Defining Effective Combinations of Immune Checkpoint Blockade and Oncolytic Virotherapy

Purpose: Recent data from randomized clinical trials with oncolytic viral therapies and with cancer immunotherapies have finally recapitulated the promise these platforms demonstrated in preclinical models. Perhaps the greatest advance with oncolytic virotherapy has been the appreciation of the importance of activation of the immune response in therapeutic activity. Meanwhile, the understanding that blockade of immune checkpoints (with antibodies that block the binding of PD1 to PDL1 or CTLA4 to B7-2) is critical for an effective antitumor immune response has revitalized the field of immunotherapy. The combination of immune activation using an oncolytic virus and blockade of immune checkpoints is therefore a logical next step. Experimental Design: Here, we explore such combinations and demonstrate their potential to produce enhanced responses in mouse tumor models. Different combinations and regimens were explored in immunocompetent mouse models of renal and colorectal cancer. Bioluminescence imaging and immune assays were used to determine the mechanisms mediating synergistic or antagonistic combinations. Results: Interaction between immune checkpoint inhibitors and oncolytic virotherapy was found to be complex, with correct selection of viral strain, antibody, and timing of the combination being critical for synergistic effects. Indeed, some combinations produced antagonistic effects and loss of therapeutic activity. A period of oncolytic viral replication and directed targeting of the immune response against the tumor were required for the most beneficial effects, with CD8+ and NK, but not CD4+ cells mediating the effects. Conclusions: These considerations will be critical in the design of the inevitable clinical translation of these combination approaches. Clin Cancer Res; 21(24); 5543–51. ©2015 AACR. See related commentary by Slaney and Darcy, p. 5417

[1]  S. Thorne Immunotherapeutic Potential of Oncolytic Vaccinia Virus , 2014, Front. Oncol..

[2]  J. Wolchok,et al.  Localized Oncolytic Virotherapy Overcomes Systemic Tumor Resistance to Immune Checkpoint Blockade Immunotherapy , 2014, Science Translational Medicine.

[3]  T. Hermiston,et al.  Armed Therapeutic Viruses – A Disruptive Therapy on the Horizon of Cancer Immunotherapy , 2014, Front. Immunol..

[4]  N. Senzer,et al.  OPTiM: A randomized phase III trial of talimogene laherparepvec (T-VEC) versus subcutaneous (SC) granulocyte-macrophage colony-stimulating factor (GM-CSF) for the treatment (tx) of unresected stage IIIB/C and IV melanoma. , 2013, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[5]  D. Bartlett,et al.  Crosstalk between immune cell and oncolytic vaccinia therapy enhances tumor trafficking and antitumor effects. , 2013, Molecular therapy : the journal of the American Society of Gene Therapy.

[6]  M. Bloomston,et al.  Randomized dose-finding clinical trial of oncolytic immunotherapeutic vaccinia JX-594 in liver cancer , 2013, Nature Medicine.

[7]  D. Pardoll Immunology beats cancer: a blueprint for successful translation , 2012, Nature Immunology.

[8]  R. Vile,et al.  Systemic combination virotherapy for melanoma with tumor antigen-expressing vesicular stomatitis virus and adoptive T-cell transfer. , 2012, Cancer research.

[9]  C. Drake,et al.  Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. , 2012, The New England journal of medicine.

[10]  David C. Smith,et al.  Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. , 2012, The New England journal of medicine.

[11]  J. Turchi,et al.  Abstract 4753: Targeting nucleotide excision repair: Chemical synthetic lethality and biology-based combination therapy , 2012 .

[12]  G. Cheng,et al.  Treating tumors with a vaccinia virus expressing IFNβ illustrates the complex relationships between oncolytic ability and immunogenicity. , 2012, Molecular therapy : the journal of the American Society of Gene Therapy.

[13]  Drew M. Pardoll,et al.  The blockade of immune checkpoints in cancer immunotherapy , 2012, Nature Reviews Cancer.

[14]  A. Hemminki,et al.  Targeted cancer immunotherapy with oncolytic adenovirus coding for a fully human monoclonal antibody specific for CTLA-4 , 2011, Gene Therapy.

[15]  J. Wolchok,et al.  Novel cancer immunotherapy agents with survival benefit: recent successes and next steps , 2011, Nature Reviews Cancer.

[16]  Z. Guo,et al.  Chemokine expression from oncolytic vaccinia virus enhances vaccine therapies of cancer. , 2011, Molecular therapy : the journal of the American Society of Gene Therapy.

[17]  C. Contag,et al.  Targeting localized immune suppression within the tumor through repeat cycles of immune cell-oncolytic virus combination therapy. , 2010, Molecular therapy : the journal of the American Society of Gene Therapy.

[18]  S. Kim-Schulze,et al.  Local and Distant Immunity Induced by Intralesional Vaccination with an Oncolytic Herpes Virus Encoding GM-CSF in Patients with Stage IIIc and IV Melanoma , 2010, Annals of Surgical Oncology.

[19]  S. Chatterjee,et al.  Combination of CTL‐associated Antigen‐4 Blockade and Depletion of CD25+ Regulatory T Cells Enhance Tumour Immunity of Dendritic Cell‐based Vaccine in a Mouse Model of Colon Cancer , 2010, Scandinavian journal of immunology.

[20]  F. Errington,et al.  Immune-Mediated Antitumor Activity of Reovirus Is Required for Therapy and Is Independent of Direct Viral Oncolysis and Replication , 2009, Clinical Cancer Research.

[21]  C. Contag,et al.  Chemical control of protein stability and function in living mice , 2008, Nature Medicine.

[22]  C. Contag,et al.  Enhancing poxvirus oncolytic effects through increased spread and immune evasion. , 2008, Cancer research.

[23]  D. Kirn,et al.  Targeting of Interferon-Beta to Produce a Specific, Multi-Mechanistic Oncolytic Vaccinia Virus , 2007, PLoS medicine.

[24]  D. Kirn,et al.  Rational strain selection and engineering creates a broad-spectrum, systemically effective oncolytic poxvirus, JX-963. , 2007, The Journal of clinical investigation.

[25]  F. Marincola,et al.  Eradication of solid human breast tumors in nude mice with an intravenously injected light-emitting oncolytic vaccinia virus. , 2007, Cancer research.

[26]  Drew M. Pardoll,et al.  Immunostimulatory monoclonal antibodies for cancer therapy , 2007, Nature Reviews Cancer.

[27]  Jiwon Kim,et al.  Systemic armed oncolytic and immunologic therapy for cancer with JX-594, a targeted poxvirus expressing GM-CSF. , 2006, Molecular therapy : the journal of the American Society of Gene Therapy.

[28]  S. Quezada,et al.  CTLA4 blockade and GM-CSF combination immunotherapy alters the intratumor balance of effector and regulatory T cells. , 2006, The Journal of clinical investigation.

[29]  Andrew Pekosz,et al.  Bioluminescence imaging of vaccinia virus: effects of interferon on viral replication and spread. , 2005, Virology.

[30]  S. Libutti,et al.  Systemic cancer therapy with a tumor-selective vaccinia virus mutant lacking thymidine kinase and vaccinia growth factor genes. , 2001, Cancer research.

[31]  J. Allison,et al.  Combination Immunotherapy of B16 Melanoma Using Anti–Cytotoxic T Lymphocyte–Associated Antigen 4 (Ctla-4) and Granulocyte/Macrophage Colony-Stimulating Factor (Gm-Csf)-Producing Vaccines Induces Rejection of Subcutaneous and Metastatic Tumors Accompanied by Autoimmune Depigmentation , 1999, The Journal of experimental medicine.

[32]  B. Moss,et al.  Compact, synthetic, vaccinia virus early/late promoter for protein expression. , 1997, BioTechniques.

[33]  J. Allison,et al.  Enhancement of Antitumor Immunity by CTLA-4 Blockade , 1996, Science.

[34]  R. Buller,et al.  Vaccinia Virus B18R Gene Encodes a Type I Interferon-binding Protein That Blocks Interferon α Transmembrane Signaling (*) , 1995, The Journal of Biological Chemistry.

[35]  S. Thorne,et al.  Regulating cytokine function enhances safety and activity of genetic cancer therapies. , 2013, Molecular therapy : the journal of the American Society of Gene Therapy.