1
|
Sun W, Hu S, Wang X. Advances and clinical applications of immune checkpoint inhibitors in hematological malignancies. Cancer Commun (Lond) 2024; 44:1071-1097. [PMID: 39073258 PMCID: PMC11492363 DOI: 10.1002/cac2.12587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 06/09/2024] [Accepted: 06/25/2024] [Indexed: 07/30/2024] Open
Abstract
Immune checkpoints are differentially expressed on various immune cells to regulate immune responses in tumor microenvironment. Tumor cells can activate the immune checkpoint pathway to establish an immunosuppressive tumor microenvironment and inhibit the anti-tumor immune response, which may lead to tumor progression by evading immune surveillance. Interrupting co-inhibitory signaling pathways with immune checkpoint inhibitors (ICIs) could reinvigorate the anti-tumor immune response and promote immune-mediated eradication of tumor cells. As a milestone in tumor treatment, ICIs have been firstly used in solid tumors and subsequently expanded to hematological malignancies, which are in their infancy. Currently, immune checkpoints have been investigated as promising biomarkers and therapeutic targets in hematological malignancies, and novel immune checkpoints, such as signal regulatory protein α (SIRPα) and tumor necrosis factor-alpha-inducible protein 8-like 2 (TIPE2), are constantly being discovered. Numerous ICIs have received clinical approval for clinical application in the treatment of hematological malignancies, especially when used in combination with other strategies, including oncolytic viruses (OVs), neoantigen vaccines, bispecific antibodies (bsAb), bio-nanomaterials, tumor vaccines, and cytokine-induced killer (CIK) cells. Moreover, the proportion of individuals with hematological malignancies benefiting from ICIs remains lower than expected due to multiple mechanisms of drug resistance and immune-related adverse events (irAEs). Close monitoring and appropriate intervention are needed to mitigate irAEs while using ICIs. This review provided a comprehensive overview of immune checkpoints on different immune cells, the latest advances of ICIs and highlighted the clinical applications of immune checkpoints in hematological malignancies, including biomarkers, targets, combination of ICIs with other therapies, mechanisms of resistance to ICIs, and irAEs, which can provide novel insight into the future exploration of ICIs in tumor treatment.
Collapse
Affiliation(s)
- Wenyue Sun
- Department of HematologyShandong Provincial HospitalShandong UniversityJinanShandongP. R. China
| | - Shunfeng Hu
- Department of HematologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP. R. China
| | - Xin Wang
- Department of HematologyShandong Provincial HospitalShandong UniversityJinanShandongP. R. China
- Department of HematologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP. R. China
- Taishan Scholars Program of Shandong ProvinceJinanShandongP. R. China
- Branch of National Clinical Research Center for Hematologic DiseasesJinanShandongP. R. China
- National Clinical Research Center for Hematologic Diseasesthe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuP. R. China
| |
Collapse
|
2
|
Bastin DJ, Quizi J, Kennedy MA, Kekre N, Auer RC. Current challenges in the manufacture of clinical-grade autologous whole cell vaccines for hematological malignancies. Cytotherapy 2022; 24:979-989. [PMID: 35562303 DOI: 10.1016/j.jcyt.2022.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/21/2022] [Accepted: 03/21/2022] [Indexed: 11/03/2022]
Abstract
Autologous whole cell vaccines use a patient's own tumor cells as a source of antigen to elicit an anti-tumor immune response in vivo. Recently, the authors conducted a systematic review of clinical trials employing these products in hematological cancers that showed a favorable safety profile and trend toward efficacy. However, it was noted that manufacturing challenges limit both the efficacy and clinical implementation of these vaccine products. In the current literature review, the authors sought to define the issues surrounding the manufacture of autologous whole cell products for hematological cancers. The authors describe key factors, including the acquisition, culture, cryopreservation and transduction of malignant cells, that require optimization for further advancement of the field. Furthermore, the authors provide a summary of pre-clinical work that informs how the identified challenges may be overcome. The authors also highlight areas in which future basic research would be of benefit to the field. The goal of this review is to provide a roadmap for investigators seeking to advance the field of autologous cell vaccines as it applies to hematological malignancies.
Collapse
Affiliation(s)
- Donald J Bastin
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada; Schulich School of Medicine, Western University, London, Canada
| | - Jennifer Quizi
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Michael A Kennedy
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada
| | - Natasha Kekre
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada; Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Rebecca C Auer
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Canada; Faculty of Medicine, University of Ottawa, Ottawa, Canada; Department of Surgery, University of Ottawa, Ottawa, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada.
| |
Collapse
|
3
|
Batalla-Covello J, Ngai HW, Flores L, McDonald M, Hyde C, Gonzaga J, Hammad M, Gutova M, Portnow J, Synold T, Curiel DT, Lesniak MS, Aboody KS, Mooney R. Multiple Treatment Cycles of Neural Stem Cell Delivered Oncolytic Adenovirus for the Treatment of Glioblastoma. Cancers (Basel) 2021; 13:6320. [PMID: 34944938 PMCID: PMC8699772 DOI: 10.3390/cancers13246320] [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: 11/04/2021] [Accepted: 12/09/2021] [Indexed: 11/16/2022] Open
Abstract
Tumor tropic neural stem cells (NSCs) can improve the anti-tumor efficacy of oncovirotherapy agents by protecting them from rapid clearance by the immune system and delivering them to multiple distant tumor sites. We recently completed a first-in-human trial assessing the safety of a single intracerebral dose of NSC-delivered CRAd-Survivin-pk7 (NSC.CRAd-S-pk7) combined with radiation and chemotherapy in newly diagnosed high-grade glioma patients. The maximum feasible dose was determined to be 150 million NSC.CRAd-Sp-k7 (1.875 × 1011 viral particles). Higher doses were not assessed due to volume limitations for intracerebral administration and the inability to further concentrate the study agent. It is possible that therapeutic efficacy could be maximized by administering even higher doses. Here, we report IND-enabling studies in which an improvement in treatment efficacy is achieved in immunocompetent mice by administering multiple treatment cycles intracerebrally. The results imply that pre-existing immunity does not preclude therapeutic benefits attainable by administering multiple rounds of an oncolytic adenovirus directly into the brain.
Collapse
Affiliation(s)
- Jennifer Batalla-Covello
- Department of Developmental and Stem Cell Biology, City of Hope, Duarte, CA 91010, USA; (J.B.-C.); (H.W.N.); (L.F.); (M.M.); (C.H.); (J.G.); (M.H.); (M.G.)
| | - Hoi Wa Ngai
- Department of Developmental and Stem Cell Biology, City of Hope, Duarte, CA 91010, USA; (J.B.-C.); (H.W.N.); (L.F.); (M.M.); (C.H.); (J.G.); (M.H.); (M.G.)
| | - Linda Flores
- Department of Developmental and Stem Cell Biology, City of Hope, Duarte, CA 91010, USA; (J.B.-C.); (H.W.N.); (L.F.); (M.M.); (C.H.); (J.G.); (M.H.); (M.G.)
| | - Marisa McDonald
- Department of Developmental and Stem Cell Biology, City of Hope, Duarte, CA 91010, USA; (J.B.-C.); (H.W.N.); (L.F.); (M.M.); (C.H.); (J.G.); (M.H.); (M.G.)
| | - Caitlyn Hyde
- Department of Developmental and Stem Cell Biology, City of Hope, Duarte, CA 91010, USA; (J.B.-C.); (H.W.N.); (L.F.); (M.M.); (C.H.); (J.G.); (M.H.); (M.G.)
| | - Joanna Gonzaga
- Department of Developmental and Stem Cell Biology, City of Hope, Duarte, CA 91010, USA; (J.B.-C.); (H.W.N.); (L.F.); (M.M.); (C.H.); (J.G.); (M.H.); (M.G.)
| | - Mohamed Hammad
- Department of Developmental and Stem Cell Biology, City of Hope, Duarte, CA 91010, USA; (J.B.-C.); (H.W.N.); (L.F.); (M.M.); (C.H.); (J.G.); (M.H.); (M.G.)
| | - Margarita Gutova
- Department of Developmental and Stem Cell Biology, City of Hope, Duarte, CA 91010, USA; (J.B.-C.); (H.W.N.); (L.F.); (M.M.); (C.H.); (J.G.); (M.H.); (M.G.)
| | - Jana Portnow
- Department of Medical Oncology, City of Hope, Duarte, CA 91010, USA;
| | - Tim Synold
- Department of Cancer Biology, City of Hope, Duarte, CA 91010, USA;
| | - David T. Curiel
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA;
| | - Maciej S. Lesniak
- Department of Neurological Surgery, Northwestern University, Chicago, IL 60611, USA;
| | - Karen S. Aboody
- Department of Developmental and Stem Cell Biology, City of Hope, Duarte, CA 91010, USA; (J.B.-C.); (H.W.N.); (L.F.); (M.M.); (C.H.); (J.G.); (M.H.); (M.G.)
| | - Rachael Mooney
- Department of Developmental and Stem Cell Biology, City of Hope, Duarte, CA 91010, USA; (J.B.-C.); (H.W.N.); (L.F.); (M.M.); (C.H.); (J.G.); (M.H.); (M.G.)
| |
Collapse
|
4
|
Huang FY, Wang JY, Dai SZ, Lin YY, Sun Y, Zhang L, Lu Z, Cao R, Tan GH. A recombinant oncolytic Newcastle virus expressing MIP-3α promotes systemic antitumor immunity. J Immunother Cancer 2021; 8:jitc-2019-000330. [PMID: 32759233 PMCID: PMC7410001 DOI: 10.1136/jitc-2019-000330] [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] [Accepted: 07/06/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The oncolytic Newcastle disease virus (NDV) is inherently able to trigger the lysis of tumor cells and induce the immunogenic cell death (ICD) of tumor cells and is also an excellent gene-engineering vector. The macrophage inflammatory protein-3α (MIP-3α) is a specific chemokine for dendritic cells (DCs). Thus, we constructed a recombinant NDV expressing MIP-3α (NDV-MIP3α) as an in vivo DC vaccine for amplifying antitumor immunities. METHODS The recombinant NDV-MIP3α was constructed by the insertion of MIP-3α cDNA between the P and M genes. Western blotting assay and ELISA were used to detect MIP-3α, HMGB1, IgG, and ATP in the supernatant and sera. The chemotaxis of DCs was examined by Transwell chambers. The phenotypes of the immune cells (eg, DCs) were analyzed by flow cytometry. The antitumor efficiency of NDV-MIP3α was observed in B16 and CT26 tumor-bearing mice. Immunofluorescence and immunohistochemistry were applied to observe the ecto-calreticulin (CRT) and intratumoral attraction of DCs. Adoptive transfer of splenocytes and antibodies and depletion of T-cell subsets were used to evaluate the relationship between antitumor immunities and the role of the T-cell subtype. RESULTS The findings show that NDV-MIP3α has almost the same capabilities of tumor lysis and induction of ICD as the wild-type NDV (NDV-WT). MIP-3α secreted by NDV-MIP3α could successfully attract DCs in vitro and in vivo. Both B16 and CT26 cells infected with NDV-MIP3α could strongly promote DC maturation and activation. Compared with NDV-WT, intratumoral injection of NDV-MIP3α and the adoptive transfer of T lymphocytes from mice injected with NDV-MIP3α resulted in a significant suppression of B16 and CT26 tumor growth. The NDV-MIP3α-induced production of tumor-specific cellular and humoral immune responses was dependent on CD8+ T cells and partially on CD4+ T cells. A significant reversion of tumor microenvironments was found in the mice injected with NDV-MIP3α. CONCLUSIONS Compared with NDV-WT, the recombinant NDV-MIP3α as an in vivo DC vaccine demonstrates enhanced antitumor activities through the induction of stronger system immunities and modulation of the tumor microenvironment. This strategy may be a potential approach for the generation of an in vivo DC vaccine.
Collapse
Affiliation(s)
- Feng-Ying Huang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Jin-Yan Wang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Shu-Zhen Dai
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Ying-Ying Lin
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Yan Sun
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Liming Zhang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Zhuoxuan Lu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Rong Cao
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| | - Guang-Hong Tan
- Key Laboratory of Tropical Translational Medicine of Ministry of Education & Hainan Provincial Key Laboratory of Tropical Medicine, Hainan Medical University, Haikou, Hainan, China
| |
Collapse
|
5
|
Niavarani SR, Lawson C, Boudaud M, Simard C, Tai LH. Oncolytic vesicular stomatitis virus-based cellular vaccine improves triple-negative breast cancer outcome by enhancing natural killer and CD8 + T-cell functionality. J Immunother Cancer 2021; 8:jitc-2019-000465. [PMID: 32179632 PMCID: PMC7073779 DOI: 10.1136/jitc-2019-000465] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2020] [Indexed: 01/19/2023] Open
Affiliation(s)
- Seyedeh-Raheleh Niavarani
- Immunology and Cell Biology, Université de Sherbrooke, Faculté de médecine et des sciences de la santé, Sherbrooke, Quebec, Canada
| | - Christine Lawson
- Immunology and Cell Biology, Université de Sherbrooke, Faculté de médecine et des sciences de la santé, Sherbrooke, Quebec, Canada
| | - Marie Boudaud
- Pediatrics, Université de Sherbrooke, Faculté de médecine et des sciences de la santé, Sherbrooke, Quebec, Canada
| | - Camille Simard
- Pharmacology and Physiology, Université de Sherbrooke, Faculté de médecine et des sciences de la santé, Sherbrooke, Quebec, Canada
| | - Lee-Hwa Tai
- Immunology and Cell Biology, Université de Sherbrooke, Faculté de médecine et des sciences de la santé, Sherbrooke, Quebec, Canada .,Centre de recherche du CHUS, Sherbrooke, Quebec, Canada
| |
Collapse
|
6
|
Oncolytic virotherapy in hematopoietic stem cell transplantation. Hum Immunol 2021; 82:640-648. [PMID: 34119352 DOI: 10.1016/j.humimm.2021.05.007] [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: 04/08/2021] [Revised: 05/29/2021] [Accepted: 05/29/2021] [Indexed: 12/28/2022]
Abstract
Hematopoietic stem cell transplantation (HSCT) is a curative option for various hematologic malignancies. However, fatal complications, such as relapse and graft-versus-host disease (GVHD) hampered favorable HSCT outcomes. Cancer cells remained in the body following the conditioning regimen, or those contaminating the autologous graft can cause relapse. Although the relapse is much lesser in allogeneic HSCT, GVHD is still a life-threatening complication in this type of HSCT. Researchers are seeking various strategies to reduce relapse and GVHD in HSCT with minimum effects on the engraftment and immune-reconstitution. Oncolytic viruses (OVs) are emerging anti-cancer agents with promising results in battling solid tumors. OVs can selectively replicate in the malignant cells in which the antiviral immune responses have defected. Hence, they could be used as a purging agent to eradicate the tumoral contamination of autologous grafts with no damages to hematopoietic stem cells. Moreover, they have been shown to alleviate GVHD complications through modulating alloreactive T cell responses. Primary results promise using OVs as a strategy to reduce both relapse and GVHD in the HSCT without affecting hematologic and immunologic engraftment. Herein, we provide the latest findings in the field of OV therapy in HSCT and discuss their pros and cons.
Collapse
|
7
|
Hyaluronidase expression within tumors increases virotherapy efficacy and T cell accumulation. MOLECULAR THERAPY-ONCOLYTICS 2021; 22:27-35. [PMID: 34377767 PMCID: PMC8321894 DOI: 10.1016/j.omto.2021.05.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 05/19/2021] [Indexed: 12/28/2022]
Abstract
Oncolytic viruses (OVs) preferentially infect and selectively replicate in cancer cells. OVs have been tested in clinical trials as monotherapy or in combination with chemotherapy, radiotherapy, and immunotherapy. However, the dense extracellular matrix hampers the intratumoral spreading and efficacy of OVs. Previously we described VCN-01, an oncolytic adenovirus expressing a soluble version of human sperm hyaluronidase (hyal) PH20, which exhibited enhanced intratumoral distribution and antitumor activity in different models. Here, we present two oncolytic adenoviruses designed to increase the secretion of PH20 compared to VCN-01. ICO15K-40SAPH20, encoding PH20 under an Ad40 splice acceptor, and ICO15K-E1aPH20 expressing PH20 fused to the E1A gene by P2A peptide. We demonstrate that increased hyal activity improves antitumor efficacy in both a sensitive immunodeficient model and an immunocompetent model. Moreover, we show that hyal activity impacts T cell accumulation in tumors, highlighting the value of a hyaluronidase-expressing virus for combinations with other immunotherapies in cancers involving dense stroma.
Collapse
|
8
|
Bastin DJ, Khan ST, Montroy J, Kennedy MA, Forbes N, Martel AB, Baker L, Gresham L, Boucher DM, Wong B, Shorr R, Diallo JS, Fergusson DA, Lalu MM, Auer RC, Kekre N. Safety and efficacy of autologous whole cell vaccines in hematologic malignancies: A systematic review and meta-analysis. Hematol Oncol 2021; 39:448-464. [PMID: 33963789 DOI: 10.1002/hon.2875] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 04/26/2021] [Indexed: 01/07/2023]
Abstract
Autologous cell vaccines use a patient's tumor cells to stimulate a broad antitumor response in vivo. This approach shows promise for treating hematologic cancers in early phase clinical trials, but overall safety and efficacy remain poorly described. We conducted a systematic review assessing the use of autologous cell vaccination in treating hematologic cancers. Primary outcomes of interest were safety and clinical response, with secondary outcomes including survival, relapse rate, correlative immune assays and health-quality related metrics. We performed a search of MEDLINE, Embase and the Cochrane Register of Controlled Trials including any interventional trial employing an autologous, whole cell product in any hematologic malignancy. Risk of bias was assessed using a modified Institute of Health Economics tool. Across 20 single arm studies, only 341 of 592 enrolled participants received one or more vaccinations. Primary reasons for not receiving vaccination included rapid disease progression/death and manufacturing challenges. Overall, few high-grade adverse events were observed. One death was reported and attributed to a GM-CSF producing allogeneic cell line co-administered with the autologous vaccine. Of 58 evaluable patients, the complete response rate was 21.0% [95% CI, 10.4%-37.8%)] and overall response rate was 35.8% (95% CI, 24.4%-49.0%). Of 97 evaluable patients for survival, the 5-years overall survival rate was 64.9% (95% CI, 52.6%-77.2%) and disease-free survival was 59.7% (95% CI, 47.7%-71.7%). We conclude that, in hematologic malignancies, based on limited available data, autologous cell vaccines are safe and display a trend towards efficacy but that challenges exist in vaccine manufacture and administration.
Collapse
Affiliation(s)
- Donald J Bastin
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Schulich School of Medicine, Western University, London, ON, Canada
| | - Sarwat T Khan
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Joshua Montroy
- Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Michael A Kennedy
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Nicole Forbes
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Andre B Martel
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Surgery, University of Ottawa, Ottawa, Ontario, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Laura Baker
- Department of Surgery, University of Ottawa, Ottawa, Ontario, Canada
| | - Louise Gresham
- Department of Surgery, University of Ottawa, Ottawa, Ontario, Canada
| | - Dominique M Boucher
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Boaz Wong
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Risa Shorr
- Learning Services, The Ottawa Hospital, Ottawa, ON, Canada
| | - Jean-Simon Diallo
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Dean A Fergusson
- Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada.,School of Epidemiology and Public Health, University of Ottawa, Ottawa, Ontario, Canada
| | - Manoj M Lalu
- Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada.,Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada.,Regenerative Medicine Program, The Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Rebecca C Auer
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Department of Surgery, University of Ottawa, Ottawa, Ontario, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada.,Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Natasha Kekre
- Cancer Therapeutics Program, The Ottawa Hospital Research Institute, Ottawa, ON, Canada.,Faculty of Medicine, University of Ottawa, Ottawa, Canada.,Department of Medicine and The Ottawa Hospital, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
9
|
Muñoz-Alía MÁ, Nace RA, Tischer A, Zhang L, Bah ES, Auton M, Russell SJ. MeV-Stealth: A CD46-specific oncolytic measles virus resistant to neutralization by measles-immune human serum. PLoS Pathog 2021; 17:e1009283. [PMID: 33534834 PMCID: PMC7886131 DOI: 10.1371/journal.ppat.1009283] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 02/16/2021] [Accepted: 01/05/2021] [Indexed: 02/07/2023] Open
Abstract
The frequent overexpression of CD46 in malignant tumors has provided a basis to use vaccine-lineage measles virus (MeV) as an oncolytic virotherapy platform. However, widespread measles seropositivity limits the systemic deployment of oncolytic MeV for the treatment of metastatic neoplasia. Here, we report the development of MeV-Stealth, a modified vaccine MeV strain that exhibits oncolytic properties and escapes antimeasles antibodies in vivo. We engineered this virus using homologous envelope glycoproteins from the closely-related but serologically non-cross reactive canine distemper virus (CDV). By fusing a high-affinity CD46 specific single-chain antibody fragment (scFv) to the CDV-Hemagglutinin (H), ablating its tropism for human nectin-4 and modifying the CDV-Fusion (F) signal peptide we achieved efficient retargeting to CD46. A receptor binding affinity of ~20 nM was required to trigger CD46-dependent intercellular fusion at levels comparable to the original MeV H/F complex and to achieve similar antitumor efficacy in myeloma and ovarian tumor-bearing mice models. In mice passively immunized with measles-immune serum, treatment of ovarian tumors with MeV-Stealth significantly increased overall survival compared with treatment with vaccine-lineage MeV. Our results show that MeV-Stealth effectively targets and lyses CD46-expressing cancer cells in mouse models of ovarian cancer and myeloma, and evades inhibition by human measles-immune serum. MeV-Stealth could therefore represent a strong alternative to current oncolytic MeV strains for treatment of measles-immune cancer patients. Vaccine strains of the measles virus (MeV) have been shown to be promising anti-cancer agents because of the frequent overexpression of the host-cell receptor CD46 in human malignancies. However, anti-MeV antibodies in the human population severely restrict the use of MeV as an oncolytic agent. Here, we engineered a neutralization-resistant MeV vaccine, MeV-Stealth, by replacing its envelope glycoproteins with receptor-targeted glycoproteins from wild-type canine distemper virus. By fully-retargeting the new envelope to the receptor CD46, we found that in mouse models of ovarian cancer and myeloma MeV-Stealth displayed oncolytic properties similar to the parental MeV vaccine. Furthermore, we found that passive immunization with measles-immune human serum did not eliminate the oncolytic potency of the MeV-Stealth, whereas it did destroy the potency of the parental MeV strain. The virus we here report may be considered a suitable oncolytic agent for the treatment of MeV-immune patients.
Collapse
Affiliation(s)
- Miguel Ángel Muñoz-Alía
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail: (MÁM-A); (SJR)
| | - Rebecca A. Nace
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Alexander Tischer
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Lianwen Zhang
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Eugene S. Bah
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, United States of America
| | - Matthew Auton
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Stephen J. Russell
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
- Division of Hematology, Mayo Clinic, Rochester, Minnesota, United States of America
- * E-mail: (MÁM-A); (SJR)
| |
Collapse
|
10
|
Shao X, Wang X, Guo X, Jiang K, Ye T, Chen J, Fang J, Gu L, Wang S, Zhang G, Meng S, Xu Q. STAT3 Contributes To Oncolytic Newcastle Disease Virus-Induced Immunogenic Cell Death in Melanoma Cells. Front Oncol 2019; 9:436. [PMID: 31192135 PMCID: PMC6548873 DOI: 10.3389/fonc.2019.00436] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 05/07/2019] [Indexed: 12/15/2022] Open
Abstract
Background: Oncolytic viruses (OVs) are emerging as potent inducers of immunogenic cell death (ICD), releasing danger-associated molecular patterns (DAMPs) that induce potent anticancer immunity. Oncolytic Newcastle disease virus (NDV) has been shown to educe ICD in both glioma and lung cancer cells. The objective of this study is to investigate whether oncolytic NDV induces ICD in melanoma cells and how it is regulated. Methods: Various time points were actuated to check the expression and release of ICD markers induced by NDV strain, NDV/FMW in melanoma cell lines. The expression and release of ICD markers induced by oncolytic NDV strain, NDV/FMW, in melanoma cell lines at various time points were determined. Surface-exposed calreticulin (CRT) was inspected by confocal imaging. The supernatants of NDV/FMW infected cells were collected and concentrated for the determination of ATP secretion by ELISA, HMGB1, and HSP70/90 expression by immunoblot (IB) analysis. Pharmacological inhibition of apoptosis, autophagy, necroptosis, ER Stress, and STAT3 (signal transducer and activator of transcription 3) was achieved by treatment with small molecule inhibitors. Melanoma cell lines stably depleted of STAT3 were established with lentiviral constructs. Supernatants from NDV-infected cells were intratumorally injected to mice bearing melanoma cells-derived tumors. Results: Oncolytic NDV induced CRT exposure, the release of HMGB1 and HSP70/90 as well as secretion of ATP in melanoma cells. Inhibition of apoptosis, autophagy, necroptosis or ER stress attenuated NDV/FMW-induced release of HMGB1 and HSP70/90. Moreover, NDV/FMW-induced ICD markers in melanoma cells were also suppressed by either treatment with a STAT3 inhibitor or shRNA-mediated depletion of STAT3. Of translational importance, treatment of mice bearing melanoma cells-derived tumors with supernatants from NDV/FMW-infected cells significantly inhibited tumor growth. Conclusions: Our data authenticate that oncolytic NDV/FMW might be a potent inducer of ICD in melanoma cells, which is amalgamated with several forms of cell death. We also show that STAT3 plays a role in NDV/FMW-induced ICD in melanoma cells. Together, our data highlight oncolytic NDV as propitious for cancer therapeutics by stimulatingan anti-melanoma immune response.
Collapse
Affiliation(s)
- Xiaoyan Shao
- Department of Medical Oncology, School of Medicine, Shanghai Tenths People's Hospital, Tongji University, Shanghai, China.,Department of Oncology, Dermatology Hospital, TongJi University, Shanghai, China.,Tongji University Cancer Center, Shanghai, China
| | - Xueke Wang
- Department of Medical Oncology, School of Medicine, Shanghai Tenths People's Hospital, Tongji University, Shanghai, China.,Department of Oncology, Dermatology Hospital, TongJi University, Shanghai, China.,Tongji University Cancer Center, Shanghai, China
| | - Xianling Guo
- Department of Medical Oncology, School of Medicine, Shanghai Tenths People's Hospital, Tongji University, Shanghai, China.,Department of Oncology, Dermatology Hospital, TongJi University, Shanghai, China.,Tongji University Cancer Center, Shanghai, China
| | - Ke Jiang
- Dalian Medical University Cancer Center, Institute of Cancer Stem Cell, Dalian, China
| | - Tian Ye
- Dalian Medical University Cancer Center, Institute of Cancer Stem Cell, Dalian, China
| | - Jianhua Chen
- Department of Medical Oncology, School of Medicine, Shanghai Tenths People's Hospital, Tongji University, Shanghai, China.,Department of Oncology, Dermatology Hospital, TongJi University, Shanghai, China.,Tongji University Cancer Center, Shanghai, China
| | - Juemin Fang
- Department of Medical Oncology, School of Medicine, Shanghai Tenths People's Hospital, Tongji University, Shanghai, China.,Department of Oncology, Dermatology Hospital, TongJi University, Shanghai, China.,Tongji University Cancer Center, Shanghai, China
| | - Linaer Gu
- Department of Medical Oncology, School of Medicine, Shanghai Tenths People's Hospital, Tongji University, Shanghai, China.,Department of Oncology, Dermatology Hospital, TongJi University, Shanghai, China.,Tongji University Cancer Center, Shanghai, China
| | - Sitong Wang
- Department of Medical Oncology, School of Medicine, Shanghai Tenths People's Hospital, Tongji University, Shanghai, China.,Department of Oncology, Dermatology Hospital, TongJi University, Shanghai, China.,Tongji University Cancer Center, Shanghai, China
| | - Guirong Zhang
- Central laboratory, Cancer School of Medicine, Liaoning Cancer Hospital and Institute, Hospital of China Medical University, Shenyang, China
| | - Songshu Meng
- Dalian Medical University Cancer Center, Institute of Cancer Stem Cell, Dalian, China
| | - Qing Xu
- Department of Medical Oncology, School of Medicine, Shanghai Tenths People's Hospital, Tongji University, Shanghai, China.,Department of Oncology, Dermatology Hospital, TongJi University, Shanghai, China.,Tongji University Cancer Center, Shanghai, China
| |
Collapse
|
11
|
Treatment of Metastatic Disease through Natural Killer Cell Modulation by Infected Cell Vaccines. Viruses 2019; 11:v11050434. [PMID: 31083491 PMCID: PMC6563237 DOI: 10.3390/v11050434] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 05/02/2019] [Accepted: 05/10/2019] [Indexed: 12/28/2022] Open
Abstract
Oncolytic viruses (OVs) are a form of immunotherapy that release tumor antigens in the context of highly immunogenic viral signals following tumor-targeted infection and destruction. Emerging preclinical and clinical evidence suggests that this in situ vaccine effect is critical for successful viro-immunotherapy. In this review, we discuss the application of OV as an infected cell vaccine (ICV) as one method of enhancing the potency and breadth of anti-tumoral immunity. We focus on understanding and manipulating the critical role of natural killer (NK) cells and their interactions with other immune cells to promote a clinical outcome. With a synergistic tumor killing and immune activating mechanism, ICVs represent a valuable new addition to the cancer fighting toolbox with the potential to treat malignant disease.
Collapse
|
12
|
Sivanandam V, LaRocca CJ, Chen NG, Fong Y, Warner SG. Oncolytic Viruses and Immune Checkpoint Inhibition: The Best of Both Worlds. MOLECULAR THERAPY-ONCOLYTICS 2019; 13:93-106. [PMID: 31080879 PMCID: PMC6503136 DOI: 10.1016/j.omto.2019.04.003] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Cancer immunotherapy and the emergence of immune checkpoint inhibitors have markedly changed the treatment paradigm for many cancers. They function to disrupt cancer cell evasion of the immune response and activate sustained anti-tumor immunity. Oncolytic viruses have also emerged as an additional therapeutic agent for cancer treatment. These viruses are designed to target and kill tumor cells while leaving the normal cells unharmed. As part of this process, oncolytic virus infection stimulates anti-cancer immune responses that augment the efficacy of checkpoint inhibition. These viruses have the capability of transforming a “cold” tumor microenvironment with few immune effector cells into a “hot” environment with increased immune cell and cytokine infiltration. For this reason, there are multiple ongoing clinical trials that combine oncolytic virotherapy and immune checkpoint inhibitors. This review will detail the key oncolytic viruses in preclinical and clinical studies and highlight the results of their testing with checkpoint inhibitors.
Collapse
Affiliation(s)
- Venkatesh Sivanandam
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA
| | | | - Nanhai G Chen
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Yuman Fong
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Susanne G Warner
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
13
|
Pol JG, Atherton MJ, Bridle BW, Stephenson KB, Le Boeuf F, Hummel JL, Martin CG, Pomoransky J, Breitbach CJ, Diallo JS, Stojdl DF, Bell JC, Wan Y, Lichty BD. Development and applications of oncolytic Maraba virus vaccines. Oncolytic Virother 2018; 7:117-128. [PMID: 30538968 PMCID: PMC6263248 DOI: 10.2147/ov.s154494] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Oncolytic activity of the MG1 strain of the Maraba vesiculovirus has proven efficacy in numerous preclinical cancer models, and relied not only on a direct cytotoxicity but also on the induction of both innate and adaptive antitumor immunity. To further expand tumor-specific T-cell effector and long-lasting memory compartments, we introduced the MG1 virus in a prime-boost cancer vaccine strategy. To this aim, a replication-incompetent adenoviral [Ad] vector together with the oncolytic MG1 have each been armed with a transgene expressing a same tumor antigen. Immune priming with the Ad vaccine subsequently boosted with the MG1 vaccine mounted tumor-specific responses of remarkable magnitude, which significantly prolonged survival in various murine cancer models. Based on these promising results, we validated the safety profile of the Ad:MG1 oncolytic vaccination strategy in nonhuman primates and initiated clinical investigations in cancer patients. Two clinical trials are currently under way (NCT02285816; NCT02879760). The present review will recapitulate the discoveries that led to the development of MG1 oncolytic vaccines from bench to bedside.
Collapse
Affiliation(s)
- Jonathan G Pol
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM), U1138, Paris, France
- Team 11 labelled Ligue Nationale contre le Cancer, Cordeliers Research Center, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Sorbonne Universités/Université Pierre et Marie Curie/Paris VI, Paris, France
| | - Matthew J Atherton
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada,
| | - Byram W Bridle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | | | - Fabrice Le Boeuf
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Jeff L Hummel
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada,
- Clinical Trial Division, CANSWERS, Georgetown, ON, Canada
| | | | | | | | - Jean-Simon Diallo
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - David F Stojdl
- Turnstone Biologics, Ottawa, ON, Canada,
- Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada
| | - John C Bell
- Turnstone Biologics, Ottawa, ON, Canada,
- Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Yonghong Wan
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada,
| | - Brian D Lichty
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada,
- Turnstone Biologics, Ottawa, ON, Canada,
| |
Collapse
|
14
|
Berkeley RA, Steele LP, Mulder AA, van den Wollenberg DJM, Kottke TJ, Thompson J, Coffey M, Hoeben RC, Vile RG, Melcher A, Ilett EJ. Antibody-Neutralized Reovirus Is Effective in Oncolytic Virotherapy. Cancer Immunol Res 2018; 6:1161-1173. [PMID: 30209061 DOI: 10.1158/2326-6066.cir-18-0309] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 08/14/2018] [Accepted: 08/16/2018] [Indexed: 11/16/2022]
Abstract
Immunotherapy is showing promise for otherwise incurable cancers. Oncolytic viruses (OVs), developed as direct cytotoxic agents, mediate their antitumor effects via activation of the immune system. However, OVs also stimulate antiviral immune responses, including the induction of OV-neutralizing antibodies. Current dogma suggests that the presence of preexisting antiviral neutralizing antibodies in patients, or their development during viral therapy, is a barrier to systemic OV delivery, rendering repeat systemic treatments ineffective. However, we have found that human monocytes loaded with preformed reovirus-antibody complexes, in which the reovirus is fully neutralized, deliver functional replicative reovirus to tumor cells, resulting in tumor cell infection and lysis. This delivery mechanism is mediated, at least in part, by antibody receptors (in particular FcγRIII) that mediate uptake and internalization of the reovirus/antibody complexes by the monocytes. This finding has implications for oncolytic virotherapy and for the design of clinical OV treatment strategies. Cancer Immunol Res; 6(10); 1161-73. ©2018 AACR.
Collapse
Affiliation(s)
- Robert A Berkeley
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Lynette P Steele
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom
| | - Aat A Mulder
- Leiden University Medical Centre, Department of Molecular Cell Biology, Leiden, the Netherlands
| | | | | | - Jill Thompson
- Department of Immunology, Mayo Clinic, Rochester, Minnesota
| | - Matthew Coffey
- Oncolytics Biotech Incorporated, Calgary, Alberta, Canada
| | - Rob C Hoeben
- Leiden University Medical Centre, Department of Molecular Cell Biology, Leiden, the Netherlands
| | - Richard G Vile
- Department of Immunology, Mayo Clinic, Rochester, Minnesota
| | - Alan Melcher
- Institute of Cancer Research, London, United Kingdom
| | - Elizabeth J Ilett
- Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, United Kingdom.
| |
Collapse
|
15
|
Melcher A. Oncolytic Virotherapy: Single Cycle Cures or Repeat Treatments? (Repeat Dosing Is Crucial!). Mol Ther 2018; 26:1875-1876. [PMID: 30017879 PMCID: PMC6094867 DOI: 10.1016/j.ymthe.2018.07.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Affiliation(s)
- Alan Melcher
- Institute of Cancer Research, Department of Radiotherapy and Imaging, London, UK.
| |
Collapse
|
16
|
Abstract
Hematological malignancies manifest as lymphoma, leukemia, and myeloma, and remain a burden on society. From initial therapy to endless relapse-related treatment, societal burden is felt not only in the context of healthcare cost, but also in the compromised quality of life of patients. Long-term therapeutic strategies have become the standard in keeping hematological malignancies at bay as these cancers develop resistance to each round of therapy with time. As a result, there is a continual need for the development of new drugs to combat resistant disease in order to prolong patient life, if not to produce a cure. This review aims to summarize advances in targeting lymphoma, leukemia, and myeloma through both cutting-edge and well established platforms. Current standard of treatment will be reviewed for these malignancies and emphasis will be made on new therapy development in the areas of antibody engineering, epigenetic small molecule inhibiting drugs, vaccine development, and chimeric antigen receptor cell engineering. In addition, platforms for the delivery of these and other drugs will be reviewed including antibody-drug conjugates, micro- and nanoparticles, and multimodal hydrogels. Lastly, we propose that tissue engineered constructs for hematological malignancies are the missing link in targeted drug discovery alongside mouse and patient-derived xenograft models.
Collapse
|
17
|
Falls T, Roy DG, Bell JC, Bourgeois-Daigneault MC. Murine Tumor Models for Oncolytic Rhabdo-Virotherapy. ILAR J 2017; 57:73-85. [PMID: 27034397 DOI: 10.1093/ilar/ilv048] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The preclinical optimization and validation of novel treatments for cancer therapy requires the use of laboratory animals. Although in vitro experiments using tumor cell lines and ex vivo treatment of patient tumor samples provide a remarkable first-line tool for the initial study of tumoricidal potential, tumor-bearing animals remain the primary option to study delivery, efficacy, and safety of therapies in the context of a complete tumor microenvironment and functional immune system. In this review, we will describe the use of murine tumor models for oncolytic virotherapy using vesicular stomatitis virus. We will discuss studies using immunocompetent and immunodeficient models with respect to toxicity and therapeutic treatments, as well as the various techniques and tools available to study cancer therapy with Rhabdoviruses.
Collapse
Affiliation(s)
- Theresa Falls
- Theresa Falls is a research technician at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada. Dominic Guy Roy is a Ph.D candidate at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a Ph.D candidate in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. John Cameron Bell is a senior researcher at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and professor in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. Marie-Claude Bourgeois-Daigneault is a postdoctoral fellow at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a postdoctoral fellow in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada
| | - Dominic Guy Roy
- Theresa Falls is a research technician at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada. Dominic Guy Roy is a Ph.D candidate at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a Ph.D candidate in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. John Cameron Bell is a senior researcher at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and professor in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. Marie-Claude Bourgeois-Daigneault is a postdoctoral fellow at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a postdoctoral fellow in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada
| | - John Cameron Bell
- Theresa Falls is a research technician at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada. Dominic Guy Roy is a Ph.D candidate at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a Ph.D candidate in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. John Cameron Bell is a senior researcher at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and professor in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. Marie-Claude Bourgeois-Daigneault is a postdoctoral fellow at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a postdoctoral fellow in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada
| | - Marie-Claude Bourgeois-Daigneault
- Theresa Falls is a research technician at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada. Dominic Guy Roy is a Ph.D candidate at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a Ph.D candidate in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. John Cameron Bell is a senior researcher at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and professor in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada. Marie-Claude Bourgeois-Daigneault is a postdoctoral fellow at the Centre for Innovative Cancer Research at Ottawa Hospital Research Institute in Ottawa, Canada, and a postdoctoral fellow in the Biochemistry, Microbiology, and Immunology Department at the University of Ottawa in Ottawa, Canada
| |
Collapse
|
18
|
Taking a Stab at Cancer; Oncolytic Virus-Mediated Anti-Cancer Vaccination Strategies. Biomedicines 2017; 5:biomedicines5010003. [PMID: 28536346 PMCID: PMC5423491 DOI: 10.3390/biomedicines5010003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 12/20/2016] [Accepted: 12/22/2016] [Indexed: 12/14/2022] Open
Abstract
Vaccines have classically been used for disease prevention. Modern clinical vaccines are continuously being developed for both traditional use as well as for new applications. Typically thought of in terms of infectious disease control, vaccination approaches can alternatively be adapted as a cancer therapy. Vaccines targeting cancer antigens can be used to induce anti-tumour immunity and have demonstrated therapeutic efficacy both pre-clinically and clinically. Various approaches now exist and further establish the tremendous potential and adaptability of anti-cancer vaccination. Classical strategies include ex vivo-loaded immune cells, RNA- or DNA-based vaccines and tumour cell lysates. Recent oncolytic virus development has resulted in a surge of novel viruses engineered to induce powerful tumour-specific immune responses. In addition to their use as cancer vaccines, oncolytic viruses have the added benefit of being directly cytolytic to cancer cells and thus promote antigen recognition within a highly immune-stimulating tumour microenvironment. While oncolytic viruses are perfectly equipped for efficient immunization, this complicates their use upon previous exposure. Indeed, the host's anti-viral counter-attacks often impair multiple-dosing regimens. In this review we will focus on the use of oncolytic viruses for anti-tumour vaccination. We will explore different strategies as well as ways to circumvent some of their limitations.
Collapse
|
19
|
Domingo-Musibay E, Yamamoto M. Gene and virotherapy for hematological malignancies. Int J Hematol 2016; 104:29-41. [PMID: 27289361 PMCID: PMC5089843 DOI: 10.1007/s12185-016-2031-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 05/22/2016] [Accepted: 05/24/2016] [Indexed: 12/12/2022]
Abstract
Recent years have seen a transformation in the treatment of hematological malignancies. Advances in gene therapy and molecular techniques and significant gains in computational abilities have supported the rapid development of safer and better tolerated therapies for many patients with hematologic cancers. In this review, we discuss novel applications of gene therapy, including immunomodulation and gene silencing, and report on the rise of oncolytic viruses for use in the treatment of malignancies arising in cells of the blood, lymph, and marrow. We discuss the relationship of the tropism of wildtype viruses and their oncolytic behavior as well as the tumoricidal and immunostimulatory properties of a number of attenuated and recombinant viruses currently in clinical development in countries around the world. While we have focused on promising virotherapy applications for future development, we also present a historical perspective and identify areas of potential clinical and regulatory practice change. We outline several of the virus systems being developed for applications in hematology, and summarize efficacy data in the context of ongoing or future human clinical testing. We also present the advantages and limitations of gene and virus therapy, including challenges and opportunities for improved treatment tolerability and outcomes for patients with hematologic malignancies.
Collapse
Affiliation(s)
- Evidio Domingo-Musibay
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Masato Yamamoto
- Division of Basic and Translational Research, Department of Surgery, University of Minnesota, MoosT 11-210, MMC195, 515 Delaware St SE, Minneapolis, MN, 55455, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
20
|
Li X, Liu X, Zhao Y, Zhong R, Song A, Sun L. Effect of thymosin α₁ on the phenotypic and functional maturation of dendritic cells from children with acute lymphoblastic leukemia. Mol Med Rep 2015; 12:6093-7. [PMID: 26239360 DOI: 10.3892/mmr.2015.4153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 06/15/2015] [Indexed: 11/06/2022] Open
Abstract
To determine the effect of thymosin α1 (Tα1) on the phenotypic and functional maturation of HL‑60 cells, freeze‑thaw antigen‑loaded dendritic cells (DCs) were derived from peripheral blood mononuclear cells (PBMCs) of children with acute lymphoblastic leukemia (ALL). The DCs were generated from the PBMC samples that were collected from the PB of 10 consecutive ALL children. On day 3 of culturing, the cells in the antigen + no Tα1 (AN) and antigen + Tα1 (AT) groups were incubated with 100 µl lysates obtained from freeze‑thaw cycling. After 5 days of incubation, the AT group was administered with 100 ng/ml Tα1. On day 8, the DCs were stained with fluorescein isothiocyanate‑conjugated cluster of differentiation (CD)1a, CD83 and HLA‑DR antibodies and analyzed by flow cytometry. In addition, the killing activity of cytotoxic T lymphocytes (CTLs) from the different groups on wild‑type leukemia cells was measured. The DCs in the AT group exhibited more apparent, characteristic dendritic morphologies than the control and AN group DCs. Furthermore, the lowest expression level of CD1a, and the highest expression of CD83 and HLA‑DR were observed in the AT group when compared with the AN and control groups (P<0.05). The lactate dehydrogenase release assay demonstrated that the killing rate of CTL in the AT group was significantly higher than that in the control and AN groups (P<0.01). Thus, Tα1 may markedly promote the phenotypic and functional maturation of DCs, and may serve as a suitable immunomodulator of DC‑based immunotherapy for treatment of hematological malignancies.
Collapse
Affiliation(s)
- Xuerong Li
- Department of Pediatric Hematology and Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Xiaodan Liu
- Department of Pediatric Hematology and Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Yanxia Zhao
- Department of Pediatric Hematology and Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Ren Zhong
- Department of Pediatric Hematology and Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Aiqin Song
- Department of Pediatric Hematology and Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Lirong Sun
- Department of Pediatric Hematology and Oncology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| |
Collapse
|
21
|
Arulanandam R, Batenchuk C, Varette O, Zakaria C, Garcia V, Forbes NE, Davis C, Krishnan R, Karmacharya R, Cox J, Sinha A, Babawy A, Waite K, Weinstein E, Falls T, Chen A, Hamill J, De Silva N, Conrad DP, Atkins H, Garson K, Ilkow C, Kærn M, Vanderhyden B, Sonenberg N, Alain T, Le Boeuf F, Bell JC, Diallo JS. Microtubule disruption synergizes with oncolytic virotherapy by inhibiting interferon translation and potentiating bystander killing. Nat Commun 2015; 6:6410. [PMID: 25817275 DOI: 10.1038/ncomms7410] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 01/27/2015] [Indexed: 12/11/2022] Open
Abstract
In this study, we show that several microtubule-destabilizing agents used for decades for treatment of cancer and other diseases also sensitize cancer cells to oncolytic rhabdoviruses and improve therapeutic outcomes in resistant murine cancer models. Drug-induced microtubule destabilization leads to superior viral spread in cancer cells by disrupting type I IFN mRNA translation, leading to decreased IFN protein expression and secretion. Furthermore, microtubule-destabilizing agents specifically promote cancer cell death following stimulation by a subset of infection-induced cytokines, thereby increasing viral bystander effects. This study reveals a previously unappreciated role for microtubule structures in the regulation of the innate cellular antiviral response and demonstrates that unexpected combinations of approved chemotherapeutics and biological agents can lead to improved therapeutic outcomes.
Collapse
Affiliation(s)
- Rozanne Arulanandam
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario, Canada K1H 8L6
| | - Cory Batenchuk
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario, Canada K1H 8L6
| | - Oliver Varette
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario, Canada K1H 8L6
| | - Chadi Zakaria
- Department of Biochemistry, Goodman Cancer Center, McGill University, 1160 Pine Avenue West, Montréal, Quebec, Canada H3A 1A3
| | - Vanessa Garcia
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario, Canada K1H 8L6
| | - Nicole E Forbes
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario, Canada K1H 8L6
| | - Colin Davis
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario, Canada K1H 8L6
| | - Ramya Krishnan
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario, Canada K1H 8L6
| | - Raunak Karmacharya
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario, Canada K1H 8L6
| | - Julie Cox
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario, Canada K1H 8L6
| | - Anisha Sinha
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario, Canada K1H 8L6
| | - Andrew Babawy
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario, Canada K1H 8L6
| | - Katherine Waite
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario, Canada K1H 8L6
| | - Erica Weinstein
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario, Canada K1H 8L6
| | - Theresa Falls
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario, Canada K1H 8L6
| | - Andrew Chen
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario, Canada K1H 8L6
| | - Jeff Hamill
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario, Canada K1H 8L6
| | - Naomi De Silva
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario, Canada K1H 8L6
| | - David P Conrad
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario, Canada K1H 8L6
| | - Harold Atkins
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario, Canada K1H 8L6
| | - Kenneth Garson
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario, Canada K1H 8L6
| | - Carolina Ilkow
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario, Canada K1H 8L6
| | - Mads Kærn
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, Ontario, Canada K1H 8M5
| | - Barbara Vanderhyden
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario, Canada K1H 8L6
| | - Nahum Sonenberg
- Department of Biochemistry, Goodman Cancer Center, McGill University, 1160 Pine Avenue West, Montréal, Quebec, Canada H3A 1A3
| | - Tommy Alain
- Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, Ontario, Canada K1H 8L1
| | - Fabrice Le Boeuf
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario, Canada K1H 8L6
| | - John C Bell
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario, Canada K1H 8L6
| | - Jean-Simon Diallo
- Center for Innovative Cancer Research, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, Ontario, Canada K1H 8L6
| |
Collapse
|
22
|
Tsang JJ, Atkins HL. The ex vivo purge of cancer cells using oncolytic viruses: recent advances and clinical implications. Oncolytic Virother 2015; 4:13-23. [PMID: 27512666 PMCID: PMC4918373 DOI: 10.2147/ov.s45525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Hematological malignancies are treated with intensive high-dose chemotherapy, with or without radiation. This is followed by hematopoietic stem cell (HSC) transplantation (HSCT) to rescue or reconstitute hematopoiesis damaged by the anticancer therapy. Autologous HSC grafts may contain cancer cells and purging could further improve treatment outcomes. Similarly, allogeneic HSCT may be improved by selectively purging alloreactive effector cells from the graft rather than wholesale immune cell depletion. Viral agents that selectively replicate in specific cell populations are being studied in experimental models of cancer and immunological diseases and have potential applications in the context of HSC graft engineering. This review describes preclinical studies involving oncolytic virus strains of adenovirus, herpes simplex virus type 1, myxoma virus, and reovirus as ex vivo purging agents for HSC grafts, as well as in vitro and in vivo experimental studies using oncolytic coxsackievirus, measles virus, parvovirus, vaccinia virus, and vesicular stomatitis virus to eradicate hematopoietic malignancies. Alternative ex vivo oncolytic virus strategies are also outlined that aim to reduce the risk of relapse following autologous HSCT and mitigate morbidity and mortality due to graft-versus-host disease in allogeneic HSCT.
Collapse
Affiliation(s)
- Jovian J Tsang
- Department of Biochemistry, University of Ottawa, ON, Canada; Cancer Therapeutics, Ottawa Hospital Research Institute, ON, Canada
| | - Harold L Atkins
- Cancer Therapeutics, Ottawa Hospital Research Institute, ON, Canada; Blood and Marrow Transplant Program, The Ottawa Hospital, Ottawa, ON, Canada
| |
Collapse
|
23
|
Cytokine conditioning enhances systemic delivery and therapy of an oncolytic virus. Mol Ther 2014; 22:1851-63. [PMID: 24957982 DOI: 10.1038/mt.2014.118] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Accepted: 06/18/2014] [Indexed: 12/11/2022] Open
Abstract
Optimum clinical protocols require systemic delivery of oncolytic viruses in the presence of an intact immune system. We show that preconditioning with immune modulators, or loading virus onto carrier cells ex vivo, enhances virus-mediated antitumor activity. Our early trials of systemic reovirus delivery showed that after infusion reovirus could be recovered from blood cells--but not from plasma--suggesting that rapid association with blood cells may protect virus from neutralizing antibody. We therefore postulated that stimulation of potential carrier cells directly in vivo before intravenous viral delivery would enhance delivery of cell-associated virus to tumor. We show that mobilization of the CD11b(+) cell compartment by granulocyte macrophage-colony stimulating factor immediately before intravenous reovirus, eliminated detectable tumor in mice with small B16 melanomas, and achieved highly significant therapy in mice bearing well-established tumors. Unexpectedly, cytokine conditioning therapy was most effective in the presence of preexisting neutralizing antibody. Consistent with this, reovirus bound by neutralizing antibody effectively accessed monocytes/macrophages and was handed off to tumor cells. Thus, preconditioning with cytokine stimulated recipient cells in vivo for enhanced viral delivery to tumors. Moreover, preexisting neutralizing antibody to an oncolytic virus may, therefore, even be exploited for systemic delivery to tumors in the clinic.
Collapse
|
24
|
Roy DG, Bell JC. Cell carriers for oncolytic viruses: current challenges and future directions. Oncolytic Virother 2013; 2:47-56. [PMID: 27512657 PMCID: PMC4918354 DOI: 10.2147/ov.s36623] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The optimal route for clinical delivery of oncolytic viruses is thought to be systemic intravenous injection; however, the immune system is armed with several highly efficient mechanisms to remove pathogens from the circulatory system. To overcome the challenges faced in trying to delivery oncolytic viruses specifically to tumors via the bloodstream, carrier cells have been investigated to determine their suitability as delivery vehicles for systemic administration of oncolytic viruses. Cell carriers protect viruses from neutralization, one of the most limiting aspects of oncolytic virus interaction with the immune system. Cell carriers can also possess inherent tumor tropism, thus directing the delivery of the virus more specifically to a tumor. With preclinical studies already demonstrating the success and feasibility of this approach with multiple oncolytic viruses, clinical evaluation of cell-mediated delivery of viruses is on the horizon. Meanwhile, ongoing preclinical studies are aimed at identifying new cellular vehicles for oncolytic viruses and improving current promising cell carrier platforms.
Collapse
Affiliation(s)
- Dominic G Roy
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada; Department of Biochemistry, Immunology and Microbiology, University of Ottawa, Ottawa, ON, Canada
| | - John C Bell
- Centre for Innovative Cancer Therapeutics, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada; Department of Biochemistry, Immunology and Microbiology, University of Ottawa, Ottawa, ON, Canada; Department of Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
25
|
Batenchuk C, Le Boeuf F, Stubbert L, Falls T, Atkins HL, Bell JC, Conrad DP. Non-replicating rhabdovirus-derived particles (NRRPs) eradicate acute leukemia by direct cytolysis and induction of antitumor immunity. Blood Cancer J 2013; 3:e123. [PMID: 23852158 PMCID: PMC3730201 DOI: 10.1038/bcj.2013.23] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 05/30/2013] [Accepted: 06/05/2013] [Indexed: 02/06/2023] Open
Abstract
Rhabdoviruses (RVs) are currently being pursued as anticancer therapeutics for various tumor types, notably leukemia. However, modest virion production and limited spread between noncontiguous circulating leukemic cells requires high-dose administration of RVs, which exceeds the maximum tolerable dose of the live virus. Furthermore, in severely immunosuppressed leukemic patients, the potential for uncontrolled live virus spread may compromise the safety of a live virus approach. We hypothesized that the barriers to oncolytic virotherapy in liquid tumors may be overcome by administration of high-dose non-replicating RVs. We have developed a method to produce unique high-titer bioactive yet non-replicating rhabdovirus-derived particles (NRRPs). This novel biopharmaceutical is multimodal possessing direct cytolytic and immunomodulatory activity against acute leukemia. We demonstrate that NRRP resistance in normal cells is mediated by intact antiviral defences including interferon (IFN). This data was substantiated using murine models of blast crisis. The translational promise of NRRPs was demonstrated in clinical samples obtained from patients with high-burden multidrug-resistant acute myeloid leukemia. This is the first successful attempt to eradicate disseminated cancer using a non-replicating virus-derived agent, representing a paradigm shift in our understanding of oncolytic virus-based therapies and their application toward the treatment of acute leukemia.
Collapse
Affiliation(s)
- C Batenchuk
- 1] Center for Cancer Therapeutics, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada [2] Department of Biochemistry, Immunology and Microbiology, University of Ottawa, Ottawa, Ontario, Canada [3] Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|