Effective Combination Immunotherapy with Oncolytic Adenovirus and Anti-PD-1 for Treatment of Human and Murine Ovarian Cancers

Simple Summary This study was conducted to find a new, more efficient, treatment for ovarian cancer. A combination of an oncolytic adenovirus (TILT-123) with immune checkpoint inhibitors was employed to treat ex vivo patient samples and was found statistically significantly more effective than control treatments ex vivo and showed potent efficacy towards in vivo tumor growth. Abstract Ovarian cancer (OvCa) is one of the most common gynecological cancers and has the highest mortality in this category. Tumors are often detected late, and unfortunately over 70% of OvCa patients experience relapse after first-line treatments. OvCa has shown low response rates to immune checkpoint inhibitor (ICI) treatments, thus leaving room for improvement. We have shown that oncolytic adenoviral therapy with Ad5/3-E2F-d24-hTNFa-IRES-hIL2 (aka. TILT-123) is promising for single-agent treatment of cancer, but also for sensitizing tumors for T-cell dependent immunotherapy approaches, such as ICI treatments. Therefore, this study set out to determine the effect of inhibition of the immune checkpoint inhibitors (ICI), in the context of TILT-123 therapy of OvCa. We show that simultaneous treatment of patient derived samples with TILT-123 and ICIs anti-PD-1 or anti-PD-L1 efficiently reduced overall viability. The combinations induced T cell activation, T cells expressed activation markers more often, and the treatment caused positive microenvironment changes, measured by flow cytometric assays. Furthermore, in an immunocompetent in vivo C57BL/6NHsda mouse model, tumor growth was hindered, when treated with TILT-123, ICI or both. Taken together, this study provides a rationale for using TILT-123 virotherapy in combination with TILT-123 and immune checkpoint inhibitors together in an ovarian cancer OvCa clinical trial.

[1]  A. Hemminki,et al.  Local therapy with an engineered oncolytic adenovirus enables antitumor response in non-injected melanoma tumors in mice treated with aPD-1 , 2022, Oncoimmunology.

[2]  M. Fuentes,et al.  Autoimmune Responses in Oncology: Causes and Significance , 2021, International journal of molecular sciences.

[3]  Wei Zhang,et al.  Influence of Tumor Immune Infiltration on Immune Checkpoint Inhibitor Therapeutic Efficacy: A Computational Retrospective Study , 2021, Frontiers in Immunology.

[4]  C. Czerlanis,et al.  Immune Checkpoint Blockade in Gynecologic Cancers: State of Affairs , 2020, Cancers.

[5]  A. Hemminki,et al.  Oncolytic viruses for cancer immunotherapy , 2020, Journal of Hematology & Oncology.

[6]  A. Hemminki,et al.  TNFa and IL2 Encoding Oncolytic Adenovirus Activates Pathogen and Danger-Associated Immunological Signaling , 2020, Cells.

[7]  T. D. de Gruijl,et al.  Oncolytic adenovirus shapes the ovarian tumor microenvironment for potent tumor-infiltrating lymphocyte tumor reactivity , 2020, Journal for ImmunoTherapy of Cancer.

[8]  A. Hemminki,et al.  Tumor microenvironment remodeling by an engineered oncolytic adenovirus results in improved outcome from PD-L1 inhibition , 2020, Oncoimmunology.

[9]  L. Sánchez-Lorenzo,et al.  Immunotherapy with checkpoint inhibitors in patients with ovarian cancer: Still promising? , 2019, Cancer.

[10]  P. Ascierto,et al.  Adjuvant ipilimumab versus placebo after complete resection of stage III melanoma: long-term follow-up results of the European Organisation for Research and Treatment of Cancer 18071 double-blind phase 3 randomised trial. , 2019, European journal of cancer.

[11]  J. Larkin,et al.  Pembrolizumab versus ipilimumab in advanced melanoma (KEYNOTE-006): post-hoc 5-year results from an open-label, multicentre, randomised, controlled, phase 3 study. , 2019, The Lancet. Oncology.

[12]  T. Randall,et al.  Overcoming immune suppression with epigenetic modification in ovarian cancer. , 2019, Translational research : the journal of laboratory and clinical medicine.

[13]  S. Berceli,et al.  Expression of a Functional IL-2 Receptor in Vascular Smooth Muscle Cells , 2018, The Journal of Immunology.

[14]  A. Hemminki,et al.  Abscopal Effect in Non-injected Tumors Achieved with Cytokine-Armed Oncolytic Adenovirus , 2018, Molecular therapy oncolytics.

[15]  Y. Kodera,et al.  The Current Status and Future Prospects of Oncolytic Viruses in Clinical Trials against Melanoma, Glioma, Pancreatic, and Breast Cancers , 2018, Cancers.

[16]  E. Carapuça,et al.  Oncolytic virus immunotherapies in ovarian cancer: moving beyond adenoviruses , 2018, Porto biomedical journal.

[17]  N. Lemoine,et al.  Oncolytic Viral Therapy and the Immune System: A Double-Edged Sword Against Cancer , 2018, Front. Immunol..

[18]  R. Weinberg,et al.  Understanding the tumor immune microenvironment (TIME) for effective therapy , 2018, Nature Medicine.

[19]  K. Savage,et al.  Nivolumab for Relapsed/Refractory Classic Hodgkin Lymphoma After Failure of Autologous Hematopoietic Cell Transplantation: Extended Follow-Up of the Multicohort Single-Arm Phase II CheckMate 205 Trial , 2018, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[20]  L. Sequist,et al.  Primary Patient-Derived Cancer Cells and Their Potential for Personalized Cancer Patient Care , 2017, Cell reports.

[21]  F. Lang,et al.  Oncolytic Adenovirus and Tumor-Targeting Immune Modulatory Therapy Improve Autologous Cancer Vaccination. , 2017, Cancer research.

[22]  D. Nettelbeck,et al.  Oncolytic Adenoviruses Armed with Tumor Necrosis Factor Alpha and Interleukin-2 Enable Successful Adoptive Cell Therapy , 2016, Molecular therapy oncolytics.

[23]  J. Hawiger,et al.  Interleukin 2 Activates Brain Microvascular Endothelial Cells Resulting in Destabilization of Adherens Junctions* , 2016, The Journal of Biological Chemistry.

[24]  M. Kiechle,et al.  CXCL9 and CXCL10 predict survival and are regulated by cyclooxygenase inhibition in advanced serous ovarian cancer , 2016, British Journal of Cancer.

[25]  A. Hemminki,et al.  Adenoviral Delivery of Tumor Necrosis Factor-α and Interleukin-2 Enables Successful Adoptive Cell Therapy of Immunosuppressive Melanoma. , 2016, Molecular therapy : the journal of the American Society of Gene Therapy.

[26]  J. Mathis,et al.  Syngeneic Syrian hamster tumors feature tumor-infiltrating lymphocytes allowing adoptive cell therapy enhanced by oncolytic adenovirus in a replication permissive setting , 2016, Oncoimmunology.

[27]  A. Hemminki,et al.  Favorable Alteration of Tumor Microenvironment by Immunomodulatory Cytokines for Efficient T-Cell Therapy in Solid Tumors , 2015, PloS one.

[28]  David T. W. Jones,et al.  Signatures of mutational processes in human cancer , 2013, Nature.

[29]  A. Dudley,et al.  A three-party alliance in solid tumors , 2013, Adipocyte.

[30]  A. Goy,et al.  Early Detection Biomarkers for Ovarian Cancer , 2012, Journal of oncology.

[31]  Benjamin J. Raphael,et al.  Integrated Genomic Analyses of Ovarian Carcinoma , 2011, Nature.

[32]  R. Tothill,et al.  Novel Molecular Subtypes of Serous and Endometrioid Ovarian Cancer Linked to Clinical Outcome , 2008, Clinical Cancer Research.

[33]  Gerd Ritter,et al.  Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[34]  B. Aggarwal,et al.  TNF-Induced Signaling in Apoptosis , 1999, Journal of Clinical Immunology.

[35]  A. Oberg,et al.  Oncolytic measles virus expressing the sodium iodide symporter to treat drug-resistant ovarian cancer. , 2015, Cancer research.

[36]  E. Aktas,et al.  Relationship between CD107a expression and cytotoxic activity. , 2009, Cellular immunology.