Weekly Digests
‹ Back to August

Distinct Cellular Mechanisms Underlie Anti-CTLA-4 and Anti-PD-1 Checkpoint Blockade

August 30, 2017

As the role of anti-PD-1 and anti-CTLA-4 immune checkpoint blockades (ICB) in cancer therapy continues to expand, understanding the underlying mechanisms of action becomes more imperative. Based on temporal, spatial, and cellular source differences, Wei et al. hypothesized that the anti-tumor immune responses to anti-CTLA-4 and anti-PD-1 treatment are driven by distinct mechanisms.

High dimensional mass cytometry is a powerful tool to interrogate multiple surface and intracellular markers on cells simultaneously, and coupled with unsupervised clustering analysis, it is capable of readily identifying cellular populations. To find out what goes on during the immune response following treatment, Wei et al. utilized this approach to identify and characterize the immune cells infiltrating tumors after ICB. The team transplanted syngeneic tumors into mice, which were then treated with either anti-CTLA-4 or anti-PD-1. The researchers specifically chose a tumor dose, treatment schedule, and analysis timepoint that resulted in incomplete rejection of tumors so that the tumor-infiltrating lymphocytes (TILs) could be analyzed.

In the highly immunogenic MC38 mouse colorectal tumor model, the team found an increase in the CD8+/Treg ratio after either treatment. Unsupervised clustering allowed for a deeper analysis of the data that revealed 15 distinct T cell clusters within the tumor. A phenotypically exhausted PD-1hiTIM3+CD8+ T cell population expanded more than other CD8+ T cell populations with either therapy. Based on short term labeling studies, this was likely due to increased proliferation within the tumor microenvironment, rather than increased T cell infiltration. The frequency of Treg cells decreased after both treatments, although, unsurprisingly, the effect was more pronounced following anti-CTLA-4 treatment. Finally, a significant increase in TBET+ Th1-like CD4+ effector cells was observed following treatment with anti-CTLA-4 but not anti-PD-1. A detailed look at the gene expression of MC38 tumor infiltrating cells revealed that ICOS+CD4+ T cells underwent significant, but mostly non-overlapping, transcriptional changes in response to anti-CTLA-4 and anti-PD-1 treatments, supporting the hypothesis that the two therapies have distinct underlying mechanisms.

The TIL analysis yielded nearly identical results in the B16/BL6 mouse melanoma model, an intrinsically weakly immunogenic tumor which required a GVAX tumor vaccine to boost overall T cell infiltration prior to ICB administration. Combined, the data from the two different mouse models suggest that the cellular mechanisms of both ICBs are different from one another, primarily with regard to CD4+ effector cell expansion, and are independent of tumor type.

To better understand the role of each tumor-infiltrating T cell subpopulation in the anti-tumor response, Wei et al. explored the correlations between T cell populations and tumor growth within the B16/BL6 model, and the results are presented below.

The two CD8+ T cell populations that were negatively correlated with tumor growth (MC2 and MC10) were activated and expanded after either ICB treatment. Surprisingly, one CD8+ phenotype (MC11) was positively correlated with tumor growth. This population was non-proliferative and may represent T cells with a central memory phenotype that are unrelated to the tumor, suggesting that infiltration of excess tumor-irrelevant cells may inhibit the anti-tumor response. Further analysis showed that the high proliferative rate of CD8+ T cells within the tumor in the later stages of the response was not necessarily correlated with anti-tumor effect. Overall, this detailed analysis indicates that only certain subpopulations of tumor-infiltrating T cells are involved in the response to ICB, and they may serve as predictive markers in the clinical setting. However, due to the study design of partial tumor regression, it is unclear whether the same mechanisms and cell populations would be observed during complete tumor regression, or whether the identified tumor-infiltrating T cell subsets are both necessary and sufficient for tumor rejection. In addition, the question remains whether all or only some distinct subsets of the phenotypically exhausted T cells that expanded following ICB treatment are functionally revived by ICB.

In a translational study, the researchers also performed mass cytometry analysis on resected tumors from 7 patients with metastatic melanoma who were treated with anti-PD-1, anti-CTLA-4, or a combination of both. They discovered that in the human setting, only two T cell subsets were significantly expanded in ICB-treated tumors compared to normal donor blood: the CD45RO+PD-1+TBET+EOMES+CD8+ T cell phenotype (exhausted, terminally differentiated), and the CD45RO+ICOS+PD-1loTBET+ Th1-like CD4+ effector T cell phenotype. These T cell populations were analogous to the corresponding phenotypes in the murine models (MC10 and MC3, respectively). The frequency of the Th1-like CD4+ effector T cells in human melanomas was higher after anti-CTLA-4 treatment compared to anti-PD-1 treatment, which was consistent with preclinical data.

Together, these murine and human results demonstrate that the mechanisms of anti-PD-1 and anti-CTLA-4 blockade are both similar (in the shared expansion of exhausted CD8+ populations by both therapies) and distinct (in the unique expansion of Th1-like CD4+ T cells by anti-CTLA-4 only), and contribute new insights and hypotheses to the optimization of combination therapies in the future.

by Anna Scherer

References:

Wei S.C., Levine J.H., Cogdill A.P., Zhao Y., Anang N.A.S., Andrews M.C., Sharma P., Wang J., Wargo J.A., Pe'er D., Allison J.P. Distinct Cellular Mechanisms Underlie Anti-CTLA-4 and Anti-PD-1 Checkpoint Blockade. Cell. 2017 Aug 9.

In the Spotlight...

Hematopoietic stem cell involvement in BCR-ABL1-positive ALL as potential mechanism of resistance to blinatumomab therapy

In two B-cell precursor acute lymphoblastic leukemia patients who relapsed after CD19-targeting blinatumomab, Nagel et al. used fluorescent in situ hybridization with BCR-ABL probes to demonstrate that relapsing cells were due to CD19- BCR-ABL1-positive hematopoietic stem cell progenitor and myeloid lineage cells, suggesting that combination therapies are needed to target both CD19+ leukemic B cells and CD19- malignant precursor cells.

Novel prostate cancer immunotherapy with a DNA-encoded anti-prostate-specific membrane antigen monoclonal antibody

Muthumani et al. developed a synthetic DNA plasmid encoding a PSMA-specific mAb. The in vitro- and/or in vivo-produced therapeutic mAbs bound Fc receptors and mediated ADCC in vitro. In C57BL/6 mice challenged with PSMA-expressing TRAMP-C2 cells, the DNA vaccine suppressed tumor growth and prolonged survival via NK cell-mediated ADCC.

Trastuzumab Increases HER2 Uptake and Cross-Presentation by Dendritic Cells

Gall et al. demonstrate in vitro and in vivo that trastuzumab facilitates the FcR-mediated uptake of soluble HER2 by dendritic cells, which increases cross-presentation of the immunodominant HER2 epitope E75. This priming of adaptive immunity in trastuzumab-treated HER2+ breast cancer patients may explain the synergy between HER2-derived peptide vaccines and trastuzumab observed in ongoing clinical trials.

CMTM6 maintains the expression of PD-L1 and regulates anti-tumour immunity

Using a genome-wide CRISPR-Cas9 screen, Burr et al. identified the ubiquitously expressed protein CMTM6 as a critical regulator of PD-L1. CMTM6 co-localizes with PD-L1 at the plasma membrane and maintains PD-L1 cell surface expression by preventing PD-L1 from being targeted for lysosome-mediated degradation during endocytic recycling. Both human in vitro and murine in vivo results support consideration of CMTM6 as a novel immunotherapeutic target.

Identification of CMTM6 and CMTM4 as PD-L1 protein regulators

Using a FACS-based haploid genetic screen, Mezzadra et al. identified CMTM6 as a regulator of PD-L1 on tumor and dendritic cells, and CMTM4 as a backup regulatory protein. CMTM6/4 associate with PD-L1 at the cell surface and act by protecting PD-L1 from ubiquitination and destabilization by STUB1, increasing PD-L1 protein half life. Genetic knockout of CMTM6/4 in tumor cells reduced PD-L1 surface expression and restored T cell function.

Novel landscape of HLA-G isoforms expressed in clear cell renal cell carcinoma patients

HLA-G is a newly discovered immune checkpoint affecting both innate and adaptive immune cells. Using antibodies, RNAseq, and RT-PCR, Roux et al. uncovered significant inter- and intra-tumoral HLA-G heterogeneity in ccRCC patients and described additional HLA-G isoforms that resulted from alternative splicing due to exon skipping or intron retention. This extensive heterogeneity suggests HLA-G is a complex immune and diagnostic target.

Agonist anti-GITR antibody significantly enhances the therapeutic efficacy of Listeria monocytogenes-based immunotherapy

A combination of a Listeria-E7 vaccine and an agonist anti-GITR antibody yielded a synergistic anti-tumor effect (60% complete tumor regression) and long-term systemic immunity in HPV-E6/E7 TC-1 tumor-bearing mice. The effect is likely due to an increase in infiltrating and peripheral antigen-specific CD8+ T cells, a dilution of T regulatory cells via expansion of FoxP3- CD4+ cells, and the stabilization of myeloid-derived suppressor cells.

Treatment of Patients With Metastatic Cancer Using a Major Histocompatibility Complex Class II-Restricted T-Cell Receptor Targeting the Cancer Germline Antigen MAGE-A3

Treatment of seventeen patients with various solid tumors with autologous CD4+ T cells engineered to express an MHC II-restricted TCR targeting the cancer germline antigen MAGE-A3 resulted in one patient with an objective ongoing complete response and three patients with partial responses of varying durations. All patients showed increased cytokine expression with limited toxicity. T cell persistence was related to initial dose, but no markers correlated with clinical outcomes.

Macrophage Polarization Contributes to Glioblastoma Eradication by Combination Immunovirotherapy and Immune Checkpoint Blockade

Using a non-immunogenic, intracranial glioblastoma stem-like cell tumor and the more aggressive CT-2A model, Saha et al. show that the triplet of an IL-12 expressing oncolytic herpes virus, anti-CTLA-4, and anti-PD-1 checkpoint therapy led to significant long term survival and resistance to re-challenge, overcoming multiple forms of immunosuppression. Noteworthy were the criticality of CD4+ T cells, near criticality of CD8+ cells, and the increase in M1-polarized tumor associated macrophages.

Genomic and immune heterogeneity are associated with differential responses to therapy in melanoma

In melanoma patients bearing multiple “synchronous” metastases, intrapatient immune heterogeneity (immune cell infiltrates, TCR clonality) was consistently more pervasive than, but correlated with intrapatient genomic heterogeneity. Checkpoint or targeted therapy revealed substantial clinical response heterogeneity between lesions as well. A novel radiomic “texture analysis” of CT images at baseline was predictive of clinical response.

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.