Novel Genetic Melanoma Vaccines Based on Induced Pluripotent Stem Cells or Melanosphere-Derived Stem-Like Cells Display High Efficacy in a murine Tumor Rejection Model

Therapeutic cancer vaccines have elicited renewed interest due to the development of immune checkpoint inhibitors. The role of these vaccines is to induce specific effector cells for killing cancer cells. Cancer stem cells (CSCs) are responsible for tumor growth and progression. Accordingly, they are targets for various cancer therapies, including immunotherapy. Here, we demonstrate the effectiveness of melanoma vaccines composed of genetically modified tumor cells admixed with melanoma stem-like cells (MSC) or induced pluripotent stem cells (iPSCs). Two vaccines were constructed. The first vaccine contained cells derived from B16F10 melanospheres (SFs) with CSC characteristics. The second vaccine contained syngeneic murine induced pluripotent stem cells (miPSCs). iPSCs or SF cells were admixed with B16F10 cells, modified with the designer cytokine Hyper-IL6(H6) (B16/H6). Control mice received B16/H6 cells, B16F10 cells or PBS. Immunization with either vaccine significantly inhibited tumor growth and increased disease-free survival (DFS) and overall survival (OS) in C57BL/6 mice. Mice treated with the SF or iPSC vaccine demonstrated increased activation of the immune response in the vaccination site and tumor microenvironment compared to those treated with B16/H6, B16F10 or PBS. Higher infiltration of dendritic cells (DCs) monocytes, and natural killer (NK) cells; lower numbers of myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs); higher levels of the cytokines INFγ and IL-12 were observed with the novel vaccines than with the control treatments. In vitro restimulation of splenocytes derived from mice immunized with B16F10 cell, SF cell or miPSC lysates increased the proliferation of CD4+ T helper lymphocytes and secretion of cytokines. An increased serum titer of antibodies directed against B16F10 cells was found in mice immunized with the SF vaccine. The most effective DFS and OS extensions were reached with the miPSCs vaccine. The described results form the basis for a novel platform for the next generation of cancer vaccines composed of allogeneic cancer-specific cells modified with a molecular adjuvant gene and admixed with allogeneic miPSCs or SFs.

[1]  Sakshi Sahni,et al.  Role of Anti-PD-1 Antibodies in Advanced Melanoma: The Era of Immunotherapy , 2018, Cureus.

[2]  A. Mackiewicz,et al.  Re-induction using whole cell melanoma vaccine genetically modified to melanoma stem cells-like beyond recurrence extends long term survival of high risk resected patients - updated results , 2018, Journal of Immunotherapy for Cancer.

[3]  P. Dong,et al.  Tumor-Intrinsic PD-L1 Signaling in Cancer Initiation, Development and Treatment: Beyond Immune Evasion , 2018, Front. Oncol..

[4]  Patrycja Czerwińska,et al.  Whole cell melanoma vaccine genetically modified to stem cells like phenotype generates specific immune responses to ALDH1A1 and long-term survival in advanced melanoma patients , 2018, Oncoimmunology.

[5]  Mark M. Davis,et al.  Autologous iPSC-Based Vaccines Elicit Anti-tumor Responses In Vivo. , 2018, Cell stem cell.

[6]  K. Kabashima,et al.  Anti-PD-1 and Anti-CTLA-4 Therapies in Cancer: Mechanisms of Action, Efficacy, and Limitations , 2018, Front. Oncol..

[7]  K. Khanna,et al.  Combination Immunotherapy: Taking Cancer Vaccines to the Next Level , 2018, Front. Immunol..

[8]  J. Dou,et al.  Effective tumor immunity to melanoma mediated by B16F10 cancer stem cell vaccine , 2017, International Immunopharmacology.

[9]  A. Mackiewicz,et al.  Programmed cell death 1 checkpoint inhibitors in the treatment of patients with advanced melanoma , 2017, Contemporary oncology.

[10]  K. Kaur,et al.  Differentiation by NK cells is a prerequisite for effective targeting of cancer stem cells/poorly differentiated tumors by chemopreventive and chemotherapeutic drugs , 2017, Journal of Cancer.

[11]  K. Kaur,et al.  Novel strategies to target cancer stem cells by NK cells; studies in humanized mice. , 2017, Frontiers in bioscience.

[12]  C. Aspord,et al.  Melanoma dormancy in a mouse model is linked to GILZ/FOXO3A-dependent quiescence of disseminated stem-like cells , 2016, Scientific Reports.

[13]  Marzieh Ebrahimi,et al.  Dendritic cell based immunotherapy using tumor stem cells mediates potent antitumor immune responses. , 2016, Cancer letters.

[14]  K. Kaur,et al.  Adoptive transfer of osteoclast-expanded natural killer cells for immunotherapy targeting cancer stem-like cells in humanized mice , 2016, Cancer Immunology, Immunotherapy.

[15]  Kaori Tanaka,et al.  Aldehyde dehydrogenase 1A1 in stem cells and cancer , 2016, Oncotarget.

[16]  Yujiang Fang,et al.  The paradoxical role of IL-10 in immunity and cancer. , 2015, Cancer letters.

[17]  M. Serrano,et al.  The pluripotency factor NANOG promotes the formation of squamous cell carcinomas , 2015, Scientific Reports.

[18]  R. Stefan,et al.  Whole Cell Therapeutic Vaccine Modified With Hyper-IL6 for Combinational Treatment of Nonresected Advanced Melanoma , 2015, Medicine.

[19]  A. Mackiewicz,et al.  Immunotargeting of cancer stem cells , 2015, Contemporary oncology.

[20]  C. Creighton,et al.  VEGF drives cancer-initiating stem cells through VEGFR-2/Stat3 signaling to upregulate Myc and Sox2 , 2014, Oncogene.

[21]  N. Popitsch,et al.  CTLA-4 and PD-1/PD-L1 Blockade: New Immunotherapeutic Modalities with Durable Clinical Benefit in Melanoma Patients , 2013, Clinical Cancer Research.

[22]  A. Mackiewicz,et al.  Therapeutic gene modified cell based cancer vaccines. , 2013, Gene.

[23]  Baocun Sun,et al.  The in-vitro spheroid culture induces a more highly differentiated but tumorigenic population from melanoma cell lines , 2013, Melanoma research.

[24]  I. Mellman,et al.  Oncology meets immunology: the cancer-immunity cycle. , 2013, Immunity.

[25]  Masashi Kato,et al.  Inflammatory monocytes are potent antitumor effectors controlled by regulatory CD4+ T cells , 2013, Proceedings of the National Academy of Sciences.

[26]  M. Wiznerowicz,et al.  Long-term survival of high-risk melanoma patients immunized with a Hyper-IL-6-modified allogeneic whole-cell vaccine after complete resection , 2012, Expert opinion on investigational drugs.

[27]  A. Mackiewicz,et al.  Design of clinical trials for therapeutic cancer vaccines development. , 2009, European journal of pharmacology.

[28]  Eric Vivier,et al.  Functions of natural killer cells , 2008, Nature Immunology.

[29]  J. Scheller,et al.  The IL-6/sIL-6R complex as a novel target for therapeutic approaches , 2007, Expert opinion on therapeutic targets.

[30]  Ugo Orfanelli,et al.  Isolation and Characterization of Tumorigenic, Stem-like Neural Precursors from Human Glioblastoma , 2004, Cancer Research.

[31]  I. Weissman,et al.  Stem cells, cancer, and cancer stem cells , 2001, Nature.

[32]  H. Ertl,et al.  Vaccine regimen for prevention of sexually transmitted infections with human papillomavirus type 16. , 2001, Vaccine.

[33]  P. Schirmacher,et al.  The designer cytokine hyper-IL-6 mediates growth inhibition and GM–CSF-dependent rejection of B16 melanoma cells , 2001, Oncogene.

[34]  S. Rose-John,et al.  A bioactive designer cytokine for human hematopoietic progenitor cell expansion , 1997, Nature Biotechnology.