Association and prognostic significance of BRCA1/2-mutation status with neoantigen load, number of tumor-infiltrating lymphocytes and expression of PD-1/PD-L1 in high grade serous ovarian cancer

Kyle C Strickland, Brooke E Howitt, Sachet A Shukla, Scott Rodig, Lauren L Ritterhouse, Joyce F Liu, Judy E Garber, Dipanjan Chowdhury, Catherine J Wu, Alan D D'Andrea, Ursula A Matulonis, Panagiotis A Konstantinopoulos, Kyle C Strickland, Brooke E Howitt, Sachet A Shukla, Scott Rodig, Lauren L Ritterhouse, Joyce F Liu, Judy E Garber, Dipanjan Chowdhury, Catherine J Wu, Alan D D'Andrea, Ursula A Matulonis, Panagiotis A Konstantinopoulos

Abstract

Immune checkpoint inhibitors (e.g., anti-PD-1 and anti-PD-L1 antibodies) have demonstrated remarkable efficacy against hypermutated cancers such as melanomas and lung carcinomas. One explanation for this effect is that hypermutated lesions harbor more tumor-specific neoantigens that stimulate recruitment of an increased number of tumor-infiltrating lymphocytes (TILs), which is counterbalanced by overexpression of immune checkpoints such as PD-1 or PD-L1. Given that BRCA1/2-mutated high grade serous ovarian cancers (HGSOCs) exhibit a higher mutational load and a unique mutational signature with an elevated number of larger indels up to 50 bp, we hypothesized that they may also harbor more tumor-specific neoantigens, and, therefore, exhibit increased TILs and PD-1/PD-L1 expression. Here, we report significantly higher predicted neoantigens in BRCA1/2-mutated tumors compared to tumors without alterations in homologous recombination (HR) genes (HR-proficient tumors). Tumors with higher neoantigen load were associated with improved overall survival and higher expression of immune genes associated with tumor cytotoxicity such as genes of the TCR, the IFN-gamma and the TNFR pathways. Furthermore, immunohistochemistry studies demonstrated that BRCA1/2-mutated tumors exhibited significantly increased CD3+ and CD8+ TILs, as well as elevated expression of PD-1 and PD-L1 in tumor-associated immune cells compared to HR-proficient tumors. Survival analysis showed that both BRCA1/2-mutation status and number of TILs were independently associated with outcome. Of note, two distinct groups of HGSOCs, one with very poor prognosis (HR proficient with low number of TILs) and one with very good prognosis (BRCA1/2-mutated tumors with high number of TILs) were defined. These findings support a link between BRCA1/2-mutation status, immunogenicity and survival, and suggesting that BRCA1/2-mutated HGSOCs may be more sensitive to PD-1/PD-L1 inhibitors compared to HR-proficient HGSOCs.

Keywords: BRCA1 and BRCA2 mutations; PD-1 and PD-L1; high grade serous ovarian cancer; homologous recombination DNA repair; immunogenicity.

Conflict of interest statement

CONFLICTS OF INTEREST

The authors declare that they have no conflicts of interest to report.

Figures

Figure 1. Neoantigen load in BRCA1/2-mutated, non-BRCA1/2-mutated/HR-deficient…
Figure 1. Neoantigen load in BRCA1/2-mutated, non-BRCA1/2-mutated/HR-deficient and HR proficient cohorts, and association with outcome in the TCGA dataset
(A) Predicted neoantigen load in BRCA1/2-mutated (n = 54) vs all remaining non-BRCA1/2-mutated tumors (n = 191). (B) Predicted neoantigen load in BRCA1/2-mutated (n = 54), HR deficient/non-BRCA1/2-mutated (n = 69) and HR proficient tumors (n = 122). (C) Predicted neoantigen load of HR-deficient (n = 123) vs HR-proficient (n = 122). (D) Predicted neoantigen load of BRCA1- versus BRCA2-mutated tumors. (E) Tumors in the lowest quartile of neoantigen load were associated with significantly lower overall survival compared to the remaining tumors. Of the 60 tumors in the lower quartile, 20 were HR deficient and 40 were HR proficient. (F) Tumors in the lowest quintile of neoantigen load were associated with significantly lower overall survival compared to the remaining tumors. Of the 47 tumors in the lower quintile, 19 were HR deficient and 28 were HR proficient.
Figure 2. Outline of our study cohorts
Figure 2. Outline of our study cohorts
(A) Prediction of neoantigen load in the TCGA dataset. (B) Determination of BRCA1/2-mutated and HR proficient subsets in our institutional cohort. The BRCA1/2-mutated group was comprised of 37 HGSOCs with BRCA1/2 germline mutations (29 with BRCA1 and 8 with BRCA2 mutations) identified by genetic testing (left). The HR-proficient (HR intact) group (i.e. group without HR alterations) comprised 16 ovarian cancers which were identified in a two-step process (right). First, NGS excluded tumors with mutations in HR genes and this analysis identified 17 such tumors. Tumor was excluded based on absent BRCA1 expression by immunohistochemistry.
Figure 3. Results of BRCA1 immunohistochemistry
Figure 3. Results of BRCA1 immunohistochemistry
(A) Positive BRCA1 IHC in a representative case. BRCA1 expression was positive by IHC in 16 of the 17 tumors without HR alterations identified by NGS. (B) BRCA1 IHC was negative in one tumor that did not harbor HR alterations by NGS. Focal BRCA1 positivity was present in lymphocytes. Interestingly, review of the NGS data for this case demonstrated that the tumor had a single copy deletion of the BRCA1 gene, suggesting that BRCA1 loss in this tumor was likely due to single copy deletion of BRCA1 and epigenetic silencing of the complementary allele. (C) Corresponding area of tumor on H & E stain demonstrates the presence of intratumoral lymphocytes. (D) The presence of intraepithelial lymphocytes was confirmed by a CD3 IHC.
Figure 4. CD3+, CD4+ CD8+ and CD20+…
Figure 4. CD3+, CD4+ CD8+ and CD20+ intraepithelial lymphocytes in BRCA1/2-mutated vs HR proficient tumors
(A) Photomicrographs of representative BRCA1/2-mutated and HR-intact tumors depicting H & E staining and immunohistochemistry for CD3, CD4, CD8 and CD20. (B) Quantification and comparison of CD3+ TILs from BRCA1/2-mutated and HR intact tumors. (C) Quantification and comparison of CD4+ TILs from BRCA1/2-mutated and HR intact tumors. (D) Quantification and comparison of CD8+ TILs from BRCA1/2-mutated and HR intact tumors. (E) Quantification and comparison of CD20+ TILs from BRCA1/2-mutated and HR intact tumors.
Figure 5. PD-1 and PD-L1 expression in…
Figure 5. PD-1 and PD-L1 expression in the intraepithelial and peritumoral immune cells of BRCA1/2-mutated versus HR-proficient tumors
(A) Photomicrographs of representative BRCA1/2-mutated and HR-intact tumors depicting H & E staining and immunohistochemistry for PD-1 and PD-L1. Photomicrographs depict cases from each study group that were scored as positive. (B) Bar graphs illustrating the number of tumors with increased PD-1 and PD-L1 positive intraepithelial and peritumoral immune cells, as well as the number of tumors positive for PD-L1 in tumor cells of BRCA1/2-mutated and HR intact cases.
Figure 6. Association of CD3+ TILs and…
Figure 6. Association of CD3+ TILs and BRCA1/2-mutation status with survival in our institutional cohort
(A) Overall survival of patients with BRCA1/2-mutated (red) versus HR intact (HRP, blue) tumors. (B) Overall survival of patients with tumors containing above median number of CD3+ TILs/HPF (red) versus the remaining tumors (blue). (C) Overall survival of patients with tumors containing ≥ 13 CD3+ TILs/HPF (red) versus those containing < 13 CD3+ TILs/HPF (blue). (D) Overall survival of patients with BRCA1/2-mutated tumors with a high number of TILs (≥ 13 TILs/HPF) (red), HR proficient tumors (HRP) with a low number of TILs (< 13 TILs/HPF) (blue), and BRCA1/2-mutated tumors with low TILs or HRP tumors with high TILs (green).

References

    1. Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, Drake CG, Camacho LH, Kauh J, Odunsi K, Pitot HC, Hamid O, Bhatia S, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366:2455–2465.
    1. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, Powderly JD, Carvajal RD, Sosman JA, Atkins MB, Leming PD, Spigel DR, Antonia SJ, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med. 2012;366:2443–2454.
    1. Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, Skora AD, Luber BS, Azad NS, Laheru D, Biedrzycki B, Donehower RC, Zaheer A, et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N Engl J Med. 2015;372:2509–2520.
    1. Xiao Y, Freeman GJ. The microsatellite instable subset of colorectal cancer is a particularly good candidate for checkpoint blockade immunotherapy. Cancer Discov. 2015;5:16–18.
    1. Llosa NJ, Cruise M, Tam A, Wicks EC, Hechenbleikner EM, Taube JM, Blosser RL, Fan H, Wang H, Luber BS, Zhang M, Papadopoulos N, Kinzler KW, et al. The vigorous immune microenvironment of microsatellite instable colon cancer is balanced by multiple counter-inhibitory checkpoints. Cancer Discov. 2014;5:43–51.
    1. Hussein YR, Weigelt B, Levine DA, Schoolmeester JK, Dao LN, Balzer BL, Liles G, Karlan B, Kobel M, Lee CH, Soslow RA. Clinicopathological analysis of endometrial carcinomas harboring somatic POLE exonuclease domain mutations. Mod Pathol. 2015;28:505–514.
    1. Rooney MS, Shukla SA, Wu CJ, Getz G, Hacohen N. Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell. 2015;160:48–61.
    1. Brown SD, Warren RL, Gibb EA, Martin SD, Spinelli JJ, Nelson BH, Holt RA. Neo-antigens predicted by tumor genome meta-analysis correlate with increased patient survival. Genome Res. 2014;24:743–750.
    1. Howitt BE, Shukla SA, Sholl LM, Ritterhouse LL, Watkins JC, Rodig S, Stover E, Strickland KC, D'Andrea AD, Wu CJ, Matulonis UA, Konstantinopoulos PA. Association of Polymerase e-Mutated and Microsatellite-Instable Endometrial Cancers With Neoantigen Load, Number of Tumor-Infiltrating Lymphocytes, and Expression of PD-1 and PD-L1. JAMA Oncol. 2015;1:1319–23.
    1. Llosa NJ, Cruise M, Tam A, Wicks EC, Hechenbleikner EM, Taube JM, Blosser RL, Fan H, Wang H, Luber BS, Zhang M, Papadopoulos N, Kinzler KW, et al. The vigorous immune microenvironment of microsatellite instable colon cancer is balanced by multiple counter-inhibitory checkpoints. Cancer Discov. 2015;5:43–51.
    1. TCGA Integrated genomic analyses of ovarian carcinoma. Nature. 2011;474:609–615.
    1. Konstantinopoulos PA, Ceccaldi R, Shapiro GI, D'Andrea AD. Homologous Recombination Deficiency: Exploiting the Fundamental Vulnerability of Ovarian Cancer. Cancer Discov. 2015;5:1137–54.
    1. Bolton KL, Chenevix-Trench G, Goh C, Sadetzki S, Ramus SJ, Karlan BY, Lambrechts D, Despierre E, Barrowdale D, McGuffog L, Healey S, Easton DF, Sinilnikova O, et al. Association between BRCA1 and BRCA2 mutations and survival in women with invasive epithelial ovarian cancer. JAMA. 2012;307:382–390.
    1. Boyd J, Sonoda Y, Federici MG, Bogomolniy F, Rhei E, Maresco DL, Saigo PE, Almadrones LA, Barakat RR, Brown CL, Chi DS, Curtin JP, Poynor EA, et al. Clinicopathologic features of BRCA-linked and sporadic ovarian cancer. JAMA. 2000;283:2260–2265.
    1. Ceccaldi R, Liu JC, Amunugama R, Hajdu I, Primack B, Petalcorin MI, O'Connor KW, Konstantinopoulos PA, Elledge SJ, Boulton SJ, Yusufzai T, D'Andrea AD. Homologous-recombination-deficient tumours are dependent on Poltheta-mediated repair. Nature. 2015;518:258–262.
    1. Mateos-Gomez PA, Gong F, Nair N, Miller KM, Lazzerini-Denchi E, Sfeir A. Mammalian polymerase theta promotes alternative NHEJ and suppresses recombination. Nature. 2015;518:254–257.
    1. Yousefzadeh MJ, Wood RD. DNA polymerase POLQ and cellular defense against DNA damage. DNA repair. 2013;12:1–9.
    1. Birkbak NJ, Kochupurakkal B, Izarzugaza JM, Eklund AC, Li Y, Liu J, Szallasi Z, Matulonis UA, Richardson AL, Iglehart JD, Wang ZC. Tumor mutation burden forecasts outcome in ovarian cancer with BRCA1 or BRCA2 mutations. PLoS One. 2013;8:e80023.
    1. Alexandrov LB, Nik-Zainal S, Wedge DC, Aparicio SA, Behjati S, Biankin AV, Bignell GR, Bolli N, Borg A, Borresen-Dale AL, Boyault S, Burkhardt B, Butler AP, et al. Signatures of mutational processes in human cancer. Nature. 2013;500:415–421.
    1. Shukla SA, Rajasagi M, Tiao G, Dixon PM, Lawrence MS, Stevens J, Lane WJ, Dellagatta JL, Steelman S, Sougnez C, Cibulskis K, Kiezun A, Brusic V, et al. Comprehensive analysis of cancer-associated somatic mutations in class I HLA genes. Nat Biotechnol. 2015;33:1152–8.
    1. Nielsen M, Lundegaard C, Blicher T, Lamberth K, Harndahl M, Justesen S, Roder G, Peters B, Sette A, Lund O, Buus S. NetMHCpan, a method for quantitative predictions of peptide binding to any HLA-A and -B locus protein of known sequence. PLoS One. 2007;2:e796.
    1. Garg K, Levine DA, Olvera N, Dao F, Bisogna M, Secord AA, Berchuck A, Cerami E, Schultz N, Soslow RA. BRCA1 immunohistochemistry in a molecularly characterized cohort of ovarian high-grade serous carcinomas. Am J Surg Pathol. 2013;37:138–146.
    1. Zhang L, Conejo-Garcia JR, Katsaros D, Gimotty PA, Massobrio M, Regnani G, Makrigiannakis A, Gray H, Schlienger K, Liebman MN, Rubin SC, Coukos G. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N Engl J Med. 2003;348:203–213.
    1. Patch AM, Christie EL, Etemadmoghadam D, Garsed DW, George J, Fereday S, Nones K, Cowin P, Alsop K, Bailey PJ, Kassahn KS, Newell F, Quinn MC, et al. Whole-genome characterization of chemoresistant ovarian cancer. Nature. 2015;521:489–494.
    1. Herbst RS, Soria JC, Kowanetz M, Fine GD, Hamid O, Gordon MS, Sosman JA, McDermott DF, Powderly JD, Gettinger SN, Kohrt HE, Horn L, Lawrence DP, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014;515:563–567.
    1. Soslow RA, Han G, Park KJ, Garg K, Olvera N, Spriggs DR, Kauff ND, Levine DA. Morphologic patterns associated with BRCA1 and BRCA2 genotype in ovarian carcinoma. Mod Pathol. 2011;25:625–636.
    1. Clarke B, Tinker AV, Lee CH, Subramanian S, van de Rijn M, Turbin D, Kalloger S, Han G, Ceballos K, Cadungog MG, Huntsman DG, Coukos G, Gilks CB. Intraepithelial T cells and prognosis in ovarian carcinoma: novel associations with stage, tumor type, and BRCA1 loss. Mod Pathol. 2009;22:393–402.
    1. George J, Alsop K, Etemadmoghadam D, Hondow H, Mikeska T, Dobrovic A, deFazio A, Smyth GK, Levine DA, Mitchell G, Bowtell DD. Nonequivalent gene expression and copy number alterations in high-grade serous ovarian cancers with BRCA1 and BRCA2 mutations. Clin Cancer Res. 2013;19:3474–3484.
    1. Sato E, Olson SH, Ahn J, Bundy B, Nishikawa H, Qian F, Jungbluth AA, Frosina D, Gnjatic S, Ambrosone C, Kepner J, Odunsi T, Ritter G, et al. Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci U S A. 2005;102:18538–18543.
    1. Rajasagi M, Shukla SA, Fritsch EF, Keskin DB, DeLuca D, Carmona E, Zhang W, Sougnez C, Cibulskis K, Sidney J, Stevenson K, Ritz J, Neuberg D, et al. Systematic identification of personal tumor-specific neoantigens in chronic lymphocytic leukemia. Blood. 2014;124:453–62.
    1. Shukla SA, Rooney MS, Rajasagi M, Tiao G, Dixon PM, Lawrence MS, Stevens J, Lane WJ, Dellagatta JL, Steelman S, Sougnez C, Cibulskis K, Kiezun A, et al. Comprehensive analysis of cancer-associated somatic mutations in class I HLA genes. Nat Biotechnol. 2015;33:1152–1158.
    1. Lawrence MS, Stojanov P, Mermel CH, Robinson JT, Garraway LA, Golub TR, Meyerson M, Gabriel SB, Lander ES, Getz G. Discovery and saturation analysis of cancer genes across 21 tumour types. Nature. 2014;505:495–501.
    1. Wagle N, Berger MF, Davis MJ, Blumenstiel B, Defelice M, Pochanard P, Ducar M, Van Hummelen P, Macconaill LE, Hahn WC, Meyerson M, Gabriel SB, Garraway LA. High-throughput detection of actionable genomic alterations in clinical tumor samples by targeted, massively parallel sequencing. Cancer Discov. 2012;2:82–93.
    1. Hamanishi J, Mandai M, Iwasaki M, Okazaki T, Tanaka Y, Yamaguchi K, Higuchi T, Yagi H, Takakura K, Minato N, Honjo T, Fujii S. Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer. Proc Natl Acad Sci U S A. 2007;104:3360–3365.

Source: PubMed

3
Subscribe