Abstract
PURPOSE
Ovarian cancers can exhibit a prominent immune infiltrate, but clinical trials have not demonstrated substantive response rates to immune checkpoint blockade monotherapy. We aimed to understand genomic features associated with immunogenicity in BRCA1/2 mutation–associated cancers.
MATERIALS AND METHODS
Using the Cancer Genome Atlas whole-exome sequencing, methylation, and expression data, we analyzed 66 ovarian cancers with either germline or somatic loss of BRCA1/2 and whole-exome sequencing, immunohistochemistry, and CyTOF in 20 ovarian cancers with germline BRCA1/2 pathogenic variants from Penn.
RESULTS
We found two groups of BRCA1/2 ovarian cancers differing in their immunogenicity: (1) 37 tumors significantly enriched for PTEN loss (11, 30%) and BRCA1 promoter–hypermethylated (10, 27%; P = .0016) and (2) PTEN wild-type (28 of 29 tumors) cancers, with the latter group having longer overall survival (OS; P = .0186, median OS not reached v median OS = 66.1 months). BRCA1/2-mutant PTEN loss and BRCA1 promoter–hypermethylated cancers were characterized by the decreased composition of lymphocytes estimated by gene expression (P = .0030), cytolytic index (P = .034), and cytokine expression but higher homologous recombination deficiency scores (P = .00013). Large-scale state transitions were the primary discriminating feature (P = .001); neither mutational burden nor neoantigen burden could explain differences in immunogenicity. In Penn tumors, PTEN loss and high homologous recombination deficiency cancers exhibited fewer CD3+ (P = .05), CD8+ (P = .012), and FOXP3+ (P = .0087) T cells; decreased PRF1 expression (P = .041); and lower immune costimulatory and inhibitory molecule expression.
CONCLUSION
Our study suggests that within ovarian cancers with genetic loss of BRCA1/2 are two subsets exhibiting differential immunogenicity, with lower levels associated with PTEN loss and BRCA hypermethylation. These genomic features of BRCA1/2-associated ovarian cancers may inform considerations around how to optimally deploy immune checkpoint inhibitors in the clinic.
INTRODUCTION
BRCA1/2 are essential proteins involved in homologous recombination (HR)–based DNA repair.1 Ovarian cancers with germline and somatic alterations in BRCA1/2 share many phenotypic characteristics including defects in double-strand break repair, replication fork stalling, and mutational signatures reflective of underlying HR deficiency (HRD).2,3 Consequently, they exhibit similar therapeutic vulnerabilities, namely, sensitivity to inhibitors of poly(ADP-ribose) polymerase (PARP) and platinum-based chemotherapy.4-12 However, resistance to DNA-damaging therapy, whether intrinsic in the form of the absence of BRCA1/2 allele–specific loss of heterozygosity13 or acquired via secondary genetic events,9,14-18 has necessitated the investigation of orthogonal treatment strategies.
CONTEXT
Key Objective
To understand genomic features associated with immunogenicity in BRCA1/2 mutation–associated ovarian cancers.
Knowledge Generated
We found that BRCA1/2 mutation–associated ovarian groups clustered into two groups: Immune-High, associated with PTEN-loss and BRCA1 promoter–methylated tumors, and Immune-Low, with a significantly lower overall survival. BRCA1/2 mutation–associated ovarian cancers with PTEN loss had significantly higher homologous recombination deficiency scores, but exhibited significantly fewer CD3+, CD8+, and FOXP3+ T cells.
Relevance
Guided by molecular features, BRCA1/2 mutation–associated ovarian cancers can be divided into two groups with differing levels of immunogenicity, which may inform the use of immune checkpoint inhibitors in this patient group.
One treatment strategy that has been hypothesized to impart clinical benefits in BRCA1/2-deficient ovarian cancer is immune checkpoint blockade (ICB).19-25 BRCA1/2-deficient ovarian cancers tend to have a relatively increased neoantigen load, because of a reliance on error-prone double-strand break repair,22 which can be predictive in some tumor types of ICB response.20,22,23 BRCA1/2-deficient ovarian tumors are also characterized by a high presence of tumor-infiltrating lymphocytes (TILs),20,22,26,27 which is a positive prognostic factor for survival.20,22,24 PARP1 inhibition (PARPi), to which BRCA1/2-deficient ovarian cancers can respond,5,16,28 has also been shown to increase TILs and synergistically combine with inhibitors of the immune checkpoint protein CTLA4 in mouse models.29,30
Despite the immunogenic properties of BRCA1/2-deficient ovarian cancers, the results of clinical trials evaluating ICB have been variable.31 A single-agent phase Ib clinical trial of the programmed death-1 (PD1) inhibitor avelumab (n = 125) with epithelial ovarian cancer found an objective response rate (ORR) of 9.6% and a 1-year progression-free survival (PFS) of 10.2%.32 Patients with BRCA1/2 mutations did not selectively benefit over BRCA1/2 wild-type (WT) patients in this trial.32,33 A phase II ovarian cancer study of pembrolizumab (KEYNOTE-100, n = 367) also showed modest activity with an ORR of 8.0%; BRCA mutation status was not evaluated as a biomarker.34 In a single-institution study, BRCA mutation status, tumor mutational burden (TMB), and HRD were not associated with response to ICB in patients with ovarian cancer although a high fraction of genome altered was associated with improvement in PFS and overall survival.35 A trial combining PARP1 inhibitor niraparib with the PD1 inhibitor pembrolizumab (n = 62) showed an ORR of 18% (5% complete response and 13% partial response36), which did not differ by tumor BRCA deficiency (11 carriers, 18% response). The accompanying correlative study found that having a marker of HRD, mutational signature 3 (n = 11, 33%), a positive immune score (n = 3, 9%), or both (n = 6, 18%) was predictive of response.37 Thus, it remains unclear whether responses were driven primarily by PARPi or checkpoint blockade or the treatment of two subgroups in the population.36,38,39 Although larger phase II and III trials evaluating anti–CTLA-4 and anti-PD1/programmed cell death ligand-1 in combination with targeted therapy are underway,21,40 these results suggest that a subset of BRCA1/2 ovarian cancers may harbor immunosuppressive mechanisms that impede clinical benefits.
Herein, we investigated 86 BRCA1/2-deficient ovarian cancers using genomic data from the Cancer Genome Atlas (TCGA) and performed genomic and histopathologic analyses in ovarian cancers from our institute to determine the genetic and genomic properties associated with intrinsic immunogenicity. Our findings illustrate that transcriptional pathways, loss of PTEN, and genomic signatures of HRD collectively inform intrinsic immunogenicity in BRCA1/2-deficient ovarian cancers and may aid in the evaluation of ICB in clinical trials.
MATERIALS AND METHODS
Materials and methods describing the acquisition and sequencing of Penn tumors, the acquisition of TCGA data, immunohistochemistry, CyTOF analysis, and bioinformatics are described in the Data Supplement.
RESULTS
Genomic and Transcriptomic Properties of BRCA1/2-Deficient Ovarian Cancers
We investigated immunogenicity in ovarian cancers from TCGA, comparing BRCA1/2-deficient with HR-proficient cancers (HR-WT cancers, Data Supplement). Ovarian cancers with mutations in non-BRCA1/2 genes involved in HR6 were excluded from our analysis because of the insufficient sample size for immunogenetic analysis (n = 10 RNAseq). TMB (Methods) was significantly different across BRCA1 versus HR-WT (P = .00032, Kruskal-Wallis) and across BRCA2 versus HR-WT cancers (P = .00043, Kruskal-Wallis, Data Supplement). The number of single-nucleotide nonsynonymous variants and neoantigen load were similar across BRCA1 versus BRCA2 cancers, and BRCA1/2-deficient ovarian cancers had a significantly higher mean number of single-nucleotide nonsynonymous variants (Data Supplement; P = .0036 and P = .0025) and neoantigen loads (Data Supplement; P = .018 and P = .0066, respectively) than HR-WT cancers. BRCA1/2-deficient cancers collectively exhibited a higher HRD score, a metric of genomic instability associated with BRCA1/2 dysfunction,8,13 than HR-WT cancers (Data Supplement; P = 6.74e–05). Notably, neither HRD nor TMB were significantly correlated with sequencing coverage (Data Supplement).
We investigated biologic pathways that may differentiate BRCA1/2-deficient ovarian cancers (Methods). Clustering of gene set variation analysis scores across all cancers found that three recurring biologic functions inclusive of immune, hematologic, and tumor-suppressive (p53 and ATM) pathways were significantly upregulated in cluster c2 (Fig 1A and Data Supplement). We evaluated the relationship between PTEN status and cluster assignment, as PTEN loss is a frequent driving event in BRCA1/2 high grade serous ovarian cancer41 and has been shown to impede immune cell infiltration in other solid tumors.42,43 We confirmed in our cohort that BRCA1/2 mutations were associated with a higher rate of PTEN loss; 17.3% of BRCA1/2 cancers had homozygous PTEN loss versus 7.7% of HR WT tumors (P = .019, Fisher's exact test). PIK3CA amplification was found in 20% of all samples with higher rates in the BRCA1 hypermethylation group at 30% than those with BRCA1/2 somatic and germline mutations (14%) and non-BRCA1/2 (17%; Data Supplement). Using the chi-square test of independence, we found that BRCA1/2-deficient PTEN-WT cancers were significantly over-represented in one cluster (28 of 29 tumors), that is, c2, whereas BRCA1 promoter–hypermethylated and BRCA1/2-deficient cancers with homozygous loss of PTEN (Methods) were significantly over-represented in cluster c1 (14 of 37 tumors, Fig 1B, P = .0016), but not PIK3CA amplification. When comparing overall survival across cluster c1 versus cluster c2 by multivariate Cox analysis, we found significantly worse prognosis in cluster c1 relative to cluster c2 when adjusting for grade, HRD level, ploidy, BRCA mutation, and stage (Fig 1C, P = .0186, hazard ratio = 4.19 [CI, 1.27 to 13.83]). When comparing PFS across clusters, we found a borderline significantly worse prognosis in cluster c1 relative to cluster c2 when adjusting for the same set of covariates (Fig 1C, P = .062, hazard ratio = 3.26 [CI, 0.94 to 11.33]).
FIG 1.
Mutational and transcriptomic features of the Cancer Genome Atlas ovarian cancers with somatic or germline BRCA1/2 alterations: (A) GSVA illustrating significantly different canonical pathways (MSigDb, adjusted P value < .05) across c1 versus c2 GSVA signaling clusters in BRCA1/2 ovarian cancers; (B) chi-square test of independence illustrating frequency of mutational subtypes within hierarchical clusters in BRCA1/2 cancers (P = .0016); and (C) covariate-adjusted (grade, homologous recombination deficiency level, ploidy, stage, and BRCA1/2 mutation) OS (hazard ratio = 4.19 [CI, 1.27 to 13.83]) and PFS (hazard ratio = 3.26 [CI, 0.94 to 11.33]) curves across c1 versus c2 GSVA signaling clusters in BRCA1/2 ovarian cancers. *P < .05, **P < .01, Student's t-test, Cox Proportional Hazards. BRCA1-HM, BRCA1-hypermethylated; BRCA1/2-P, BRCA1/2 PTEN wild-type; BRCA1/2-PL, BRCA1/2 PTEN loss; FS, Frameshift indel; GSVA, gene set variation analysis; OS, overall survival; PFS, progression-free survival; SNV, single-nucleotide variant; WT, wild-type.
Immunogenicity of BRCA1/2-Deficient Ovarian Cancers by PTEN Status and Mechanism of BRCA1/2 Loss of Function
Given the transcriptional clusters observed in Figure 1A, we categorized BRCA1/2-deficient cancers on the basis of their PTEN status and mechanism of BRCA1/2 loss.44 We found no significant differences in TMB or neoantigen load when comparing on the basis of PTEN loss status within either germline or somatic BRCA1/2-deficient ovarian cancers or HR-WT cancers (Data Supplement). However, HRD scores were significantly higher in BRCA1/2-deficient PTEN-loss and BRCA1 promoter–hypermethylated cancers as compared with BRCA1/2-deficient PTEN-WT and HR-WT cancers (Fig 2A; P = .024 and P = .024, respectively). Among the three components of the HRD score (Methods), large scale state transitions (LST) were significantly lower in BRCA1/2-deficient PTEN-WT ovarian cancers when compared with BRCA1/2-deficient PTEN-loss (P = .012) and BRCA1 promoter–hypermethylated (P = .010 for LST) cancers (Fig 2B). The LST scores of BRCA1/2-deficient PTEN-WT cancers were similar to those of HR-WT cancers. Notably, non-telomeric allelic imbalance (NtAI) scores were significantly higher in BRCA1/2-deficient PTEN WT and HR-WT cancers versus BRCA1/2-deficient PTEN-loss cancers (Fig 2B).
FIG 2.
BRCA1/2 ovarian cancer immunogenicity in relation to the mechanism of BRCA1/2 loss of function and PTEN status: (A) HRD score in Penn + TCGA BRCA1/2 and TCGA HR WT (N = 108) ovarian cancers by PTEN status (PTEN loss, n = 17; PTEN WT, n = 69) versus TCGA BRCA1-hm ovarian cancers (n = 11); (B) LST, NtAI, and LOH scores in Penn + TCGA BRCA1/2 and TCGA HR WT (N = 108) ovarian cancers by PTEN status (PTEN loss, n = 17; PTEN WT, n = 69) versus TCGA BRCA1-hm ovarian cancers (n = 11); (C) in TCGA, CD8 T-cell CIind in BRCA1/2 ovarian cancers by PTEN status (PTEN loss, n = 12; PTEN WT, n = 43) versus TCGA BRCA1-hm ovarian cancers (n = 11) and TCGA HR WT (N = 108); (D) immune infiltration inferred by ESTIMATE in BRCA1/2 ovarian cancers by PTEN status (PTEN loss, n = 12; PTEN WT, n = 43) versus TCGA BRCA1-hm ovarian cancers (n = 11) and TCGA HR WT (N = 108); (E) expression of immune-regulatory molecules in BRCA1/2 ovarian cancers by PTEN status (PTEN loss, n = 12; PTEN WT, n = 43) versus TCGA BRCA1-HM ovarian cancers (n = 11); and (F) coefficient weights from SVM analysis of copy number variations illustrating gene importance across TCGA BRCA1/2-P (n = 43) versus TCGA BRCA1/2-PL (n = 12) and BRCA1-HM (n = 11) ovarian cancers. *P < .05, **P < .01, Student's t-test, analysis of variance. BRCA1/2-P, BRCA1/2 PTEN wild-type; BRCA1/2-PL, BRCA1/2 PTEN loss; CIind, cytolytic index; HR, homologous recombination; HRD, homologous recombination deficiency; iEst, immune ESTIMATE; LOH, loss of heterozygosity; LST, large scale state transitions; NtAI, non-telomeric allelic imbalance; SVM, support vector machine; TCGA, the Cancer Genome Atlas; WT, wild-type.
We determined tumor immunogenicity by comparing expression-based immune indices, including cytolytic index (CIind)45 and immune ESTIMATE (iEst) score.46 Neither index differed significantly when comparing across cancers with BRCA1 versus BRCA2 alterations or germline versus somatic BRCA1/2 alterations, or PTEN-loss versus PTEN-WT among HR-WT cancers (Data Supplement). Both indices correlated negatively with tumor purity estimated by copy number variation (CNV; see the Methods; Data Supplement; CIind: R = –0.25, P = .038; iEst: R = –0.38, P = .0013). The LST score correlated negatively with iEst scores and not with CIind (Data Supplement; CIind: R = –0.079, P = .52; iEst: R = –0.29, P = .019), whereas loss of heterozygosity score and NtAi scores did not exhibit any significant associations (data not shown). The HRD score showed a borderline significantly negative association with the iEst score, but not CIind (Data Supplement; CIind: R = –0.077, P = .53; iEst: R = –0.23, P = .067). When stratifying further, we found that BRCA1/2-deficient PTEN-loss and BRCA1 promoter–hypermethylated cancers had lower CIind (P = .034 and P = .042, respectively) and, for the latter group, decreased iEst versus BRCA1/2-deficient PTEN-WT cancers (P = .003; Figs 2C and 2D). Tumor purity was also lower in BRCA1/2-deficient PTEN-WT versus BRCA1/2-deficient PTEN-loss tumors (Data Supplement, P = 9.34e–05). We analyzed the expression of multiple immune-regulatory genes that we curated from the literature26,29,45,47,48 as having important roles with respect to response to ICB. We found significantly higher expression of ADORA2A (P = .033), DOK3 (P = .0026), HAVCR2 (P = .00055), CD28 (P = .0037), CD86 (P = .00091), ICOS (P = .05), and TNFRSF17 (P = .033) in BRCA1/2-deficient PTEN-WT tumors versus BRCA1 promoter–hypermethylated and BRCA1/2 PTEN-loss tumors (Fig 2E), consistent with the hypothesis that elevated expression of immune inhibitors (ADORA2A, DOK3, and HAVCR2) serves to counter-regulate heightened immune activity.25,26 T-cell chemoattractants (CCL5, CXCL9, CXCL10, and CXCL11) coordinately trended toward higher expression in BRCA1/2-deficient PTEN-WT tumors, but not in BRCA1 promoter–hypermethylated or BRCA1/2-deficient PTEN-loss tumors. HLA expression did not have any discernable pattern across the three groups.
We aimed to identify CNVs affecting gene regions involved in immune system function (Reactome, n = 946 immune-related genes) that may distinguish immunologically cold (BRCA1/2-deficient PTEN-loss + BRCA1-hypermethylated) cancers versus hot cancers (BRCA1/2-deficient, PTEN-WT), as focal CNVs affecting genes involved in immune function influence tumor immunophenotypes.45 By an L1-regularized support vector machine,49 we identified 42 genes distinguishing the two groups with an area under the receiver operating characteristic curve of 0.96 (Data Supplement, hg38 genomic locations). We observed deletions in EIF2AK2, NCF2, and KPNA3, among BRCA1/2-deficient PTEN-WT ovarian cancers. Gains in DEFB4A and NLRP1 were more frequently observed in BRCA1/2-deficient PTEN-loss or BRCA1 promoter–hypermethylated ovarian cancers.
Association of PTEN Loss and HRD Score With Immune Infiltration and Cytotoxicity in BRCA1/2-Deficient Penn Ovarian Cancers
To validate the immunologic effects of PTEN loss, we evaluated Penn ovarian cancers associated with germline BRCA1/2 mutations (n = 18, Data Supplement). Multiple markers corresponding to adaptive and innate immune cell function were compared by PTEN status (Methods). BRCA1/2-deficient PTEN-loss cancers (n = 5) were characterized by significantly lower intratumoral CD3+ cells (P = .05), intratumoral CD8+ (P = .012), and intratumoral and stromal FOXP3+ (P = .0087 and P = .037, respectively) immune cells (Figs 3A and 3B), revealing that PTEN loss is associated with T-cell exclusion in BRCA1/2-deficient ovarian cancer. Other immune cell types (CD4, CD20, and CD68) did not demonstrate a statistically significant difference by PTEN status (Data Supplement). Furthermore, BRCA1/2-deficient PTEN-loss cancers were characterized by lower numbers of PRF1 (perforin 1)-positive cells (P = .041, Figs 3C and 3D), suggesting lower antitumor cytolytic activity in this subset. To determine whether the difference in cytolytic activity was due to fewer CD8+ cells or due to lower immune activity on a per-cell basis, we calculated the ratio of PRF1+ to CD8+ cells and found no significant difference (P = .71) across PTEN-WT versus PTEN-loss BRCA1/2-deficient cancers. MATH score, a measure of intratumoral heterogeneity,50 was significantly higher in BRCA1/2-deficient PTEN-loss cancers than BRCA1/2-deficient PTEN-WT cancers (Fig 3E, P = .0042), in agreement with previous work demonstrating that CD8+ T-cell infiltration is negatively associated with ovarian cancers exhibiting greater clonal diversity.51
FIG 3.
Evaluation of tumor infiltrates, antitumor immune activity, and somatic mutation clonality in Penn BRCA1/2 ovarian cancers by PTEN status. Immune cell populations and immune effector molecules as a function of PTEN status in Penn ovarian cancers associated with germline BRCA1/2 mutations. (A) Levels of intratumoral and stromal CD3+, CD8+, and FoxP3+ T cells in BRCA1/2-PL (n = 5) or BRCA1/2-P (n = 13) ovarian cancers. (B) Representative 10× immunohistochemical images of CD3+, CD8+, and FoxP3+ T cells in BRCA1/2 ovarian cancers (the scale bar represents 0.1 mm at 0.01 mm increments). (C) Number of PRF1-positive cells in BRCA1/2-PL (n = 5) or BRCA1/2-P (n = 13) ovarian cancers. (D) Representative images of intratumoral PRF1 expression in BRCA1/2-PL or BRCA1/2-P ovarian cancers (the scale bar represents 0.1 mm at 0.01 mm increments). (E) MATH scores by PTEN status. Error bars, standard error, *P < .05, **P < .01, Student's t-test. BRCA1/2-P, BRCA1/2 PTEN wild-type; BRCA1/2-PL, BRCA1/2 PTEN loss; MATH, Mutant Allele Tumor Heterogeneity.
CyTOF analysis was performed on 11 available Penn tumor digests (n = 8) or ascites samples (n = 3) with BRCA1/2 mutations (2 of 11 with homozygous PTEN loss) to measure the expression of immune-regulatory molecules in CD3+ T-cell subsets in the tumor microenvironment (Methods, Data Supplement). viSNE plots mapped the location of immune subsets such as CD8+ and CD8– T cells (Data Supplement). viSNE analysis52,53 identified dense clusters and higher frequencies of T cells in BRCA1/2-deficient PTEN-WT tumors expressing relatively high amounts of inhibitory immune checkpoints CTLA4, LAG3, and TIGIT, as well as FOXP3+ cells in a representative BRCA1 ovarian cancer, in contrast to comparatively lower overall expression in a BRCA1 PTEN-loss cancer (Fig 4A). The proinflammatory and costimulatory molecules interleukin (IL)-2, interferon gamma, IL-6, and CD28 were also expressed by more T cells at high levels in the PTEN-WT cancer relative to the PTEN-loss cancer (Fig 4B), underscoring the association of PTEN loss with reduced T-cell activation in the tumor microenvironment.
FIG 4.
CyTOF analysis of immunoinhibitory and cytolytic activity in Penn BRCA1/2 ovarian cancers by PTEN status and HRD score: (A) viSNE maps of CD4+ and CD8+ T cells illustrating differences in expression of inhibitory immune checkpoint molecules CTLA4, LAG3, TIGIT, and FOXP3 from a patient with a germline BRCA1/2-PL tumor versus a patient with a BRCA1/2-P; (B) viSNE maps of CD4+ and CD8+ T cells illustrating differences in expression of proinflammatory and costimulatory immune molecules IL2, IFNG, IL6, and CD28 from a patient with a germline BRCA1/2-PL versus a patient with a BRCA1/2-P; and (C) Scatterplots illustrating negative correlations between the HRD score and the expression of inhibitory immune molecules and immune cell subsets (top) and between the HRD score and proinflammatory and immune-activating molecules (bottom) by CyTOF in BRCA1/2 ovarian cancers (n = 11). BRCA1/2-P, BRCA1/2 PTEN wild-type; BRCA1/2-PL, BRCA1/2 PTEN loss; HRD, homologous recombination deficiency; IFNG, interferon gamma; IL, interleukin; r2, Pearson Correlation Coefficient.
We investigated the association of the HRD score with immune cell subsets and immunomodulatory molecules in BRCA1/2-deficient ovarian cancers (Fig 4C). We found negative correlations between the HRD score and the frequency of tumor-associated T cells expressing CTLA4, LAG3, and CD160 immune inhibitory molecules and FOXP3+ T cells, which have been shown to accumulate at sites of cytotoxic T cells to mitigate antitumor immune attack.54,55 Furthermore, proinflammatory cytokines (interferon gamma, IL-2), cytolytic pore-forming molecules (PRF and GZMB), and markers of proliferating or activated T cells (pSTAT5, Ki67, CD25, and GITR) were all negatively correlated with the HRD score (Fig 4C). Despite the negative correlations, the P values are nonsignificant likely because of the sample size.
Notably, the BRCA1/2-deficient PTEN-loss tumors had a higher preponderance of CD103+CD69+CD127– resident memory T cells (Trm) expressing PD-1 relative to the PTEN-WT tumor (Data Supplement). Furthermore, the HRD score is positively associated with CD103+CD69+CD127– Trm cells expressing PD-1 (Data Supplement).56,57 Taken together, these results further illustrate that PTEN loss or high HRD may inform immunologic states in BRCA1/2-deficient ovarian cancers.
DISCUSSION
Our work sheds light on the tumor-immune heterogeneity in ovarian cancers with BRCA1/2 alterations and potentially gives novel insights into the treatment of these cancers with ICB. Similar to other studies of ovarian cancer, we found PIK3CA amplification in 20% of ovarian cancers but did not identify an association with immunogenicity.58 Our results are similar to immunologic studies in other cancers59-64 with PTEN loss and are also consistent with a study of 5,400 ovarian cancers, with 3,244 being high-grade serous, which demonstrated a correlation between cytoplasmic PTEN staining and low CD8+ T cells.63 Notably, in BRCA1/2 breast cancers, we also found that markers of immunogenicity were inversely correlated with the HRD score in both TGCA and local tumor analyses, although likely through distinct underlying genetic mechanisms.65 Furthermore, Davoli et al and Thorrson et al found negative correlations between CNV burden and measures of immunogenicity,51,64-69 consistent with our analysis.
Our findings are also consistent with previous observations in melanoma in which T-cell exclusion is a mechanism of mitigating immune attack in PTEN-loss cancers.69,70 Our data suggest that immune exclusion in PTEN-loss cancers may result from reduced expression of chemokines that play important roles in T-cell recruitment.70 This finding is consistent with our recent finding that a reduction of both tumor intrinsic CCL5 expression and CCL5-driven CXCL9 expression by macrophages promotes TIL desertification and immune blunting, whereas CCL5 and CXCL9 overexpression in the ovarian cancer microenvironment is associated with CD8+ T-cell infiltration.48
Our observations with HRD and immunogenicity highlight that although HRD contributes to an increase in the number of neoantigens, chronic exposure of TILs to their cognate antigens may contribute to a quiescent phenotype in ovarian cancer.71-73 CyTOF analysis showed a higher preponderance of PD1-expressing CD103+CD69+CD127– Trm cells in PTEN-loss or HRD-high cancers. Trm cells have been shown in ovarian cancer to be quiescent,71 and anti-PD1 therapy can rapidly induce tumor cytotoxicity.57,74 Notably, cancers with higher HRD are more intrinsically responsive to DNA-damaging agents,8,13 which can work with ICB to synergistically eliminate solid tumors in mouse models.29,30 Our data suggest that BRCA1/2-deficient PTEN-loss and BRCA1 promoter–hypermethylated cancers may respond to PARPi, whereas ICB may be optimal for patients BRCA1/2-deficient PTEN-WT cancer. Thus, combination therapy may be efficacious, as PARPi and ICB each treat a subgroup of BRCA1/2-deficient ovarian cancers.75
From pathway analysis, we found that BRCA1/2-deficient PTEN-WT ovarian cancers had higher expression of ATM-driven DNA repair pathways, consistent with previous studies demonstrating a prevalent role of PTEN in HR-based genomic repair.76,77 Along with lower prevalence of LSTs in this group, these results indicate that partial retention of DNA repair activity may be less immunosuppressive than full loss. These findings also indicate that BRCA1/2-deficient ovarian cancers may represent another example in which aneuploidy and large CNVs68,78 are important driving forces that regulate immune responses and may supersede TMB as the primary immune influence.47
Although nearly all BRCA1/2-deficient ovarian cancers in our study harbored a TP53 mutation, p53 signaling in BRCA1/2-deficient PTEN-WT cancers was elevated relative to the remaining cancers by gene set variation analysis. TP53 may be a key driver of immune responses in this subset of ovarian cancers, as a previous study found that pharmacologic activation of p53 in the tumor microenvironment enhances CD8-driven immunogenic cell death in mouse models.79 Furthermore, our analyses of somatic CNVs identified a greater prevalence of deletions of EIF2AK2, NCF2, and KPNA3, all of which are either targets of or influence p53 signaling, in the BRCA1/2 PTEN-loss and BRCA1 promoter–hypermethylated group.80-82 In particular, EIF2AK2 is a target of TP53 activity, serving a proapoptotic role for tumor suppression.83 The more prevalent deletion of EIF2AK2 in PTEN-WT ovarian cancers in conjunction with heightened p53 signaling suggests an intricate proinflammatory mechanism of p53 in ovarian cancer with concomitant selection against the tumor-suppressive role of EIF2AK2.
Although the study benefited from inclusion of both TCGA and locally generated data, each sample set had limitations. TGCA data did not include linked immunohistochemistry data examining immune cells, and the Penn data derived from formalin-fixed paraffin-embedded tissues were limited in terms of expression analysis. Neither sample set had whole genome sequencing data, which most accurately characterize TMB and large-scale CNVs. Several findings warrant independent validation, in particular, the CyTOF analysis and association of the expression signatures with outcomes. In addition, a study of BRCA1/2 breast cancer using publicly available TCGA and Wellcome Trust Institute data suggests that PTEN status modulates immunogenicity but, by contrast, found that PTEN loss was associated with a more T-cell–inflamed signature.84 However, PTEN loss also correlates with BRCA1 mutation status and triple-negative breast cancer status, and in their analysis, the latter may account for the enrichment of T-cell–inflamed signatures, which we and many others have found to be relatively more immunogenic than hormone-positive breast cancers.65
Taken together, our study gives novel insights into the genetic events that may contribute to immunosuppression in BRCA1/2-deficient ovarian cancers, defining a subset of immunologically cold tumors. This understanding may help craft more efficacious use of ICB in the clinic.
Michael Feldman
Research Funding: Scopio Inc (Inst)
Daniel J. Powell
Stock and Other Ownership Interests: Atara Biotherapeutics, InsTIL Bio Inc
Consulting or Advisory Role: Neon Therapeutics, Iovance Biotherapeutics, Tmunity Therapeutics Inc, InsTIL Bio Inc, Bellicum Pharmaceuticals
Research Funding: Lilly, Tmunity Therapeutics Inc, Incyte, Monojul, AstraZeneca/MedImmune, InsTIL Bio
Patents, Royalties, Other Intellectual Property: I hold patents in the field of CAR T-cell therapy in oncology and have received royalties related to their licensing to Novartis, I hold patents in the field of CAR T-cell therapy in oncology and have received royalties related to their licensing to Tmunity, and I hold patents in the field of universal immune receptor T-cell therapy in oncology and have received payments related to their licensing to Prescient Therapeutics
Travel, Accommodations, Expenses: Iovance Biotherapeutics
Susan M. Domchek
Honoraria: AstraZeneca, GlaxoSmithKline
Research Funding: AstraZeneca (Inst), Clovis Oncology (Inst)
Open Payments Link: https://openpaymentsdata.cms.gov/physician/917904
Robert H. Vonderheide
Patents, Royalties, Other Intellectual Property: Receives royalties from Children's Hospital Boston for a licensed research-only monoclonal antibody, Inventor on a licensed patient regarding cancer vaccine antigens
No other potential conflicts of interest were reported.
DISCLAIMER
The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
SUPPORT
Supported by the National Institutes of Health (P30 CA016520), Basser Center for BRCA at the University of Pennsylvania (K.L.N., S.M.D., D.J.P., and R.H.V.), Ovarian Cancer Translational Center of Excellence at the University of Pennsylvania (D.J.P.), Breast Cancer Research Foundation (K.L.N., S.M.D., and R.H.V.), Susan G Komen Foundation (S.M.D), Rooney Family Foundation (K.L.N. and S.M.D.), National Institutes of Health (K12-CA076931; K.N.M.), Konner Family Foundation (K.N.M.), the Parker Institute for Cancer Immunotherapy (R.H.V. and K.L.N.), the National Center for Advancing Translational Sciences of the National Institutes of Health (TL1TR001880; A.A.K.), and the National Human Genome Research Institute of the National Institutes of Health (5T32HG009495—02; A.A.K.).
DATA SHARING STATEMENT
The WES data that support this study have been deposited in the National Center for Biotechnology Information (NCBI)'s Sequence Read Archive (SRA, https://www.ncbi.nlm.nih.gov/sra) with BioProject ID PRJNA38804 and can be accessed at http://www.ncbi.nlm.nih.gov/bioproject/388048. The TCGA data are available from the National Cancer Institute's Genome Data Commons (https://gdc.cancer.gov/). The remaining data are available within the article, and its Supplementary Information files are available from the authors upon request.
AUTHOR CONTRIBUTIONS
Conception and design: Adam A. Kraya, Kara N. Maxwell, Michael Feldman, Susan M. Domchek, Robert H. Vonderheide, Katherine L. Nathanson
Financial support: Katherine L. Nathanson
Administrative support: Katherine L. Nathanson
Provision of study materials or patients: Katherine L. Nathanson
Collection and assembly of data: Adam A. Kraya, Monika A. Eiva, Michael Feldman, Anupma Nayak, Daniel J. Powell, Susan M. Domchek, Katherine L. Nathanson
Data analysis and interpretation: All authors
Manuscript writing: All authors
Final approval of manuscript: All authors
Accountable for all aspects of the work: All authors
AUTHORS' DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST
The following represents disclosure information provided by authors of this manuscript. All relationships are considered compensated unless otherwise noted. Relationships are self-held unless noted. I = Immediate Family Member, Inst = My Institution. Relationships may not relate to the subject matter of this manuscript. For more information about ASCO's conflict of interest policy, please refer to www.asco.org/rwc or ascopubs.org/po/author-center.
Open Payments is a public database containing information reported by companies about payments made to US-licensed physicians (Open Payments).
Michael Feldman
Research Funding: Scopio Inc (Inst)
Daniel J. Powell
Stock and Other Ownership Interests: Atara Biotherapeutics, InsTIL Bio Inc
Consulting or Advisory Role: Neon Therapeutics, Iovance Biotherapeutics, Tmunity Therapeutics Inc, InsTIL Bio Inc, Bellicum Pharmaceuticals
Research Funding: Lilly, Tmunity Therapeutics Inc, Incyte, Monojul, AstraZeneca/MedImmune, InsTIL Bio
Patents, Royalties, Other Intellectual Property: I hold patents in the field of CAR T-cell therapy in oncology and have received royalties related to their licensing to Novartis, I hold patents in the field of CAR T-cell therapy in oncology and have received royalties related to their licensing to Tmunity, and I hold patents in the field of universal immune receptor T-cell therapy in oncology and have received payments related to their licensing to Prescient Therapeutics
Travel, Accommodations, Expenses: Iovance Biotherapeutics
Susan M. Domchek
Honoraria: AstraZeneca, GlaxoSmithKline
Research Funding: AstraZeneca (Inst), Clovis Oncology (Inst)
Open Payments Link: https://openpaymentsdata.cms.gov/physician/917904
Robert H. Vonderheide
Patents, Royalties, Other Intellectual Property: Receives royalties from Children's Hospital Boston for a licensed research-only monoclonal antibody, Inventor on a licensed patient regarding cancer vaccine antigens
No other potential conflicts of interest were reported.
REFERENCES
- 1.Maxwell KN, Domchek SM.Cancer treatment according to BRCA1 and BRCA2 mutations Nat Rev Clin Oncol 9520–5282012 [DOI] [PubMed] [Google Scholar]
- 2.Lord CJ, Ashworth A.PARP inhibitors: Synthetic lethality in the clinic Science 3551152–11582017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Alexandrov LB, Kim J, Haradhvala NJ, et al. The repertoire of mutational signatures in human cancer Nature 57894–1012020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Audeh MW, Carmichael J, Penson RT, et al. Oral poly(ADP-ribose) polymerase inhibitor olaparib in patients with BRCA1 or BRCA2 mutations and recurrent ovarian cancer: A proof-of-concept trial Lancet 376245–2512010 [DOI] [PubMed] [Google Scholar]
- 5.Farmer H, McCabe N, Lord CJ, et al. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy Nature 434917–9212005 [DOI] [PubMed] [Google Scholar]
- 6.Konstantinopoulos PA, Ceccaldi R, Shapiro GI, et al. Homologous recombination deficiency: Exploiting the fundamental vulnerability of ovarian cancer Cancer Discov 51137–11542015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Watkins JA, Irshad S, Grigoriadis A, et al. Genomic scars as biomarkers of homologous recombination deficiency and drug response in breast and ovarian cancers. Breast Cancer Res. 2014;16:211. doi: 10.1186/bcr3670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Telli ML, Timms KM, Reid J, et al. Homologous recombination deficiency (HRD) score predicts response to platinum-containing neoadjuvant chemotherapy in patients with triple-negative breast cancer Clin Cancer Res 223764–37732016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Kondrashova O, Nguyen M, Shield-Artin K, et al. Secondary somatic mutations restoring RAD51C and RAD51D associated with acquired resistance to the PARP inhibitor rucaparib in high-grade ovarian carcinoma Cancer Discov 7984–9982017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Jenner ZB, Sood AK, Coleman RL.Evaluation of rucaparib and companion diagnostics in the PARP inhibitor landscape for recurrent ovarian cancer therapy Future Oncol 121439–14562016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Mirza MR, Monk BJ, Herrstedt J, et al. Niraparib maintenance therapy in platinum-sensitive, recurrent ovarian cancer N Engl J Med 3752154–21642016 [DOI] [PubMed] [Google Scholar]
- 12.Moore K, Colombo N, Scambia G, et al. Maintenance olaparib in patients with newly diagnosed advanced ovarian cancer N Engl J Med 3792495–25052018 [DOI] [PubMed] [Google Scholar]
- 13. Maxwell KN, Wubbenhorst B, Wenz BM, et al. BRCA locus-specific loss of heterozygosity in germline BRCA1 and BRCA2 carriers. Nat Commun. 2017;8:319. doi: 10.1038/s41467-017-00388-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Norquist B, Wurz KA, Pennil CC, et al. Secondary somatic mutations restoring BRCA1/2 predict chemotherapy resistance in hereditary ovarian carcinomas J Clin Oncol 293008–30152011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Lord CJ, Ashworth A.Mechanisms of resistance to therapies targeting BRCA-mutant cancers Nat Med 191381–13882013 [DOI] [PubMed] [Google Scholar]
- 16.Patch AM, Christie EL, Etemadmoghadam D, et al. Whole-genome characterization of chemoresistant ovarian cancer Nature 521489–4942015 [DOI] [PubMed] [Google Scholar]
- 17.Yazinski SA, Comaills V, Buisson R, et al. ATR inhibition disrupts rewired homologous recombination and fork protection pathways in PARP inhibitor-resistant BRCA-deficient cancer cells Genes Dev 31318–3322017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Jaspers JE, Kersbergen A, Boon U, et al. Loss of 53BP1 causes PARP inhibitor resistance in Brca1-mutated mouse mammary tumors Cancer Discov 368–812013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Darb-Esfahani S, Kunze CA, Kulbe H, et al. Prognostic impact of programmed cell death-1 (PD-1) and PD-ligand 1 (PD-L1) expression in cancer cells and tumor-infiltrating lymphocytes in ovarian high grade serous carcinoma Oncotarget 71486–14992016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Clarke B, Tinker AV, Lee CH, et al. Intraepithelial T cells and prognosis in ovarian carcinoma: Novel associations with stage, tumor type, and BRCA1 loss Mod Pathol 22393–4022009 [DOI] [PubMed] [Google Scholar]
- 21. Gaillard SL, Secord AA, Monk B. The role of immune checkpoint inhibition in the treatment of ovarian cancer. Gynecol Oncol Res Pract. 2016;3:11. doi: 10.1186/s40661-016-0033-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Strickland KC, Howitt BE, Shukla SA, et al. 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 Oncotarget 713587–135982016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Soslow RA, Han G, Park KJ, et al. Morphologic patterns associated with BRCA1 and BRCA2 genotype in ovarian carcinoma Mod Pathol 25625–6362012 [DOI] [PubMed] [Google Scholar]
- 24.Zhang L, Conejo-Garcia JR, Katsaros D, et al. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer N Engl J Med 348203–2132003 [DOI] [PubMed] [Google Scholar]
- 25.Sato E, Olson SH, Ahn J, 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 USA 10218538–185432005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Llosa NJ, Cruise M, Tam A, et al. The vigorous immune microenvironment of microsatellite instable colon cancer is balanced by multiple counter-inhibitory checkpoints Cancer Discov 543–512015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Hussein YR, Weigelt B, Levine DA, et al. Clinicopathological analysis of endometrial carcinomas harboring somatic POLE exonuclease domain mutations Mod Pathol 28505–5142015 [DOI] [PubMed] [Google Scholar]
- 28.Kaufman B, Shapira-Frommer R, Schmutzler RK, et al. Olaparib monotherapy in patients with advanced cancer and a germline BRCA1/2 mutation J Clin Oncol 33244–2502015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Huang J, Wang L, Cong Z, et al. The PARP1 inhibitor BMN 673 exhibits immunoregulatory effects in a Brca1(-/-) murine model of ovarian cancer Biochem Biophys Res Commun 463551–5562015 [DOI] [PubMed] [Google Scholar]
- 30.Higuchi T, Flies DB, Marjon NA, et al. CTLA-4 blockade synergizes therapeutically with PARP inhibition in BRCA1-deficient ovarian cancer Cancer Immunol Res 31257–12682015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Chodon T, Lugade AA, Battaglia S, et al. Emerging role and future directions of immunotherapy in advanced ovarian cancer Hematol Oncol Clin North Am 321025–10392018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Disis ML, Taylor MH, Kelly K, et al. Efficacy and safety of avelumab for patients with recurrent or refractory ovarian cancer: Phase 1b results from the JAVELIN solid tumor trial JAMA Oncol 5393–4012019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Disis ML, Patel MR, Pant S, et al. Avelumab (MSB0010718C; anti-PD-L1) in patients with recurrent/refractory ovarian cancer from the JAVELIN Solid Tumor phase Ib trial: Safety and clinical activity. J Clin Oncol. 2016;15 abstr 5533. [Google Scholar]
- 34.Matulonis UA, Shapira-Frommer R, Santin AD, et al. Antitumor activity and safety of pembrolizumab in patients with advanced recurrent ovarian cancer: Results from the phase II KEYNOTE-100 study Ann Oncol 301080–10872019 [DOI] [PubMed] [Google Scholar]
- 35. Liu YL, Selenica P, Zhou Q, et al. BRCA mutations, homologous DNA repair deficiency, tumor mutational burden, and response to immune checkpoint inhibition in recurrent ovarian cancer. JCO Precis Oncol. doi: 10.1200/PO.20.00069. 10.1200/PO.20.00069 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Konstantinopoulos PA, Waggoner S, Vidal GA, et al. Single-arm phases 1 and 2 trial of niraparib in combination with pembrolizumab in patients with recurrent platinum-resistant ovarian carcinoma JAMA Oncol 51141–11492019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Färkkilä A, Gulhan DC, Casado J, et al. Immunogenomic profiling determines responses to combined PARP and PD-1 inhibition in ovarian cancer. Nat Commun. 2020;11:1459. doi: 10.1038/s41467-020-15315-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Kristeleit R, Shapiro GI, Burris HA, et al. A phase I-II study of the oral PARP inhibitor rucaparib in patients with germline BRCA1/2-mutated ovarian carcinoma or other solid tumors Clin Cancer Res 234095–41062017 [DOI] [PubMed] [Google Scholar]
- 39.Oza AM, Tinker AV, Oaknin A, et al. Antitumor activity and safety of the PARP inhibitor rucaparib in patients with high-grade ovarian carcinoma and a germline or somatic BRCA1 or BRCA2 mutation: Integrated analysis of data from Study 10 and ARIEL2 Gynecol Oncol 147267–2752017 [DOI] [PubMed] [Google Scholar]
- 40.Castellano T, Moore KN, Holman LL.An overview of immune checkpoint inhibitors in gynecologic cancers Clin Ther 40372–3882018 [DOI] [PubMed] [Google Scholar]
- 41. Merino DM, McShane LM, Fabrizio D, et al. Establishing guidelines to harmonize tumor mutational burden (TMB): In silico assessment of variation in TMB quantification across diagnostic platforms: phase I of the Friends of Cancer Research TMB Harmonization Project. J Immunother Cancer. 2020;8:e000147. doi: 10.1136/jitc-2019-000147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Martins FC, Santiago ID, Trinh A, et al. Combined image and genomic analysis of high-grade serous ovarian cancer reveals PTEN loss as a common driver event and prognostic classifier. Genome Biol. 2014;15:526. doi: 10.1186/s13059-014-0526-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Lin Z, Huang L, Li SL, et al. PTEN loss correlates with T cell exclusion across human cancers. BMC Cancer. 2021;21:429. doi: 10.1186/s12885-021-08114-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Chiang JW, Karlan BY, Cass L, et al. BRCA1 promoter methylation predicts adverse ovarian cancer prognosis Gynecol Oncol 101403–4102006 [DOI] [PubMed] [Google Scholar]
- 45.Rooney MS, Shukla SA, Wu CJ, et al. Molecular and genetic properties of tumors associated with local immune cytolytic activity Cell 16048–612015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Yoshihara K, Shahmoradgoli M, Martínez E, et al. Inferring tumour purity and stromal and immune cell admixture from expression data. Nat Commun. 2013;4:2612. doi: 10.1038/ncomms3612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Balli D, Rech AJ, Stanger BZ, et al. Immune cytolytic activity stratifies molecular subsets of human pancreatic cancer Clin Cancer Res 233129–31382017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Dangaj D, Bruand M, Grimm AJ, et al. Cooperation between constitutive and inducible chemokines enables T cell engraftment and immune attack in solid tumors Cancer Cell 35885–900.e102019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Huang S, Cai N, Pacheco PP, et al. Applications of support vector machine (SVM) learning in cancer genomics Cancer Genomics Proteomics 1541–512018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Mroz EA, Rocco JW.MATH, a novel measure of intratumor genetic heterogeneity, is high in poor-outcome classes of head and neck squamous cell carcinoma Oral Oncol 49211–2152013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Zhang AW, McPherson A, Milne K, et al. Interfaces of malignant and immunologic clonal dynamics in ovarian cancer Cell 1731755–1769.e222018 [DOI] [PubMed] [Google Scholar]
- 52.Amir ED, Davis KL, Tadmor MD, et al. viSNE enables visualization of high dimensional single-cell data and reveals phenotypic heterogeneity of leukemia Nat Biotechnol 31545–5522013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.van der Maaten L.Accelerating t-SNE using tree-based algorithms J Mach Learn Res 153221–32452014 [Google Scholar]
- 54.Le DT, Ladle BH, Lee T, et al. CD8(+) Foxp3(+) tumor infiltrating lymphocytes accumulate in the context of an effective anti-tumor response Int J Cancer 129636–6472011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Jang JE, Hajdu CH, Liot C, et al. Crosstalk between regulatory T cells and tumor-associated dendritic cells negates anti-tumor immunity in pancreatic cancer Cell Rep 20558–5712017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Webb JR, Milne K, Watson P, et al. Tumor-infiltrating lymphocytes expressing the tissue resident memory marker CD103 are associated with increased survival in high-grade serous ovarian cancer Clin Cancer Res 20434–4442014 [DOI] [PubMed] [Google Scholar]
- 57. Blanc C, Hans S, Tran T, et al. Targeting resident memory T cells for cancer immunotherapy. Front Immunol. 2018;9:1722. doi: 10.3389/fimmu.2018.01722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Press JZ, De Luca A, Boyd N, et al. Ovarian carcinomas with genetic and epigenetic BRCA1 loss have distinct molecular abnormalities. BMC Cancer. 2008;8:17. doi: 10.1186/1471-2407-8-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Peng W, Chen JQ, Liu C, et al. Loss of PTEN promotes resistance to T cell-mediated immunotherapy Cancer Discov 6202–2162016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.George S, Miao D, Demetri GD, et al. Loss of PTEN is associated with resistance to anti-PD-1 checkpoint blockade therapy in metastatic uterine leiomyosarcoma Immunity 46197–2042017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Garcia AJ, Ruscetti M, Arenzana TL, et al. Pten null prostate epithelium promotes localized myeloid-derived suppressor cell expansion and immune suppression during tumor initiation and progression Mol Cell Biol 342017–20282014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Toso A, Revandkar A, Di Mitri D, et al. Enhancing chemotherapy efficacy in Pten-deficient prostate tumors by activating the senescence-associated antitumor immunity Cell Rep 975–892014 [DOI] [PubMed] [Google Scholar]
- 63.Martins FC, Couturier DL, Paterson A, et al. Clinical and pathological associations of PTEN expression in ovarian cancer: A multicentre study from the ovarian tumour tissue analysis consortium Br J Cancer 123793–8022020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Thorsson V, Gibbs DL, Brown SD, et al. The immune landscape of cancer Immunity 48812–830.e142018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Kraya AA, Maxwell KN, Wubbenhorst B, et al. Genomic signatures predict the immunogenicity of BRCA-deficient breast cancer Clin Cancer Res 254363–43742019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Davoli T, Uno H, Wooten EC, et al. Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Science. 2017;355:6322. doi: 10.1126/science.aaf8399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Buccitelli C, Salgueiro L, Rowald K, et al. Pan-cancer analysis distinguishes transcriptional changes of aneuploidy from proliferation Genome Res 27501–5112017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Taylor AM, Shih J, Ha G, et al. Genomic and functional approaches to understanding cancer aneuploidy Cancer Cell 33676–689.e32018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Jerby-Arnon L, Shah P, Cuoco MS, et al. A cancer cell program promotes T cell exclusion and resistance to checkpoint blockade Cell 175984–997.e242018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Gregor CE, Foeng J, Comerford I, et al. Chemokine-driven CD4(+) T cell homing: New concepts and recent advances Adv Immunol 135119–1812017 [DOI] [PubMed] [Google Scholar]
- 71.Webb JR, Milne K, Nelson BH.PD-1 and CD103 are widely coexpressed on prognostically favorable intraepithelial CD8 T cells in human ovarian cancer Cancer Immunol Res 3926–9352015 [DOI] [PubMed] [Google Scholar]
- 72.Matsuzaki J, Gnjatic S, Mhawech-Fauceglia P, et al. Tumor-infiltrating NY-ESO-1-specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer Proc Natl Acad Sci USA 1077875–78802010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Bakhoum SF, Cantley LC.The multifaceted role of chromosomal instability in cancer and its microenvironment Cell 1741347–13602018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Enamorado M, Iborra S, Priego E, et al. Enhanced anti-tumour immunity requires the interplay between resident and circulating memory CD8(+) T cells. Nat Comm. 2017;8:16073. doi: 10.1038/ncomms16073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Mehta AK, Cheney EM, Hartl CA, et al. Targeting immunosuppressive macrophages overcomes PARP inhibitor resistance in BRCA1-associated triple-negative breast cancer Nat Cancer 266–822021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Bassi C, Ho J, Srikumar T, et al. Nuclear PTEN controls DNA repair and sensitivity to genotoxic stress Science 341395–3992013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Mansour WY, Tennstedt P, Volquardsen J, et al. Loss of PTEN-assisted G2/M checkpoint impedes homologous recombination repair and enhances radio-curability and PARP inhibitor treatment response in prostate cancer. Sci Rep. 2018;8:3947. doi: 10.1038/s41598-018-22289-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Li J, Byrne KT, Yan F, et al. Tumor cell-intrinsic factors underlie heterogeneity of immune cell infiltration and response to immunotherapy Immunity 49178–193.e72018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Guo G, Yu M, Xiao W, et al. Local activation of p53 in the tumor microenvironment overcomes immune suppression and enhances antitumor immunity Cancer Res 772292–23052017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Dierckx T, Khouri R, Menezes SM, et al. IFN-beta induces greater antiproliferative and proapoptotic effects and increased p53 signaling compared with IFN-alpha in PBMCs of Adult T-cell Leukemia/Lymphoma patients. Blood Cancer J. 2017;7:e519. doi: 10.1038/bcj.2016.126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Chen KS, Kwon WS, Kim J, et al. A novel TP53-KPNA3 translocation defines a de novo treatment-resistant clone in osteosarcoma. Cold Spring Harb Mol Case Stud. 2016;2:a000992. doi: 10.1101/mcs.a000992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Italiano D, Lena AM, Melino G, et al. Identification of NCF2/p67phox as a novel p53 target gene Cell Cycle 114589–45962012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Yoon CH, Lee ES, Lim DS, et al. PKR, a p53 target gene, plays a crucial role in the tumor-suppressor function of p53 Proc Natl Acad Sci USA 1067852–78572009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Wen WX, Leong CO. Association of BRCA1- and BRCA2-deficiency with mutation burden, expression of PD-L1/PD-1, immune infiltrates, and T cell-inflamed signature in breast cancer. PLoS One. 2019;14:e0215381. doi: 10.1371/journal.pone.0215381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Spurdle AB, Healey S, Devereau A, et al. ENIGMA—Evidence-based network for the interpretation of germline mutant alleles: An international initiative to evaluate risk and clinical significance associated with sequence variation in BRCA1 and BRCA2 genes Hum Mutat 332–72012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Favero F, Joshi T, Marquard AM, et al. Sequenza: Allele-specific copy number and mutation profiles from tumor sequencing data Ann Oncol 2664–702015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Szolek A, Schubert B, Mohr C, et al. OptiType: Precision HLA typing from next-generation sequencing data Bioinformatics 303310–33162014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Karosiene E, Lundegaard C, Lund O, et al. NetMHCcons: A consensus method for the major histocompatibility complex class I predictions Immunogenetics 64177–1862012 [DOI] [PubMed] [Google Scholar]
- 89. Hänzelmann S, Castelo R, Guinney J. GSVA: Gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics. 2013;14:7. doi: 10.1186/1471-2105-14-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Liberzon A, Birger C, Thorvaldsdóttir H, et al. The Molecular Signatures Database (MSigDB) hallmark gene set collection Cell Syst 1417–4252015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Ritchie ME, Phipson B, Wu D, et al. Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43:e47. doi: 10.1093/nar/gkv007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Gu Z, Eils R, Schlesner M.Complex heatmaps reveal patterns and correlations in multidimensional genomic data Bioinformatics 322847–28492016 [DOI] [PubMed] [Google Scholar]
- 93.Therneau T.A Package for Survival Analysis in R. 2021. v3.2-13. https://CRAN.R-project.org/package=survival
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
The WES data that support this study have been deposited in the National Center for Biotechnology Information (NCBI)'s Sequence Read Archive (SRA, https://www.ncbi.nlm.nih.gov/sra) with BioProject ID PRJNA38804 and can be accessed at http://www.ncbi.nlm.nih.gov/bioproject/388048. The TCGA data are available from the National Cancer Institute's Genome Data Commons (https://gdc.cancer.gov/). The remaining data are available within the article, and its Supplementary Information files are available from the authors upon request.