Weekly Digests
‹ Back to March

Cancer immunity — can it stem from stem cells?

March 14, 2018

Induced pluripotent stem cells (iPSCs), lineage-specific adult cells that are reprogrammed to an earlier pluripotent state, bypass the ethical controversy associated with embryonic stem cell use. Because iPSCs and cancer cells overlap in the expression of many tumor-associated genes, iPSCs can prime the immune system for both known and potentially unknown tumor-associated antigens (TAAs) and tumor-specific antigens (TSAs). With this rationale in mind, Kooreman et al. explored the use of iPSCs as a generalizable vaccine in multiple models of cancer in their paper published in Cell Stem Cell.

Using the Toll-like receptor ligand CpG (“C”), previously shown to be an effective adjuvant in association with a whole-cell vaccine, and irradiated iPSCs from murine FVB cells (“I”), the team began their work by combining CpG with FVB strain iPSCs (C+I). After optimizing the immune response with once-weekly injections of C+I for four weeks, 40 FVB mice were vaccinated with either PBS, C, I, or C+I. After four weeks of vaccinations, all mice were subcutaneously injected with DB7 breast cancer cells. All mice initially developed tumors at one week, but tumors regressed in 7 of 10 mice in the C+I group. All other groups had progressive tumor growth.

Four weeks after DB7 tumor cell inoculation, five mice from each group were sacrificed for further study of the spleen, blood, and draining lymph nodes (dLNs); the remaining five mice from each group were maintained for long-term survival studies. Mice treated with C+I had significant increases in effector and memory cytotoxic CD8+ T cell populations in both the spleen and dLNs. Splenocytes showed an increased release of IFNγ in response to DB7 tumor lysate, demonstrating tumor specificity. Mature antigen-presenting cells (APCs) and helper T cell populations were also increased in the dLNs of C+I-treated mice. Two of the five C+I-treated FVB mice survived for one year; at that time, both mice had antibody titers against iPSCs and DB7, similar to those seen before the extended incubation period, and were able to completely reject reintroduced cancer cells.

To analyze the effect of the iPSC vaccine in other models of cancer, the same experiments were repeated in B16F0 melanoma mice with C57BL/6 iPSCs. At two weeks, tumor growth was significantly lower in the C+I mice. Flow cytometry demonstrated a significant decrease in CD4+CD25+Foxp3+ Tregs in the blood, an increase in mature APCs, and an increase in effector and memory Th cells in dLNs. In a third variation of the same experiment, performed in the AC29 mesothelioma murine model, tumor regression was successfully predicted by B and T cells expressing IL-2, IL-4, and IL-5 in C+I-vaccinated mice. Importantly, these mice had significantly lower systemic cytokine levels than PBS controls, pointing to a localized immune response and a potentially low risk of organ toxicity.

To demonstrate that the immune responses were due to antigens shared by both the cancer cell line and iPSCs – “two-way immunity” – the researchers turned to adoptive T cell transfer. T cells were prepared from C+I-vaccinated and vehicle-vaccinated mice and transferred into orthotopic DB7 tumor-bearing mice. Cancer immunity was demonstrated by a significant reduction in tumor size in the mice receiving the adoptive transfer of C+I isolated T cells compared with controls. Separately, tumor-experienced lymphocytes (TELs) were extracted from dLNs near the DB7 tumor site of C+I-vaccinated mice and vehicle-vaccinated mice and adoptively transferred into NOD-SCID mice inoculated with iPSC cells. NOD-SCID mice that received TELs from C+I-treated mice exhibited significantly reduced teratoma size, demonstrating iPSC immunity.

For their final experiment, Kooreman et al. investigated the effectiveness of the C+I vaccine as an adjuvant after tumor resection. B16F0 cells were injected into the lower backs of C57BL/6 mice and the tumors were R1-resected (microscopic residual tumor) or R2-resected (macroscopic residual tumor) two weeks later. With two adjuvant rounds of C+I, R2-resected mice had no visible tumors within their resection area, while vehicle-vaccinated mice did. After four weeks of vaccination with either C+I or CpG alone, R1-resected mice showed reduced tumor load in dLNs. Interestingly, only C+I generated significantly lower tumor recurrence in areas farther from the vaccination site, indicating the systemic effect of C+I vaccination.

Results of the experiments performed by Kooreman et al. demonstrate the effectiveness of iPSCs as a whole-cell prophylactic vaccine targeting known and unknown tumor-associated antigens in multiple models of cancer. While the results can’t be directly translated from mouse models to human studies, iPSCs hold promise, particularly as a rapidly available adjuvant therapy following primary tumor reduction.

by Brynn Vessey

References:

Kooreman N.G., Kim Y., de Almeida P.E., Termglinchan V., Diecke S., Shao N.Y., Wei T.T., Yi H., Dey D., Nelakanti R., Brouwer T.P., Paik D.T., Sagiv-Barfi I., Han A., Quax P.H.A., Hamming J.F., Levy R., Davis M.M., Wu J.C. Autologous iPSC-Based Vaccines Elicit Anti-tumor Responses In Vivo. Cell Stem Cell. 2018 Feb 8.

In the Spotlight...

Functional Anti-TIGIT Antibodies Regulate Development of Autoimmunity and Antitumor Immunity

Dixon et al. generated agonistic and blocking anti-mouse antibodies (Abs) against the coinhibitory receptor TIGIT that modulated T cell response. In mice, agonistic anti-TIGIT Abs reduced the severity of autoimmune disease. Antagonistic anti-TIGIT Ab combined with anti-PD-1 improved tumor control in mice with colon carcinoma or glioblastoma by enhancing T cell proliferation, antitumor cytokine production, and long-term immunity.

Cancer vaccine formulation dictates synergy with CTLA-4 and PD-L1 checkpoint blockade therapy

Hailemichael et al. demonstrate that addition of gp100 peptide vaccine formulated in incomplete Freund’s adjuvant, but not other nonpersistent vaccine formulations, to GVAX/anti-CTLA-4 surprisingly sequestered both gp100-specific effector T cells (Teffs) and other tumor-specific Teffs at the vaccination site, diminishing tumor control. The persistent, localized antigen initially attracted gp100 Teffs that in turn recruited myeloid cells and Teffs of other specificities to the vaccination site via IFNγ, chemokine signaling, and upregulation of adhesion molecules.

Hypomethylating agents in combination with immune checkpoint inhibitors in acute myeloid leukemia and myelodysplastic syndromes

Daver et al. outline the benefit of coupling immune-stimulating hypomethylating agents (HMAs), (including upregulation of IFNγ pathway genes and costimulatory molecules in immune cells, and increased expression of cancer testis antigens and HLA class I in tumor cells) with checkpoint therapy to counter the HMA-induced increase in cellular exhaustion markers PD-1 and/or CTLA-4. Early trials of anti-PD-1 therapy in combination with HMA treatment show promising results in acute myeloid leukemia and myelodysplastic syndrome.

Histone deacetylase (HDAC) inhibitor ACY241 enhances anti-tumor activities of antigen-specific central memory cytotoxic T lymphocytes against multiple myeloma and solid tumors

Bae et al. elucidate the immune cell-modulating activities of the histone deacetylase 6 inhibitor, ACY241, in bone marrow cell cultures from multiple myeloma (MM) patients. They show that ACY241 decreased levels of MM cells, MDSCs, and Tregs, reduced PD-L1 expression by tumor and Tregs, and increased HLA and CD80/86 expression in tumor and DCs. Peptide-stimulated CD8+ T cells treated with ACY241 upregulated co-stimulatory and functional markers, downregulated PD-1, and were driven to a memory phenotype via stimulation of the AKT/mTOR pathway.

Identification of alpha-fetoprotein-specific T cell receptors for hepatocellular carcinoma immunotherapy

Zhu et al. used a lentivector and peptide prime/boost immunization in human HLA-A2 transgenic mice to identify CD8+ T cells and corresponding TCR genes that recognize the hepatocellular carcinoma (HCC)-associated antigen α-fetoprotein (AFP). Human T cells engineered to express the murine AFP-specific TCRs showed potent and specific cytotoxicity against HLA-A2+AFP+ HepG2 HCC tumor cells in vitro. Adoptive transfer of the engineered cells into immunodeficient mice efficiently prevented/eradicated large, established HepG2 HCC tumors.

Heterologous human/rat HER2-specific exosome-targeted T cell vaccine stimulates potent humoral and CTL responses leading to enhanced circumvention of HER2 tolerance in double transgenic HLA-A2/HER2 mice

Xie et al. used a vaccine strategy in which antigenic heterologous human/rat HER2/neu fusion protein, HuRt, was encoded and delivered via adenoviral vector to dendritic cells (DCs). Antigen-expressing exosomes purified from DCs were then transferred to a culture of Con-A-activated CD4+ T cells, converting them into antigen-presenting cells. The resulting T cell vaccine stimulated potent, HER2-specific antibody and CTL responses that had prophylactic and therapeutic antitumor efficacy, even in a setting of established HER2 tolerance.

Pre-existing Immunity to Oncolytic Virus Potentiates Its Immunotherapeutic Efficacy

Ricca et al. addressed the question of whether pre-existing antiviral immunity impacted the efficacy of intratumoral oncolytic virus therapy by first pre-establishing a systemic immune response against Newcastle Disease Virus (NDV) prior to tumor implantation and intratumoral virotherapy. Surprisingly, the efficacy of intratumoral oncolytic viral therapy increased in animals immune to NDV, leading to reduced Tregs and enhanced infiltration of distal tumors by conventional CD4+ and CD8+ T cells. CD8+ T cells, and to a lesser extent NK cells, were critical to this effect.

Everything New this Week In...

Close Modal

Small change for you. Big change for us!

This Thanksgiving season, show your support for cancer research by donating your change.

In less than a minute, link your credit card with our partner RoundUp App.

Every purchase you make with that card will be rounded up and the change will be donated to ACIR.

All transactions are securely made through Stripe.