Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2017 Sep 21;2(18):e93265.
doi: 10.1172/jci.insight.93265.

Combination central tolerance and peripheral checkpoint blockade unleashes antimelanoma immunity

Affiliations

Combination central tolerance and peripheral checkpoint blockade unleashes antimelanoma immunity

Pearl Bakhru et al. JCI Insight. .

Abstract

Blockade of immune checkpoint proteins (e.g., CTLA-4, PD-1) improves overall survival in advanced melanoma; however, therapeutic benefit is limited to only a subset of patients. Because checkpoint blockade acts by "removing the brakes" on effector T cells, the efficacy of checkpoint blockade may be constrained by the limited pool of melanoma-reactive T cells in the periphery. In the thymus, autoimmune regulator (Aire) promotes deletion of T cells reactive against self-antigens that are also expressed by tumors. Thus, while protecting against autoimmunity, Aire also limits the generation of melanoma-reactive T cells. Here, we show that Aire deficiency in mice expands the pool of CD4+ T cells capable of melanoma cell eradication and has additive effects with anti-CTLA-4 antibody in slowing melanoma tumor growth and increasing survival. Moreover, pharmacologic blockade of central T cell tolerance and peripheral checkpoint blockade in combination enhanced antimelanoma immunity in a synergistic manner. In melanoma patients treated with anti-CTLA-4 antibody, clinical response to therapy was associated with a human Aire polymorphism. Together, these findings suggest that Aire-mediated central tolerance constrains the efficacy of peripheral checkpoint inhibition and point to simultaneous blockade of Aire and checkpoint inhibitors as a novel strategy to enhance antimelanoma immunity.

Keywords: Cancer immunotherapy; Cellular immune response; Immunology; Melanoma.

PubMed Disclaimer

Conflict of interest statement

Conflict of interest: The authors have declared that no conflict of interest exists.

Figures

Figure 1
Figure 1. Association between Aire and antimelanoma effects of CTLA-4 blockade in mice and humans.
(A and B) WT and AireGW/+ mice were s.c. injected with 1 × 105 B16 melanoma cells followed by anti–CTLA-4 (aCTLA-4) antibody or isotype control (iso) antibody treatment. Mice in each group were followed for B16 melanoma tumor growth and survival. n = 7–12 per group, cumulative of at least 2 independent experiments. Mann-Whitney U test. *P < 0.05. (C and D) Comparison of progression-free survival (PFS) and overall survival (OS) probability between patients with Aire SNP rs1055311 (TT versus CT/CC).
Figure 2
Figure 2. Aire deficiency increases CD4+ T cell cytolytic function to reduce melanoma growth.
CD4+ splenocytes were transferred from WT and AireGW/+ donors into RAG–/– recipients, followed by s.c. B16 melanoma injection on day 7. Recipients were followed for tumor growth and survival. (A and B) B16 melanoma tumor growth and survival was measured after B16 inoculation in recipients; n = 15 per group. Mann-Whitney U test. *P < 0.05, **P < 0.01. (C–E) Tumor-infiltrating lymphocytes (TIL) were harvested on day 19 following B16 melanoma inoculation in recipients of either WT or AireGW/+ CD4+ splenocytes. Absolute numbers of CD4 tumor-infiltrating cells are shown in C. Two-tailed t test. Representative flow cytometry plots and average cumulative frequencies of Ki67+ (D) and KLRG1+ and granzyme B+ (E), among CD4+ T cells. Two-tailed t test. *P < 0.05, ****P < 0.0001. (F) Representative flow cytometry histograms and average absolute CellTrace Violet–labeled B16 cell numbers after coincubation with WT and AireGW/+ CD4+ T cells. Two-tailed t test. *P < 0.05. (G) Average relative B16 cell numbers (log2) after coincubation with CD4+ T cells from WT and AireGW/+ mice, along with pan-granzyme inhibitor (3, 4 Dichloroisocoumarin). One-way ANOVA and two-tailed t tests, with P values adjusted using Hommel’s correction for multiple comparisons. *P < 0.05.
Figure 3
Figure 3. Additive effect of Aire deficiency and anti–CTLA-4 antibody on T cell responses.
(A and B) Tumor-infiltrating lymphocytes (TIL) were harvested on day 19 following B16 melanoma inoculation in WT and AireGW/+ mice treated with aCTLA-4 or isotype control antibody (iso). Representative flow cytometry plots and average cumulative frequencies of Ki67+ (A) and KLRG1+ and granzyme B+ (B) among CD4+ T cells. n = 9–12 in each group. One-way ANOVA and two-tailed t tests with P values adjusted using Hommel’s correction for multiple comparisons. *P < 0.05, **P < 0.01. (C) ELISPOT with splenocytes harvested on day 14 following B16 melanoma inoculation in WT and AireGW/+ mice treated with anti–CTLA-4 antibody (aCTLA-4) or iso. Cumulative spot forming cells (SFC)/5×105 cells secreting IL-2 with OVA and TRP-1 was analyzed. Each data point represents an individual animal. One-way ANOVA and two-tailed t tests with P values adjusted using Hommel’s correction for multiple comparisons. *P < 0.05.
Figure 4
Figure 4. Additive antimelanoma effect of Aire deficiency and anti–CTLA-4 antibody in TRP-1 CD4+ TCR Tg mice.
(A and B) Aire sufficient (WT) and AireGW/+ TRP-1 TCR Tg mice were s.c. injected with 1 × 105 B16 melanoma cells followed by anti–CTLA-4 antibody (aCTLA-4) or isotype control antibody (Iso) treatment, as outlined. Mice in each group were followed for B16 melanoma tumor growth (A) and survival (B). n = 7–12 per group, cumulative of at least 2 independent experiments. Mann-Whitney U test. *P < 0.05. (C) Examples of periporbital and tail vitiligo in AireGW/+ mice treated with aCTLA-4 antibody.
Figure 5
Figure 5. Aire deficiency does not exacerbate the development of experimental autoimmune colitis associated with anti–CTLA-4 antibody treatment.
(A) Percent initial body weight of RAG–/– recipients after transfer of WT or AireGW/+CD4+CD25CD45RBhi splenocytes. After transfer, recipients were treated with anti–CTLA-4 antibody (aCTLA-4), untreated, or treated with isotype control antibody (iso). Cumulative data from 2 independent experiments are shown. *P < 0.05 comparing aCTLA-4 antibody treatment versus untreated/iso treatment in recipients of WT cells. (B) Representative H&E-stained sections of descending colons and (C) average histopathological scores of colons from recipients of CD4+CD25CD45RBhi splenocytes derived from WT and AireGW/+ mice and administered aCTLA-4 or untreated/iso (-). Gray arrowheads, immune cell infiltration in lamina propria; black arrowheads, immune cell infiltration in the submucosa. One-way ANOVA and two-tailed t tests with P values adjusted using Hommel’s correction for multiple comparisons. *P < 0.05.
Figure 6
Figure 6. Combination aRANKL and aCTLA-4 antibody administration enhances immune rejection of melanoma.
(A) Schematic of treatment regimen, in which anti-RANKL (aRANKL) or isotype control (iso) antibody was injected every other day for 11 days, followed by s.c. B16 melanoma injection (2 × 104 cells) on day 18. GVAX with or without anti-CTLA-4 (aCTLA-4) antibody was given on days 3, 6, and 10 following melanoma injection. (B) Survival curves in GVAX-treated mice receiving indicated combinations of therapy. n = 10 for each group. Mann-Whitney U test. *P < 0.05. Tumor-infiltrating lymphocytes (TIL) were harvested on day 19 following 1 × 105 B16 melanoma cell inoculation. Representative flow cytometry plots and average cumulative frequencies of Ki67+ (C) and KLRG1+ and granzyme B+ (D) among CD4+ T cells was measured. n = 9–12 in each group. One-way ANOVA and two-tailed t tests with P values adjusted using Hommel’s correction for multiple comparisons. *P < 0.05.

Similar articles

Cited by

References

    1. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12(4):252–264. doi: 10.1038/nrc3239. - DOI - PMC - PubMed
    1. Hodi FS, et al. Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med. 2010;363(8):711–723. doi: 10.1056/NEJMoa1003466. - DOI - PMC - PubMed
    1. Schadendorf D, et al. Pooled Analysis of Long-Term Survival Data From Phase II and Phase III Trials of Ipilimumab in Unresectable or Metastatic Melanoma. J Clin Oncol. 2015;33(17):1889–1894. doi: 10.1200/JCO.2014.56.2736. - DOI - PMC - PubMed
    1. Wolchok JD, et al. Nivolumab plus ipilimumab in advanced melanoma. N Engl J Med. 2013;369(2):122–133. doi: 10.1056/NEJMoa1302369. - DOI - PMC - PubMed
    1. Ribas A, et al. Pembrolizumab versus investigator-choice chemotherapy for ipilimumab-refractory melanoma (KEYNOTE-002): a randomised, controlled, phase 2 trial. Lancet Oncol. 2015;16(8):908–918. doi: 10.1016/S1470-2045(15)00083-2. - DOI - PMC - PubMed

Publication types