, France
| | - Marie-Claude Bourgeois-Daigneault
- Institut du Cancer de Montréal, CHUM, Montreal, Quebec, Canada.,Department of Microbiology, Infectious diseases and Immunology, Université de Montréal, Montreal, Quebec, Canada
| | - Morgan Craig
- Sainte-Justine University Hospital Research Centre, Montreal, Quebec, Canada .,Department of Mathematics and Statistics, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
66
|
Middelburg J, Kemper K, Engelberts P, Labrijn AF, Schuurman J, van Hall T. Overcoming Challenges for CD3-Bispecific Antibody Therapy in Solid Tumors. Cancers (Basel) 2021; 13:287. [PMID: 33466732 PMCID: PMC7829968 DOI: 10.3390/cancers13020287] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/08/2021] [Accepted: 01/10/2021] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy of cancer with CD3-bispecific antibodies is an approved therapeutic option for some hematological malignancies and is under clinical investigation for solid cancers. However, the treatment of solid tumors faces more pronounced hurdles, such as increased on-target off-tumor toxicities, sparse T-cell infiltration and impaired T-cell quality due to the presence of an immunosuppressive tumor microenvironment, which affect the safety and limit efficacy of CD3-bispecific antibody therapy. In this review, we provide a brief status update of the CD3-bispecific antibody therapy field and identify intrinsic hurdles in solid cancers. Furthermore, we describe potential combinatorial approaches to overcome these challenges in order to generate selective and more effective responses.
Collapse
Affiliation(s)
- Jim Middelburg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
| | - Kristel Kemper
- Genmab, 3584 CT Utrecht, The Netherlands; (K.K.); (P.E.); (A.F.L.); (J.S.)
| | - Patrick Engelberts
- Genmab, 3584 CT Utrecht, The Netherlands; (K.K.); (P.E.); (A.F.L.); (J.S.)
| | - Aran F. Labrijn
- Genmab, 3584 CT Utrecht, The Netherlands; (K.K.); (P.E.); (A.F.L.); (J.S.)
| | - Janine Schuurman
- Genmab, 3584 CT Utrecht, The Netherlands; (K.K.); (P.E.); (A.F.L.); (J.S.)
| | - Thorbald van Hall
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
| |
Collapse
|
67
|
Vannini A, Leoni V, Sanapo M, Gianni T, Giordani G, Gatta V, Barboni C, Zaghini A, Campadelli-Fiume G. Immunotherapeutic Efficacy of Retargeted oHSVs Designed for Propagation in an Ad Hoc Cell Line. Cancers (Basel) 2021; 13:E266. [PMID: 33445744 PMCID: PMC7828196 DOI: 10.3390/cancers13020266] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/07/2021] [Accepted: 01/08/2021] [Indexed: 02/06/2023] Open
Abstract
Our laboratory has pursued the generation of cancer-specific oncolytic herpes simplex viruses (oHSVs) which ensure high efficacy while maintaining a high safety profile. Their blueprint included retargeting to a Tumor-Associated Antigen, e.g., HER2, coupled to detargeting from natural receptors to avoid off-target and off-tumor infections and preservation of the full complement of unmodified viral genes. These oHSVs are "fully virulent in their target cancer cells". The 3rd generation retargeted oHSVs carry two distinct retargeting moieties, which enable infection of a producer cell line and of the target cancer cells, respectively. They can be propagated in an ad hoc Vero cell derivative at about tenfold higher yields than 1st generation recombinants, and more effectively replicate in human cancer cell lines. The R-335 and R-337 prototypes were armed with murine IL-12. Intratumorally-administered R-337 conferred almost complete protection from LLC-1-HER2 primary tumors, unleashed the tumor microenvironment immunosuppression, synergized with the checkpoint blockade and conferred long-term vaccination against distant challenge tumors. In summary, the problem intrinsic to the propagation of retargeted oHSVs-which strictly require cells positive for targeted receptors-was solved in 3rd generation viruses. They are effective as immunotherapeutic agents against primary tumors and as antigen-agnostic vaccines.
Collapse
Affiliation(s)
- Andrea Vannini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40126 Bologna, Italy; (V.L.); (M.S.); (T.G.); (V.G.)
| | - Valerio Leoni
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40126 Bologna, Italy; (V.L.); (M.S.); (T.G.); (V.G.)
| | - Mara Sanapo
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40126 Bologna, Italy; (V.L.); (M.S.); (T.G.); (V.G.)
| | - Tatiana Gianni
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40126 Bologna, Italy; (V.L.); (M.S.); (T.G.); (V.G.)
| | - Giorgia Giordani
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy;
| | - Valentina Gatta
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40126 Bologna, Italy; (V.L.); (M.S.); (T.G.); (V.G.)
| | - Catia Barboni
- Department of Veterinary Medical Sciences, University of Bologna, 40126 Bologna, Italy; (C.B.); (A.Z.)
| | - Anna Zaghini
- Department of Veterinary Medical Sciences, University of Bologna, 40126 Bologna, Italy; (C.B.); (A.Z.)
| | - Gabriella Campadelli-Fiume
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40126 Bologna, Italy; (V.L.); (M.S.); (T.G.); (V.G.)
| |
Collapse
|
68
|
Zhang S, Rabkin SD. The discovery and development of oncolytic viruses: are they the future of cancer immunotherapy? Expert Opin Drug Discov 2020; 16:391-410. [PMID: 33232188 DOI: 10.1080/17460441.2021.1850689] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Despite diverse treatment modalities and novel therapies, many cancers and patients are not effectively treated. Cancer immunotherapy has recently achieved breakthrough status yet is not effective in all cancer types or patients and can generate serious adverse effects. Oncolytic viruses (OVs) are a promising new therapeutic modality that harnesses virus biology and host interactions to treat cancer. OVs, genetically engineered or natural, preferentially replicate in and kill cancer cells, sparing normal cells/tissues, and mediating anti-tumor immunity.Areas covered: This review focuses on OVs as cancer therapeutic agents from a historical perspective, especially strategies to boost their immunotherapeutic activities. OVs offer a multifaceted platform, whose activities are modulated based on the parental virus and genetic alterations. In addition to direct viral effects, many OVs can be armed with therapeutic transgenes to also act as gene therapy vectors, and/or combined with other drugs or therapies.Expert opinion: OVs are an amazingly versatile and malleable class of cancer therapies. They tend to target cellular and host physiology as opposed to specific genetic alterations, which potentially enables broad responsiveness. The biological complexity of OVs have hindered their translation; however, the recent approval of talimogene laherparepvec (T-Vec) has invigorated the field.
Collapse
Affiliation(s)
- Shunchuan Zhang
- Molecular Neurosurgery Laboratory and the Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Samuel D Rabkin
- Molecular Neurosurgery Laboratory and the Brain Tumor Research Center, Department of Neurosurgery, Massachusetts General Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| |
Collapse
|
69
|
Jamieson TR, Poutou J, Ilkow CS. Redirecting oncolytic viruses: Engineering opportunists to take control of the tumour microenvironment. Cytokine Growth Factor Rev 2020; 56:102-114. [DOI: 10.1016/j.cytogfr.2020.07.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 07/02/2020] [Indexed: 12/12/2022]
|
70
|
Froechlich G, Caiazza C, Gentile C, D’Alise AM, De Lucia M, Langone F, Leoni G, Cotugno G, Scisciola V, Nicosia A, Scarselli E, Mallardo M, Sasso E, Zambrano N. Integrity of the Antiviral STING-mediated DNA Sensing in Tumor Cells Is Required to Sustain the Immunotherapeutic Efficacy of Herpes Simplex Oncolytic Virus. Cancers (Basel) 2020; 12:cancers12113407. [PMID: 33213060 PMCID: PMC7698602 DOI: 10.3390/cancers12113407] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Oncolytic viruses are emerging immunotherapeutics in cancer treatments. The conflicting role of innate immunity in the antitumor activity of oncolytic viruses is still a matter of debate. The STING-dependent DNA sensing axis is considered detrimental for viral replication and cancer cell clearance. Accordingly, we observed that STING loss in tumor cells was associated with improved lytic potential by a herpes-based oncolytic virus. However, STING-knockout cancer cells infected with the oncolytic virus showed impaired immunogenicity, as immunogenic cell death was improperly triggered. In agreement with these observations, STING-knockout tumors raised in a murine syngeneic model were more resistant to a combined treatment of the oncolytic virus with PD-1 blockade. The present study demonstrates the antitumor benefit of antiviral immunity and sheds lights on the mechanisms of immune resistance to oncovirotherapy exerted by STING-loss in tumor cells. Abstract The dichotomic contribution of cancer cell lysis and tumor immunogenicity is considered essential for effective oncovirotherapy, suggesting that the innate antiviral immune response is a hurdle for efficacy of oncolytic viruses. However, emerging evidence is resizing this view. By sensing cytosolic DNA, the cyclic GMP-AMP synthase (cGAS) and stimulator of interferon genes (STING) axis can both counteract viral spread and contribute to the elicitation of adaptive immunity via type I interferon responses. In this paper, we analyzed the tumor-resident function of Sting-mediated DNA sensing in a combined approach of oncovirotherapy and PD-1 immune checkpoint blockade, in an immunocompetent murine model. While supporting increased lytic potential by oncolytic HER2-retargeted HSV-1 in vitro and in vivo, Sting-knockout tumors showed molecular signatures of an immunosuppressive tumor microenvironment. These signatures were correspondingly associated with ineffectiveness of the combination therapy in a model of established tumors. Results suggest that the impairment in antiviral response of Sting-knockout tumors, while favoring viral replication, is not able to elicit an adequate immunotherapeutic effect, due to lack of immunogenic cell death and the inability of Sting-knockout cancer cells to promote anti-tumor adaptive immune responses. Accordingly, we propose that antiviral, tumor-resident Sting provides fundamental contributions to immunotherapeutic efficacy of oncolytic viruses.
Collapse
Affiliation(s)
- Guendalina Froechlich
- CEINGE Biotecnologie Avanzate S.C.aR.L., Via G. Salvatore 486, 80145 Naples, Italy; (G.F.); (C.G.); (V.S.); (A.N.); (N.Z.)
| | - Carmen Caiazza
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy; (C.C.); (M.M.)
| | - Chiara Gentile
- CEINGE Biotecnologie Avanzate S.C.aR.L., Via G. Salvatore 486, 80145 Naples, Italy; (G.F.); (C.G.); (V.S.); (A.N.); (N.Z.)
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy; (C.C.); (M.M.)
| | - Anna Morena D’Alise
- Nouscom S.R.L., Via di Castel Romano 100, 00128 Rome, Italy; (A.M.D.); (M.D.L.); (F.L.); (G.L.); (G.C.); (E.S.)
| | - Maria De Lucia
- Nouscom S.R.L., Via di Castel Romano 100, 00128 Rome, Italy; (A.M.D.); (M.D.L.); (F.L.); (G.L.); (G.C.); (E.S.)
| | - Francesca Langone
- Nouscom S.R.L., Via di Castel Romano 100, 00128 Rome, Italy; (A.M.D.); (M.D.L.); (F.L.); (G.L.); (G.C.); (E.S.)
| | - Guido Leoni
- Nouscom S.R.L., Via di Castel Romano 100, 00128 Rome, Italy; (A.M.D.); (M.D.L.); (F.L.); (G.L.); (G.C.); (E.S.)
| | - Gabriella Cotugno
- Nouscom S.R.L., Via di Castel Romano 100, 00128 Rome, Italy; (A.M.D.); (M.D.L.); (F.L.); (G.L.); (G.C.); (E.S.)
| | - Vittorio Scisciola
- CEINGE Biotecnologie Avanzate S.C.aR.L., Via G. Salvatore 486, 80145 Naples, Italy; (G.F.); (C.G.); (V.S.); (A.N.); (N.Z.)
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy; (C.C.); (M.M.)
| | - Alfredo Nicosia
- CEINGE Biotecnologie Avanzate S.C.aR.L., Via G. Salvatore 486, 80145 Naples, Italy; (G.F.); (C.G.); (V.S.); (A.N.); (N.Z.)
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy; (C.C.); (M.M.)
- Nouscom S.R.L., Via di Castel Romano 100, 00128 Rome, Italy; (A.M.D.); (M.D.L.); (F.L.); (G.L.); (G.C.); (E.S.)
| | - Elisa Scarselli
- Nouscom S.R.L., Via di Castel Romano 100, 00128 Rome, Italy; (A.M.D.); (M.D.L.); (F.L.); (G.L.); (G.C.); (E.S.)
| | - Massimo Mallardo
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy; (C.C.); (M.M.)
| | - Emanuele Sasso
- CEINGE Biotecnologie Avanzate S.C.aR.L., Via G. Salvatore 486, 80145 Naples, Italy; (G.F.); (C.G.); (V.S.); (A.N.); (N.Z.)
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy; (C.C.); (M.M.)
- Nouscom S.R.L., Via di Castel Romano 100, 00128 Rome, Italy; (A.M.D.); (M.D.L.); (F.L.); (G.L.); (G.C.); (E.S.)
- Correspondence:
| | - Nicola Zambrano
- CEINGE Biotecnologie Avanzate S.C.aR.L., Via G. Salvatore 486, 80145 Naples, Italy; (G.F.); (C.G.); (V.S.); (A.N.); (N.Z.)
- Department of Molecular Medicine and Medical Biotechnology (DMMBM), University of Naples Federico II, Via Pansini 5, 80131 Naples, Italy; (C.C.); (M.M.)
| |
Collapse
|
71
|
Kuryk L, Bertinato L, Staniszewska M, Pancer K, Wieczorek M, Salmaso S, Caliceti P, Garofalo M. From Conventional Therapies to Immunotherapy: Melanoma Treatment in Review. Cancers (Basel) 2020; 12:cancers12103057. [PMID: 33092131 PMCID: PMC7589099 DOI: 10.3390/cancers12103057] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/16/2020] [Accepted: 10/18/2020] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Here, we review the current state of knowledge in the field of cancer immunotherapy, focusing on the scientific rationale for the use of oncolytic viruses, checkpoint inhibitors and their combination to combat melanomas. Attention is also given to the immunological aspects of cancer therapy and the shift from conventional therapy towards immunotherapy. This review brings together information on how immunotherapy can be applied to support other cancer therapies in order to maximize the efficacy of melanoma treatment and improve clinical outcomes. Abstract In this review, we discuss the use of oncolytic viruses and checkpoint inhibitors in cancer immunotherapy in melanoma, with a particular focus on combinatory therapies. Oncolytic viruses are promising and novel anti-cancer agents, currently under investigation in many clinical trials both as monotherapy and in combination with other therapeutics. They have shown the ability to exhibit synergistic anticancer activity with checkpoint inhibitors, chemotherapy, radiotherapy. A coupling between oncolytic viruses and checkpoint inhibitors is a well-accepted strategy for future cancer therapies. However, eradicating advanced cancers and tailoring the immune response for complete tumor clearance is an ongoing problem. Despite current advances in cancer research, monotherapy has shown limited efficacy against solid tumors. Therefore, current improvements in virus targeting, genetic modification, enhanced immunogenicity, improved oncolytic properties and combination strategies have a potential to widen the applications of immuno-oncology (IO) in cancer treatment. Here, we summarize the strategy of combinatory therapy with an oncolytic vector to combat melanoma and highlight the need to optimize current practices and improve clinical outcomes.
Collapse
Affiliation(s)
- Lukasz Kuryk
- Department of Virology, National Institute of Public Health-National Institute of Hygiene, Chocimska 24, 00-791 Warsaw, Poland; (K.P.); (M.W.)
- Clinical Science, Targovax Oy, Saukonpaadenranta 2, 00180 Helsinki, Finland
- Correspondence: (L.K.); (M.G.)
| | - Laura Bertinato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy; (L.B.); (S.S.); (P.C.)
| | - Monika Staniszewska
- Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland;
- Centre for Advanced Materials and Technologies, Warsaw University of Technology, Poleczki 19, 02-822 Warsaw, Poland
| | - Katarzyna Pancer
- Department of Virology, National Institute of Public Health-National Institute of Hygiene, Chocimska 24, 00-791 Warsaw, Poland; (K.P.); (M.W.)
| | - Magdalena Wieczorek
- Department of Virology, National Institute of Public Health-National Institute of Hygiene, Chocimska 24, 00-791 Warsaw, Poland; (K.P.); (M.W.)
| | - Stefano Salmaso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy; (L.B.); (S.S.); (P.C.)
| | - Paolo Caliceti
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy; (L.B.); (S.S.); (P.C.)
| | - Mariangela Garofalo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy; (L.B.); (S.S.); (P.C.)
- Correspondence: (L.K.); (M.G.)
| |
Collapse
|
72
|
Shukla S, Hu H, Cai H, Chan SK, Boone CE, Beiss V, Chariou PL, Steinmetz NF. Plant Viruses and Bacteriophage-Based Reagents for Diagnosis and Therapy. Annu Rev Virol 2020; 7:559-587. [PMID: 32991265 PMCID: PMC8018517 DOI: 10.1146/annurev-virology-010720-052252] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Viral nanotechnology exploits the prefabricated nanostructures of viruses, which are already abundant in nature. With well-defined molecular architectures, viral nanocarriers offer unprecedented opportunities for precise structural and functional manipulation using genetic engineering and/or bio-orthogonal chemistries. In this manner, they can be loaded with diverse molecular payloads for targeted delivery. Mammalian viruses are already established in the clinic for gene therapy and immunotherapy, and inactivated viruses or virus-like particles have long been used as vaccines. More recently, plant viruses and bacteriophages have been developed as nanocarriers for diagnostic imaging, vaccine and drug delivery, and combined diagnosis/therapy (theranostics). The first wave of these novel virus-based tools has completed clinical development and is poised to make an impact on clinical practice.
Collapse
Affiliation(s)
- Sourabh Shukla
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, USA
| | - He Hu
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, USA
| | - Hui Cai
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, USA
| | - Soo-Khim Chan
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, USA
| | - Christine E Boone
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, USA
| | - Veronique Beiss
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, USA
| | - Paul L Chariou
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, USA
| | - Nicole F Steinmetz
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, USA
- Department of Radiology, University of California, San Diego, La Jolla, California 92093, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, California 92093, USA
- Moores Cancer Center and Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, California 92093, USA;
| |
Collapse
|
73
|
Intrinsic Oncolytic Activity of Hoshino Mumps Virus Vaccine Strain Against Human Fibrosarcoma and Cervical Cancer Cell Lines. INTERNATIONAL JOURNAL OF CANCER MANAGEMENT 2020. [DOI: 10.5812/ijcm.103111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: The use of oncolytic viruses as therapeutic agents is a promising treatment for various human cancers. Several viruses have been extensively examined to achieve tumor cell death. Objectives: This study aimed at evaluating the natural oncolytic activity of mumps Hoshino vaccine strain against two human cancer cell lines, that is, HT1080 fibrosarcoma and HeLa cervical adenocarcinoma cell lines. Methods: The cytolytic activity of the virus was evaluated using an MTT assay. Apoptosis was detected by Annexin-V/propidium iodide (PI) staining and analyzed via flow cytometry. To indicate viral replication in vivo, nude mice with HeLa heterografts were treated with the Hoshino strain of mumps virus. Results: It was found that human fibrosarcoma and cervical cells were more sensitive to the mumps Hoshino strain, even at a very low multiplicity of infection (MOI) compared to normal human diploid cells. The results also showed that the Hoshino strain induced apoptosis in both cancer cells. A preliminary in vivo study revealed the significant suppression of tumor growth in the group treated with the mumps Hoshino strain compared to the control group. Conclusions: The Hoshino vaccine strain of mumps virus showed promising oncolytic activities against human fibrosarcoma and cervical adenocarcinoma cells.
Collapse
|
74
|
White K, Connor K, Clerkin J, Murphy BM, Salvucci M, O'Farrell AC, Rehm M, O'Brien D, Prehn JHM, Niclou SP, Lamfers MLM, Verreault M, Idbaih A, Verhaak R, Golebiewska A, Byrne AT. New hints towards a precision medicine strategy for IDH wild-type glioblastoma. Ann Oncol 2020; 31:1679-1692. [PMID: 32918998 DOI: 10.1016/j.annonc.2020.08.2336] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/18/2020] [Accepted: 08/31/2020] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma represents the most common primary malignancy of the central nervous system in adults and remains a largely incurable disease. The elucidation of disease subtypes based on mutational profiling, gene expression and DNA methylation has so far failed to translate into improved clinical outcomes. However, new knowledge emerging from the subtyping effort in the IDH-wild-type setting may provide directions for future precision therapies. Here, we review recent learnings in the field, and further consider how tumour microenvironment differences across subtypes may reveal novel contexts of vulnerability. We discuss recent treatment approaches and ongoing trials in the IDH-wild-type glioblastoma setting, and propose an integrated discovery stratagem incorporating multi-omics, single-cell technologies and computational approaches.
Collapse
Affiliation(s)
- K White
- Precision Cancer Medicine Group, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - K Connor
- Precision Cancer Medicine Group, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - J Clerkin
- Precision Cancer Medicine Group, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland; National Neurosurgical Department, Beaumont Hospital, Dublin, Ireland
| | - B M Murphy
- Centre for Systems Medicine, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - M Salvucci
- Centre for Systems Medicine, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - A C O'Farrell
- Precision Cancer Medicine Group, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - M Rehm
- Institute of Cell Biology and Immunology and Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| | - D O'Brien
- National Neurosurgical Department, Beaumont Hospital, Dublin, Ireland
| | - J H M Prehn
- Centre for Systems Medicine, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - S P Niclou
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg
| | - M L M Lamfers
- Department of Neurosurgery, Brain Tumor Center, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - M Verreault
- Sorbonne Université, Institut du Cerveau et de la Moelle Épinière, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neurologie, Paris, France
| | - A Idbaih
- Sorbonne Université, Institut du Cerveau et de la Moelle Épinière, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neurologie, Paris, France
| | - R Verhaak
- Jackson Laboratory for Genomic Medicine, Farmington, USA
| | - A Golebiewska
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg
| | - A T Byrne
- Precision Cancer Medicine Group, Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland.
| |
Collapse
|
75
|
Chai D, Qiu D, Zhang Z, Yuchen Shi S, Wang G, Fang L, Li H, Li H, Tian H, Zheng J. Absent in melanoma 2 enhances anti-tumour effects of CAIX promotor controlled conditionally replicative adenovirus in renal cancer. J Cell Mol Med 2020; 24:10744-10755. [PMID: 32725966 PMCID: PMC7521288 DOI: 10.1111/jcmm.15697] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 07/01/2020] [Accepted: 07/09/2020] [Indexed: 12/28/2022] Open
Abstract
Conditionally replicative adenoviruses (CRAds) were promising approach for solid tumour treatment, but its oncolytic efficiency and toxicity are still not satisfactory for further clinical application. Here, we developed the CAIX promotor (CAIXpromotor)‐controlled CRAd armed with a tumour suppressor absent in melanoma 2 (AIM2) to enhance its oncolytic potency. The CAIXpromotor‐AIM2 adenoviruses (Ad‐CAIXpromotor‐AIM2) could efficiently express E1A and AIM2 in renal cancer cells. Compared with Ad‐CAIXpromotor, Ad‐CAIXpromotor‐AIM2 significantly inhibited cell proliferation and enhanced cell apoptosis and cell killing, thus resulting in the oncolytic efficiency in 786‐O cells or OSRC‐2 cells. To explore the therapeutic effect, various Ads were intratumourally injected into OSRC‐2‐xenograft mice. The tumour growth was remarkably inhibited in Ad‐CAIXpromotor‐AIM2‐treated group as demonstrated by reduced tumour volume and weight with a low toxicity. The inflammasome inhibitor YVAD‐CMK resulted in the reduction of anti‐tumour activity by Ad‐CAIXpromotor‐AIM2 in vitro or in vivo, suggesting that inflammasome activation response was required for the enhanced therapeutic efficiency. Furthermore, lung metastasis of renal cancer mice was also suppressed by Ad‐CAIXpromotor‐AIM2 treatment accompanied by the decreased tumour fossil in lung tissues. These results indicated that the tumour‐specific Ad‐CAIXpromotor‐AIM2 could be applied for human renal cancer therapy. The therapeutic strategy of AIM2‐based CRAds could be a potential and promising approach for the therapy of primary solid or metastasis tumours.
Collapse
Affiliation(s)
- Dafei Chai
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China
| | - Dong Qiu
- Department of Urology, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China
| | - Zichun Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China
| | - Shang Yuchen Shi
- Department of Radiation Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China
| | - Gang Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China
| | - Lin Fang
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China
| | - Huizhong Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China
| | - Hailong Li
- Department of Urology, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China
| | - Hui Tian
- Cancer Institute, Xuzhou Medical University, Xuzhou, China.,Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China
| | - Junnian Zheng
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
76
|
Gilchrist VH, Jémus-Gonzalez E, Said A, Alain T. Kinase inhibitors with viral oncolysis: Unmasking pharmacoviral approaches for cancer therapy. Cytokine Growth Factor Rev 2020; 56:83-93. [PMID: 32690442 DOI: 10.1016/j.cytogfr.2020.07.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 07/02/2020] [Indexed: 12/28/2022]
Abstract
There are more than 500 kinases in the human genome, many of which are oncogenic once constitutively activated. Fortunately, numerous hyperactive kinases are druggable, and several targeted small molecule kinase inhibitors have demonstrated impressive clinical benefits in cancer treatment. However, their often cytostatic rather than cytotoxic effect on cancer cells, and the development of resistance mechanisms, remain significant limitations to these targeted therapies. Oncolytic viruses are an emerging class of immunotherapeutic agents with a specific oncotropic nature and excellent safety profile, highlighting them as a promising alternative to conventional therapeutic modalities. Nonetheless, the clinical efficacy of oncolytic virotherapy is challenged by immunological and physical barriers that limit viral delivery, replication, and spread within tumours. Several of these barriers are often associated with oncogenic kinase activity and, in some cases, worsened by the action of oncolytic viruses on kinase signaling during infection. What if inhibiting these kinases could potentiate the cancer-lytic and anti-tumour immune stimulating properties of oncolytic virotherapies? This could represent a paradigm shift in the use of specific kinase inhibitors in the clinic and provide a novel therapeutic approach to the treatment of cancers. A phase III clinical trial combining the oncolytic Vaccinia virus Pexa-Vec with the kinase inhibitor Sorafenib was initiated. While this trial failed to show any benefits over Sorafenib monotherapy in patients with advanced liver cancer, several pre-clinical studies demonstrate that targeting kinases combined with oncolytic viruses have synergistic effects highlighting this strategy as a unique avenue to cancer therapy. Herein, we review the combinations of oncolytic viruses with kinase inhibitors reported in the literature and discuss the clinical opportunities that represent these pharmacoviral approaches.
Collapse
Affiliation(s)
- Victoria Heather Gilchrist
- Children's Hospital of Eastern Ontario Research Institute, Apoptosis Research Center, Ottawa, ON, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada.
| | - Estephanie Jémus-Gonzalez
- Children's Hospital of Eastern Ontario Research Institute, Apoptosis Research Center, Ottawa, ON, Canada
| | - Aida Said
- Children's Hospital of Eastern Ontario Research Institute, Apoptosis Research Center, Ottawa, ON, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Tommy Alain
- Children's Hospital of Eastern Ontario Research Institute, Apoptosis Research Center, Ottawa, ON, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
77
|
Advances in Anti-Cancer Immunotherapy: Car-T Cell, Checkpoint Inhibitors, Dendritic Cell Vaccines, and Oncolytic Viruses, and Emerging Cellular and Molecular Targets. Cancers (Basel) 2020; 12:cancers12071826. [PMID: 32645977 PMCID: PMC7408985 DOI: 10.3390/cancers12071826] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/21/2020] [Accepted: 06/23/2020] [Indexed: 12/12/2022] Open
Abstract
Unlike traditional cancer therapies, such as surgery, radiation and chemotherapy that are typically non-specific, cancer immunotherapy harnesses the high specificity of a patient’s own immune system to selectively kill cancer cells. The immune system is the body’s main cancer surveillance system, but cancers may evade destruction thanks to various immune-suppressing mechanisms. We therefore need to deploy various immunotherapy-based strategies to help bolster the anti-tumour immune responses. These include engineering T cells to express chimeric antigen receptors (CARs) to specifically recognise tumour neoantigens, inactivating immune checkpoints, oncolytic viruses and dendritic cell (DC) vaccines, which have all shown clinical benefit in certain cancers. However, treatment efficacy remains poor due to drug-induced adverse events and immunosuppressive tendencies of the tumour microenvironment. Recent preclinical studies have unveiled novel therapies such as anti-cathepsin antibodies, galectin-1 blockade and anti-OX40 agonistic antibodies, which may be utilised as adjuvant therapies to modulate the tumour microenvironment and permit more ferocious anti-tumour immune response.
Collapse
|
78
|
Chaurasiya S, Fong Y, Warner SG. Optimizing Oncolytic Viral Design to Enhance Antitumor Efficacy: Progress and Challenges. Cancers (Basel) 2020; 12:cancers12061699. [PMID: 32604787 PMCID: PMC7352900 DOI: 10.3390/cancers12061699] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/19/2020] [Accepted: 06/23/2020] [Indexed: 12/15/2022] Open
Abstract
The field of oncolytic virotherapy has seen remarkable advancements in last two decades, leading to approval of the first oncolytic immuno-virotherapy, Talimogene Laherparepvec, for the treatment of melanoma. A plethora of preclinical and clinical studies have demonstrated excellent safety profiles of other oncolytic viruses. While oncolytic viruses show clinical promise in already immunogenic malignancies, response rates are inconsistent. Response rates are even less consistent in immunosuppressed tumor microenvironments like those found in liver, pancreas, and MSI-stable colon cancers. Therefore, the efficacy of oncolytic viruses needs to be improved for more oncolytic viruses to enter mainstream cancer therapy. One approach to increase the therapeutic efficacy of oncolytic viruses is to use them as primers for other immunotherapeutics. The amenability of oncolytic viruses to transgene-arming provides an immense opportunity for investigators to explore different ways of improving the outcome of oncolytic therapy. In this regard, genes encoding immunomodulatory proteins are the most commonly studied genes for arming oncolytic viruses. Other transgenes used to arm oncolytic viruses include those with the potential to favorably modulate tumor stroma, making it possible to image the virus distribution and increase its suitability for combination with other therapeutics. This review will detail the progress made in arming oncolytic viruses with a focus on immune-modulatory transgenes, and will discuss the challenges that need to be addressed for more armed oncolytic viruses to find widespread clinical use.
Collapse
|
79
|
Cervera-Carrascon V, Quixabeira DCA, Havunen R, Santos JM, Kutvonen E, Clubb JHA, Siurala M, Heiniö C, Zafar S, Koivula T, Lumen D, Vaha M, Garcia-Horsman A, Airaksinen AJ, Sorsa S, Anttila M, Hukkanen V, Kanerva A, Hemminki A. Comparison of Clinically Relevant Oncolytic Virus Platforms for Enhancing T Cell Therapy of Solid Tumors. MOLECULAR THERAPY-ONCOLYTICS 2020; 17:47-60. [PMID: 32322662 PMCID: PMC7163046 DOI: 10.1016/j.omto.2020.03.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 03/13/2020] [Indexed: 12/19/2022]
Abstract
Despite some promising results, the majority of patients do not benefit from T cell therapies, as tumors prevent T cells from entering the tumor, shut down their activity, or downregulate key antigens. Due to their nature and mechanism of action, oncolytic viruses have features that can help overcome many of the barriers currently facing T cell therapies of solid tumors. This study aims to understand how four different oncolytic viruses (adenovirus, vaccinia virus, herpes simplex virus, and reovirus) perform in that task. For that purpose, an immunocompetent in vivo tumor model featuring adoptive tumor-infiltrating lymphocyte (TIL) therapy was used. Tumor growth control (p < 0.001) and survival analyses suggest that adenovirus was most effective in enabling T cell therapy. The complete response rate was 62% for TILs + adenovirus versus 17.5% for TILs + PBS. Of note, TIL biodistribution did not explain efficacy differences between viruses. Instead, immunostimulatory shifts in the tumor microenvironment mirrored efficacy results. Overall, the use of oncolytic viruses can improve the utility of T cell therapies, and additional virus engineering by arming with transgenes can provide further antitumor effects. This phenomenon was seen when an unarmed oncolytic adenovirus was compared to Ad5/3-E2F-d24-hTNFa-IRES-hIL2 (TILT-123). A clinical trial is ongoing, where patients receiving TIL treatment also receive TILT-123 (ClinicalTrials.gov: NCT04217473).
Collapse
Affiliation(s)
- Victor Cervera-Carrascon
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, 00290 Helsinki, Finland.,TILT Biotherapeutics, 00290 Helsinki, Finland
| | - Dafne C A Quixabeira
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, 00290 Helsinki, Finland
| | - Riikka Havunen
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, 00290 Helsinki, Finland.,TILT Biotherapeutics, 00290 Helsinki, Finland
| | - Joao M Santos
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, 00290 Helsinki, Finland.,TILT Biotherapeutics, 00290 Helsinki, Finland
| | - Emma Kutvonen
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, 00290 Helsinki, Finland
| | - James H A Clubb
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, 00290 Helsinki, Finland.,TILT Biotherapeutics, 00290 Helsinki, Finland
| | - Mikko Siurala
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, 00290 Helsinki, Finland.,TILT Biotherapeutics, 00290 Helsinki, Finland
| | - Camilla Heiniö
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, 00290 Helsinki, Finland
| | - Sadia Zafar
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, 00290 Helsinki, Finland
| | - Teija Koivula
- Department of Chemistry, Radiochemistry, University of Helsinki, 00560 Helsinki, Finland
| | - Dave Lumen
- Department of Chemistry, Radiochemistry, University of Helsinki, 00560 Helsinki, Finland
| | - Marjo Vaha
- Regenerative Pharmacology Group, Division of Pharmacology and Pharmacotherapy, University of Helsinki, 00560 Helsinki, Finland
| | - Arturo Garcia-Horsman
- Regenerative Pharmacology Group, Division of Pharmacology and Pharmacotherapy, University of Helsinki, 00560 Helsinki, Finland
| | - Anu J Airaksinen
- Department of Chemistry, Radiochemistry, University of Helsinki, 00560 Helsinki, Finland
| | - Suvi Sorsa
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, 00290 Helsinki, Finland.,TILT Biotherapeutics, 00290 Helsinki, Finland
| | | | - Veijo Hukkanen
- Institute of Biomedicine, University of Turku, 20500 Turku, Finland
| | - Anna Kanerva
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, 00290 Helsinki, Finland.,Department of Obstetrics and Gynecology, Helsinki University Central Hospital, 00290 Helsinki, Finland
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, 00290 Helsinki, Finland.,TILT Biotherapeutics, 00290 Helsinki, Finland.,Helsinki University Hospital Comprehensive Cancer Center, 00290 Helsinki, Finland
| |
Collapse
|
80
|
Development of oncolytic virotherapy: from genetic modification to combination therapy. Front Med 2020; 14:160-184. [PMID: 32146606 PMCID: PMC7101593 DOI: 10.1007/s11684-020-0750-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 01/14/2020] [Indexed: 12/17/2022]
Abstract
Oncolytic virotherapy (OVT) is a novel form of immunotherapy using natural or genetically modified viruses to selectively replicate in and kill malignant cells. Many genetically modified oncolytic viruses (OVs) with enhanced tumor targeting, antitumor efficacy, and safety have been generated, and some of which have been assessed in clinical trials. Combining OVT with other immunotherapies can remarkably enhance the antitumor efficacy. In this work, we review the use of wild-type viruses in OVT and the strategies for OV genetic modification. We also review and discuss the combinations of OVT with other immunotherapies.
Collapse
|
81
|
Chaurasiya S, Yang A, Kang S, Lu J, Kim SI, Park AK, Sivanandam V, Zhang Z, Woo Y, Warner SG, Fong Y. Oncolytic poxvirus CF33-hNIS-ΔF14.5 favorably modulates tumor immune microenvironment and works synergistically with anti-PD-L1 antibody in a triple-negative breast cancer model. Oncoimmunology 2020; 9:1729300. [PMID: 32158622 PMCID: PMC7051185 DOI: 10.1080/2162402x.2020.1729300] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 12/31/2019] [Accepted: 01/06/2020] [Indexed: 12/28/2022] Open
Abstract
Triple-negative breast cancer is the most aggressive subtype of breast cancer and is difficult to treat. Breast cancer is considered to be poorly immunogenic and hence is less responsive to immunotherapies. We tested whether the oncolytic poxvirus CF33-hNIS-ΔF14.5 could modulate tumor immune microenvironment and make the tumors responsive to the immune checkpoint inhibitor anti-PD-L1. We found that virus infection causes the upregulation of PD-L1 levels on triple-negative breast cancer cells in vitro as well as in vivo in mice. In a mouse model of orthotopic triple-negative breast cancer, the virus was found to increase tumor infiltration by CD8+ T cells. Likewise, in mice treated with CF33-hNIS-ΔF14.5 high levels of proinflammatory cytokines IFNγ and IL-6 were found in the tumors but not in the serum. The levels of immune modulation were even higher in mice that were treated with a combination of the virus and anti-PD-L1 antibody. While CF33-hNIS-ΔF14.5 and anti-PD-L1 antibody failed to exert significant anti-tumor effect as a single agent, a combination of the two agents resulted in significant anti-tumor effect with 50% mice experiencing complete tumor regression when both agents were injected intra-tumorally. Furthermore, the ‘cured’ mice did not develop tumor after re-challenge with the same cancer cells suggesting that they developed immunity against those cancer cells. Taken together, our study shows that CF33-hNIS-ΔF14.5 favorably modulates tumor immune microenvironment in triple-negative breast cancer model making them responsive to the immune checkpoint inhibitor anti-PD-L1, and hence warrants further studies to determine the clinical applicability of this combination therapy.
Collapse
Affiliation(s)
| | - Annie Yang
- Department of Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Seonah Kang
- Department of Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Jianming Lu
- Department of Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Sang-In Kim
- Department of Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Anthony K Park
- Department of Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | | | - Zhifang Zhang
- Department of Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Yanghee Woo
- Department of Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Susanne G Warner
- Department of Surgery, City of Hope National Medical Center, Duarte, CA, USA
| | - Yuman Fong
- Department of Surgery, City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
82
|
Hadryś A, Sochanik A, McFadden G, Jazowiecka-Rakus J. Mesenchymal stem cells as carriers for systemic delivery of oncolytic viruses. Eur J Pharmacol 2020; 874:172991. [PMID: 32044323 DOI: 10.1016/j.ejphar.2020.172991] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 01/09/2020] [Accepted: 02/04/2020] [Indexed: 12/13/2022]
Abstract
Progress in genetic engineering led to the emergence of some viruses as potent anticancer therapeutics. These oncolytic viruses combine self-amplification with dual antitumor action: oncolytic (destruction of cancer cells) and immunostimulatory (eliciting acquired antitumor response against cancer epitopes). As any other viruses, they trigger antiviral response upon systemic administration. Mesenchymal stem cells are immature cells capable of self-renewing and differentiating into many cell types that belong to three germinal layers. Due to their inherent tumor tropism mesenchymal stem cells loaded with oncolytic virus can improve delivery of the therapeutic cargo to cancer sites. Shielding of oncolytic viral construct from antiviral host immune response makes these cells prospective delivery vehicles to even hard-to-reach metastatic neoplastic foci. Use of mesenchymal stem cells has been criticized by some investigators as limiting proliferative abilities of primary cells and increasing the risk of malignant transformation, as well as attenuating therapeutic responses. However, majority of preclinical studies indicate safety and efficacy of mesenchymal stem cells used as carriers of oncolytic viruses. In view of contradictory postulates, the debate continues. The review discusses mesenchymal stem cells as carriers for delivery of genetically engineered oncolytic constructs and focuses on systemic approach to oncoviral treatment of some deadly neoplasms.
Collapse
Affiliation(s)
- Agata Hadryś
- Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland; Institute of Chemistry, University of Silesia, Poland.
| | - Aleksander Sochanik
- Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland.
| | - Grant McFadden
- Biodesign Institute, Arizona State University, Tempe, AZ, USA.
| | | |
Collapse
|
83
|
Engineering Newcastle Disease Virus as an Oncolytic Vector for Intratumoral Delivery of Immune Checkpoint Inhibitors and Immunocytokines. J Virol 2020; 94:JVI.01677-19. [PMID: 31694938 DOI: 10.1128/jvi.01677-19] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 10/29/2019] [Indexed: 12/26/2022] Open
Abstract
Newcastle disease virus (NDV) is an attractive candidate for oncolytic immunotherapy due to its ability to replicate in tumor cells and potentially to overcome the inherently immunosuppressive nature of the tumor microenvironment. The advent of checkpoint blockade immunotherapy over the past few years represents a paradigm shift in cancer therapy. However, the prevalence of severe immune-related adverse events with CTLA4 and PD1 pathway blockade in clinical studies, especially in combination therapy groups, is a cause for concern. Immunotherapies with cytokines have also been extensively explored, but they have been associated with adverse events in clinical trials. Oncolytic vectors engineered to express checkpoint blockade antibodies and cytokines could provide an avenue for reducing the clinical toxicity associated with systemic therapy by concentrating the immunomodulatory payload at the site of disease. In this study, we engineered six different recombinant viruses: NDVs expressing checkpoint inhibitors (rNDV-anti-PD1 and rNDV-anti-PDL1); superagonists (rNDV-anti-CD28); and immunocytokines, where the antibodies are fused to an immunostimulatory cytokine, such as interleukin 12 (IL-12) (rNDV-anti-CD28-murine IL-12 [mIL-12], rNDV-anti-PD1-mIL-12, and rNDV-anti-PDL1-mIL-12). These six engineered viruses induced tumor control and survival benefits in both highly aggressive unilateral and bilateral B16-F10 murine melanoma models, indicative of an abscopal effect. The data represent a strong proof of concept on which further clinical evaluation could build.IMPORTANCE Checkpoint inhibitor therapy has shown tremendous efficacy, but also frequent and often severe side effects-especially when multiple drugs of the class are used simultaneously. Similarly, many investigational immunotherapy agents, which have shown promise in animal models, have failed in clinical trials due to dose-limiting toxicity when administered systemically. This study utilized a murine melanoma model to evaluate the efficacy of intratumoral injections of recombinant NDVs engineered to express multiple immunotherapeutic proteins with well-documented side effects in humans. Our results indicate that intratumoral administration of these recombinant NDVs, particularly when combined with systemic CTLA4 checkpoint inhibition, exerts a robust effect in treated and nontreated tumors, indicative of a systemic antitumoral response. The intratumoral delivery of rNDVs expressing immunotherapeutic proteins may be an effective method of targeting the immune cell populations most relevant for antitumoral immunity and allowing us to restrict the use of systemic immunotherapy agents.
Collapse
|
84
|
Lemos de Matos A, Franco LS, McFadden G. Oncolytic Viruses and the Immune System: The Dynamic Duo. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 17:349-358. [PMID: 32071927 PMCID: PMC7015832 DOI: 10.1016/j.omtm.2020.01.001] [Citation(s) in RCA: 156] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Oncolytic viruses (OVs) constitute a new and promising immunotherapeutic approach toward cancer treatment. This therapy takes advantage of the natural propensity of most tumor cells to be infected by specific OVs. Besides the direct killing potential (oncolysis), what makes OV administration attractive for the present cancer immunotherapeutic scenario is the capacity to induce two new overlapping, but distinct, immunities: anti-tumoral and anti-viral. OV infection and oncolysis naturally elicit both innate and adaptive immune responses (required for long-term anti-tumoral immunity); at the same time, the viral infection prompts an anti-viral response. In this review, we discuss the dynamic interaction between OVs and the triggered responses of the immune system. The anti-OV immunological events that lead to viral clearance and the strategies to deal with such potential loss of the therapeutic virus are discussed. Additionally, we review the immune stimulatory actions induced by OVs through different inherent strategies, such as modulation of the tumor microenvironment, the role of immunogenic cell death, and the consequences of genetically modifying OVs by arming them with therapeutic transgenes. An understanding of the balance between the OV-induced anti-tumoral versus anti-viral immunities will provide insight when choosing the appropriate virotherapy for any specific cancer.
Collapse
Affiliation(s)
- Ana Lemos de Matos
- Biodesign Center for Immunotherapy, Vaccines, and Virotherapy (B-CIVV), The Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Lina S Franco
- Biodesign Center for Immunotherapy, Vaccines, and Virotherapy (B-CIVV), The Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| | - Grant McFadden
- Biodesign Center for Immunotherapy, Vaccines, and Virotherapy (B-CIVV), The Biodesign Institute, Arizona State University, Tempe, AZ 85287, USA
| |
Collapse
|
85
|
Hutzen B, Paudel SN, Naeimi Kararoudi M, Cassady KA, Lee DA, Cripe TP. Immunotherapies for pediatric cancer: current landscape and future perspectives. Cancer Metastasis Rev 2019; 38:573-594. [PMID: 31828566 PMCID: PMC6994452 DOI: 10.1007/s10555-019-09819-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The advent of immunotherapy has revolutionized how we manage and treat cancer. While the majority of immunotherapy-related studies performed to date have focused on adult malignancies, a handful of these therapies have also recently found success within the pediatric space. In this review, we examine the immunotherapeutic agents that have achieved the approval of the US Food and Drug Administration for treating childhood cancers, highlighting their development, mechanisms of action, and the lessons learned from the seminal clinical trials that ultimately led to their approval. We also shine a spotlight on several emerging immunotherapeutic modalities that we believe are poised to have a positive impact on the treatment of pediatric malignancies in the near future.
Collapse
Affiliation(s)
- Brian Hutzen
- The Abigail Wexner Research Institute at Nationwide Children's Hospital Center for Childhood Cancer and Blood Disorders, 575 Children's Crossroad, Columbus, OH, 43215, USA
| | - Siddhi Nath Paudel
- The Abigail Wexner Research Institute at Nationwide Children's Hospital Center for Childhood Cancer and Blood Disorders, 575 Children's Crossroad, Columbus, OH, 43215, USA
- Graduate Program in Molecular, Cellular and Developmental Biology, The Ohio State University, Columbus, OH, USA
| | - Meisam Naeimi Kararoudi
- The Abigail Wexner Research Institute at Nationwide Children's Hospital Center for Childhood Cancer and Blood Disorders, 575 Children's Crossroad, Columbus, OH, 43215, USA
| | - Kevin A Cassady
- The Abigail Wexner Research Institute at Nationwide Children's Hospital Center for Childhood Cancer and Blood Disorders, 575 Children's Crossroad, Columbus, OH, 43215, USA
- Graduate Program in Molecular, Cellular and Developmental Biology, The Ohio State University, Columbus, OH, USA
- Division of Hematology/Oncology/BMT, Department of Pediatrics, Nationwide Children's Hospital, Columbus, OH, USA
- Ohio State University Wexner College of Medicine, Columbus, OH, USA
| | - Dean A Lee
- The Abigail Wexner Research Institute at Nationwide Children's Hospital Center for Childhood Cancer and Blood Disorders, 575 Children's Crossroad, Columbus, OH, 43215, USA
- Graduate Program in Molecular, Cellular and Developmental Biology, The Ohio State University, Columbus, OH, USA
- Division of Hematology/Oncology/BMT, Department of Pediatrics, Nationwide Children's Hospital, Columbus, OH, USA
- Ohio State University Wexner College of Medicine, Columbus, OH, USA
| | - Timothy P Cripe
- The Abigail Wexner Research Institute at Nationwide Children's Hospital Center for Childhood Cancer and Blood Disorders, 575 Children's Crossroad, Columbus, OH, 43215, USA.
- Graduate Program in Molecular, Cellular and Developmental Biology, The Ohio State University, Columbus, OH, USA.
- Division of Hematology/Oncology/BMT, Department of Pediatrics, Nationwide Children's Hospital, Columbus, OH, USA.
- Ohio State University Wexner College of Medicine, Columbus, OH, USA.
| |
Collapse
|