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 Jun 7;9(393):eaal4922.
doi: 10.1126/scitranslmed.aal4922.

Combined immune checkpoint blockade as a therapeutic strategy for BRCA1-mutated breast cancer

Affiliations

Combined immune checkpoint blockade as a therapeutic strategy for BRCA1-mutated breast cancer

Emma Nolan et al. Sci Transl Med. .

Abstract

Immune checkpoint inhibitors have emerged as a potent new class of anticancer therapy. They have changed the treatment landscape for a range of tumors, particularly those with a high mutational load. To date, however, modest results have been observed in breast cancer, where tumors are rarely hypermutated. Because BRCA1-associated tumors frequently exhibit a triple-negative phenotype with extensive lymphocyte infiltration, we explored their mutational load, immune profile, and response to checkpoint inhibition in a Brca1-deficient tumor model. BRCA1-mutated triple-negative breast cancers (TNBCs) exhibited an increased somatic mutational load and greater numbers of tumor-infiltrating lymphocytes, with increased expression of immunomodulatory genes including PDCD1 (PD-1) and CTLA4, when compared to TNBCs from BRCA1-wild-type patients. Cisplatin treatment combined with dual anti-programmed death-1 and anti-cytotoxic T lymphocyte-associated antigen 4 therapy substantially augmented antitumor immunity in Brca1-deficient mice, resulting in an avid systemic and intratumoral immune response. This response involved enhanced dendritic cell activation, reduced suppressive FOXP3+ regulatory T cells, and concomitant increase in the activation of tumor-infiltrating cytotoxic CD8+ and CD4+ T cells, characterized by the induction of polyfunctional cytokine-producing T cells. Dual (but not single) checkpoint blockade together with cisplatin profoundly attenuated the growth of Brca1-deficient tumors in vivo and improved survival. These findings provide a rationale for clinical studies of combined immune checkpoint blockade in BRCA1-associated TNBC.

PubMed Disclaimer

Conflict of interest statement

Competing interests: No potential conflicts of interest were disclosed by the other authors.

Figures

Fig. 1
Fig. 1. BRCA1-mutated TNBCs are enriched for TILs and have a high mutational burden
(A) Stromal TILs in BRCA1-mutated (n = 29) versus WT primary TNBC (n = 64). P = 0.037 (Mann-Whitney U test). The combined cohort was from TCGA (n = 71) and a kConFab series of BRCA1-mutant tumors (n = 22). (B) Correlogram of stromal TILs and expression of key immune genes in BRCA1-mutant primary TNBC (n = 7). Stars indicate P < 0.05. Gene expression measured in transcripts per million (TPM). Pearson product-moment correlation coefficient is displayed. (C) Scatter plots of TILs versus TPM (logarithmic scale) for key immune genes [same data as (B)]. (D) Nonsilent mutation (missense/nonsense mutations and indels) burden in BRCA1-mutant (n = 7) versus WT primary TNBC from TCGA cohort (n = 64). P = 0.05 (Mann-Whitney U test). Refer to Materials and Methods for details on box plots.
Fig. 2
Fig. 2. BRCA1-mutated TNBCs exhibit prominent lymphocytic infiltrate and PD-L1 expression
(A) Representative H&E image of a BRCA1-mutated TNBC. Scale bar, 100 μm. (B) Analysis of matched BRCA1-positive TNBC patient stromal TIL populations for H&E, OPAL staining, stromal, and intratumoral PD-L1 expression (n = 16). (C) BRCA1-mutated TNBC. H&E and accompanying section immunostained for PD-L1. Scale bars, 100 mm. (D) Representative BRCA1-mutated TNBC patient sample with high stromal TILs. The inset indicates an area with a high number of CD3+ (*), CD4+ (**), and CD8+ (***) cells. Iso, isotype-matched control antibody. Additional images are shown in fig. S1 (A and B). Scale bars, 100 mm (main image) and 20 μm (inset). (E) BRCA1-mutated TNBCs. The OPAL staining panel includes tumor marker CK18 (yellow), CD3 (red), CD4 (white), CD8 (green), FOXP3 (orange), PD-L1 (cyan), and 4′,6-diamidino-2-phenylindole (DAPI) (blue). Scale bars, 100 μm. (F) OPAL staining for PD-L1 and CK18, revealing high intratumoral PD-L1 expression. Scale bar, 100 μm. (G) Sankey plot of a fluorescence-activated cell sorting (FACS) profile of CD3+ TILs from a BRCA1-mutated tumor, showing percentages of CD8+ and CD4+ cells and PD-1 expression within these subsets (see fig. S1C).
Fig. 3
Fig. 3. Combination therapy with checkpoint inhibitors curtails the growth of Brca1-deficient tumors
Graph depicting the percentages of PD-L1+ (A) tumor cells and (B) stromal cells (Lin+) within mammary tumors harvested from MMTV-cre/Brca1fl/fl/p53+/, MMTV-Neu, MMTV-PyMT, MMTV-Wnt1, and p53+/ mice. PD-L1 expression was determined by flow cytometry on freshly harvested tumors, and the percentage of positive cells was determined by comparing PD-L1 expression to an isotype-matched control antibody. Data are means ± SEM; each data point depicts an individual tumor. *P < 0.05, **P < 0.01. (C) Overview of treatment strategy: Freshly harvested MMTV-cre/Brca1fl/fl/p53+/ tumor cells were injected into the mammary fat pads of syngeneic (F1 FVB × BALB/c) mice. Three weeks after transplantation, mice were randomized to one of six treatment arms: (i) vehicle (PBS), (ii) anti–PD-1 and anti-CTLA4, (iii) cisplatin, (iv) cisplatin and anti–PD-1, (v) cisplatin and anti-CTLA4, and (vi) cisplatin, anti-CTLA4, and anti–PD-1. Mice received cisplatin on day 1 of each treatment cycle (days 1, 21, 42, and 63) and anti–PD-1 and anti-CTLA4 on days 2, 5, and 8 of each cycle. (D) Tumor growth curves for individual mice (n = 58). Arrows depict day 1 of a treatment cycle (treatment with cisplatin or vehicle control). (E) Kaplan-Meier survival curves depicting the augmented response of MMTV-cre/Brca1fl/fl/p53+/ tumors to combination therapy. Log-rank (Mantel-Cox) P value is shown for combination cisplatin, anti–PD-1, and anti-CTLA4 therapy versus cisplatin alone.
Fig. 4
Fig. 4. Combination therapy induces an avid immune response in Brca1-deficient tumors
(A) Schematic diagram of the experimental protocol. MMTV-cre/Brca1fl/fl/p53+/ tumor cells were transplanted into the fat pads of syngeneic (F1 FVB × BALB/c) mice. Tumors, spleens, and draining lymph nodes were either harvested before treatment initiation (baseline, day 0) or 14 days after treatment with cisplatin ± anti-CTLA4/anti–PD-1. The composition and activation status of immune cells were assessed by flow cytometry. Two independent experiments were performed (n = 5 mice per group per experiment). (B) Representative FACS plots showing FOXP3 versus CD8 expression on TCRβ+ tumor-infiltrating cells in mice receiving the treatments indicated. The percentage of cells bounded by each region is shown. (C) Quantification of the ratio of CD8+/FOXP3+ T cells infiltrating the tumors of mice receiving the various treatments. UT, untreated. (D) Histograms of ICOS, CD44, and NRP1 expression on tumor-infiltrating CD8+ (top panels) or CD4+FOXP3 (bottom panels) conventional T cells from untreated mice (gray shaded), mice treated with cisplatin (gray line), or mice treated with cisplatin, anti-CTLA4, and anti–PD-1 (black line). (E) Representative FACS plots depicting CD8 versus PD-1 expression on TCRβ+ tumor-infiltrating cells in mice receiving the treatments indicated. The percentage of cells bounded by each region is shown. (F) Quantification of the proportion of PD-1+ tumor-infiltrating CD8+ T cells. (G) Histograms of PD-1 expression on tumor-infiltrating CD8+ or CD4+ conventional T cells from untreated mice(gray shaded), mice treated with cisplatin (gray line), or mice treated with cisplatin, anti-CTLA4, and anti–PD-1 (black line). (H) Expression of PD-1 versus CD8 on TCRβ+ T cells from the draining lymph nodes of mice receiving the indicated treatments (left panels) and quantification of PD-1+CD8+ T cells (right panel). Flow cytometric analysis is representative of two experiments with n = 5 mice per group. Means ± SEM are shown, with analysis by one-way analysis of variance (ANOVA) and Tukey’s post-test. *P < 0.05, **P < 0.001, ***P < 0.0001, ****P < 0.00001.

Comment in

  • Immunotherapy: Keeping breast cancer in check.
    Lieben L. Lieben L. Nat Rev Cancer. 2017 Aug;17(8):454-455. doi: 10.1038/nrc.2017.55. Epub 2017 Jul 7. Nat Rev Cancer. 2017. PMID: 28684877 No abstract available.
  • Doubling Down on BRCA-Mutated Cancer.
    Wahner Hendrickson AE, Kaufmann SH, Swisher EM. Wahner Hendrickson AE, et al. Trends Cancer. 2017 Nov;3(11):743-744. doi: 10.1016/j.trecan.2017.09.005. Epub 2017 Sep 29. Trends Cancer. 2017. PMID: 29120748

References

    1. Foulkes WD, Smith IE, Reis-Filho JS. Triple-negative breast cancer. N Engl J Med. 2010;363:1938–1948. - PubMed
    1. Lord CJ, Ashworth A. BRCAness revisited. Nat Rev Cancer. 2016;16:110–120. - PubMed
    1. Bouwman P, Jonkers J. Molecular pathways: How can BRCA-mutated tumors become resistant to PARP inhibitors? Clin Cancer Res. 2014;20:540–547. - PubMed
    1. Schreiber RD, Old LJ, Smyth MJ. Cancer immunoediting: Integrating immunity’s roles in cancer suppression and promotion. Science. 2011;331:1565–1570. - PubMed
    1. Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: A common denominator approach to cancer therapy. Cancer Cell. 2015;27:450–461. - PMC - PubMed

Publication types

MeSH terms