Purpose:

Tumors of germline BRCA1/2 mutated carriers show homologous recombination (HR) deficiency (HRD), resulting in impaired DNA double-strand break (DSB) repair and high sensitivity to PARP inhibitors. Although this therapy is expected to be effective beyond germline BRCA1/2 mutated carriers, a robust validated test to detect HRD tumors is lacking. In this study, we therefore evaluated a functional HR assay exploiting the formation of RAD51 foci in proliferating cells after ex vivo irradiation of fresh breast cancer tissue: the recombination REpair CAPacity (RECAP) test.

Experimental Design:

Fresh samples of 170 primary breast cancer were analyzed using the RECAP test. The molecular explanation for the HRD phenotype was investigated by exploring BRCA deficiencies, mutational signatures, tumor-infiltrating lymphocytes (TIL), and microsatellite instability (MSI).

Results:

RECAP was completed successfully in 125 of 170 samples (74%). Twenty-four tumors showed HRD (19%), whereas six tumors were HR intermediate (HRi; 5%). HRD was explained by BRCA deficiencies (mutations, promoter hypermethylation, deletions) in 16 cases, whereas seven HRD tumors were non-BRCA related. HRD tumors showed an increased incidence of high TIL counts (P = 0.023) compared with HR proficient (HRP) tumors and MSI was more frequently observed in the HRD group (2/20, 10%) than expected in breast cancer (1%; P = 0.017).

Conclusions:

RECAP is a robust functional HR assay detecting both BRCA1/2-deficient and BRCA1/2-proficient HRD tumors. Functional assessment of HR in a pseudo-diagnostic setting is achievable and produces robust and interpretable results.

Translational Relevance

The functional RECAP test assesses HR capacity in fresh tissue samples. This is a very accurate method for diagnosing HRD tumors, because the read-out is a dynamic process rather than a static genomic status. The main clinical implication of this study is that functional assessment of HR in a pseudo-diagnostic setting is achievable and produces robust and interpretable results. Selection of tumors based on the HRD phenotype, instead of germline BRCA mutations, can identify 50% more HRD tumors. Breast cancers with HRD phenotype showed an increased incidence of high TILs and microsatellite instability (MSI). This observation provides a basis to study whether these specific subgroups of patients with breast cancer would benefit not only from PARP inhibitors (PARPi) but also immunotherapy. Clinical trials have been initiated to evaluate the predictive value of the RECAP test for in vivo response to PARPi.

Breast cancer is the most common malignancy in women with the second highest cancer-related mortality rate (1). Approximately 3% of all breast cancer cases are due to germline mutations in BRCA1/2 (2), and in triple-negative breast cancers (TNBC) this percentage is even 10% to 20% (3). The BRCA proteins play an important role in the homologous recombination (HR) pathway, the error-free DNA double-strand break (DSB) repair pathway that operates during the S- and G2-phase of the cell cycle. HR deficiency (HRD) leading to impaired DNA DSB repair is frequently caused by, but not limited to, defects in BRCA1/2 (4).

Therapies specifically targeting tumor cells with impaired HR capacity are PARP inhibitors (PARPi), as well as classical chemotherapies such as platinum-derivates and alkylating agents (5). PARPi causes persistence of single-strand DNA breaks (SSB) by trapping PARP1 on DNA, whereas platinum-derivates cause DNA interstrand crosslinks. Both types of lesions result in replication fork stalling and/or collapse, frequently leading to DSBs that need HR for their repair (5). The targeted approach of PARPi kills tumor cells lacking HR, whereas normal cells remain unharmed, due to their normal DSB repair capacity, a phenomenon often referred to as synthetic lethality. Recently, FDA approval was granted for the use of Olaparib in germline BRCA mutated breast cancer based on the results of the Olympiad trial (6).

Although evidence is emerging that the use of PARPi could be extended beyond germline BRCA1/2 mutated cancers to sporadic cancers with BRCA-like features, a gold standard test for predicting response to treatments targeting HR is not yet available (7). Several different HRD tests exist, mostly based on genomic patterns or transcriptional predictors of BRCAness (8–12).

These genomic tests measure the accumulation of mutations and chromosomal aberrations over time, but not necessarily reflect the real-time HR status. Beyond mutational status, several other factors influence tumor behavior and therapy response, such as epigenetic changes and the microenvironment of the tumor cells. Independent of the underlying cause, the downstream effect of HR impairment (phenotype) can be assessed functionally. A functional diagnostic assay therefore has the potential for more precisely detecting patients who may benefit from PARPi than genomic assays.

A functional HRD assay was first described by Graeser et al., assessing RAD51 focus formation, a marker of HR competence, in tumor biopsies obtained 24 hours after in vivo anthracycline treatment (13). This provided the first evidence that RAD51 focus formation can serve as a predictive biomarker. To enhance clinical utility of this biomarker, test outcomes should be available before start of treatment. Therefore, we developed the homologous recombination REpair CAPacity (RECAP) test exploiting the formation of RAD51 foci in proliferating cells after ex vivo irradiation of fresh breast cancer tissue, providing a real-time HR status of the tumor (14). The aim of this study was to validate the RECAP test in an extensive cohort of primary breast cancers and provide evidence that this functional test is achievable in a pseudo-clinical setting. Additionally, thorough molecular characterization of the HRD phenotype is performed, proving that HRD tumors encompass more than only BRCA deficiencies.

Primary breast cancer specimens

Residual fresh breast cancer tissue was prospectively collected from lumpectomy of the breast or mastectomy specimens in the Erasmus MC Cancer Institute, Haven hospital, and Maasstad hospital in Rotterdam, the Netherlands, between 1 October 2011 and 1 September 2016. The first 41 patients were also included in our previous cohort (14). After macroscopic evaluation of the surgical specimen by trained pathologists, residual tumor tissue was collected for our research purposes according to the “Code of proper secondary use of human tissue in the Netherlands” established by the Dutch Federation of Medical Scientific Societies and approved by the local Medical Ethical committees (MEC-11-098). Patients who had objected to secondary use of residual tumor material for research purposes were not included in this study. Patients with ductal carcinoma in situ (DCIS) only or patients receiving neo-adjuvant chemotherapy were excluded.

RECAP test

Obtained tissue samples were immediately transferred into customized breast tissue culture medium, as described in Naipal et al. (14). Processing of samples was performed within 4 hours after the tissue was resected. Microscopic analysis of hematoxylin and eosin (HE) stained sections was performed to determine presence of invasive carcinoma. The RECAP test, a functional assay exploiting the formation of RAD51 foci in proliferating cells after ex vivo irradiation of fresh breast cancer tissue, was performed and results were analyzed as described previously (14). In brief, presence of RAD51 foci was determined in S–G2 cells only, which stain positive for Geminin. At least 30 Geminin expressing cells were counted per tumor sample. A cell was considered RAD51 positive when at least five RAD51 foci could be detected. Based on previous experiments with patient-derived xenograft (PDX) models with known BRCA status, tumors were classified as HR proficient (HRP), HR deficient (HRD), or intermediate (HRi) when more than 50%, less than 20% or between 20% and 50% of geminin positive cells showed ≥5 RAD51 foci, respectively.

Workflow of molecular characterization of the HRD phenotype

To unravel the possible molecular mechanism underlying the HRD phenotype, several molecular tests were performed retrospectively (Fig. 2; Supplementary Fig. S1). As no DNA could be obtained for one HRD sample, we conducted the analyses for 23 HRD and six HRi samples. First, BRCA sequencing and BRCA1 promoter methylation analysis was performed in HRD and HRi samples, as well as in all TNBC (n = 5), ER/PR− HER2+ (n = 2), and 21 ER/PR+ HRP tumors (total n = 28; Supplementary Fig. S1). The HRD and HRi tumors without molecular explanation for their phenotype were subjected to BRCA1 and BRCA2 MLPA analysis to identify large genomic rearrangements (LGR), as LGRs are not usually identified by targeted sequencing. In addition to this targeted approach, morphologic examination of TILs and whole exome sequencing (WES) was performed on a selection of tumors to further explore molecular aspects connected to the HRD phenotype.

DNA isolation

Isolation of DNA from 30 μm fresh frozen tissue section samples was performed using the NucleoSpin Tissue Kit (Macherey-Nagel) according to the manufacturer's instructions. Quantity and quality checks of isolated DNA were performed using the MultiNA microchip electrophoresis system (Shimadzu's Hertogenbosch), Nanodrop 2000-v.1 (Thermo Fisher Scientific), and Qubit (Thermo Fisher Scientific).

BRCA1/2 analyses

Ion semiconductor sequencing on the Ion Torrent Personal S5XL was performed according to manufacturer's instructions (Thermo Fisher Scientific). Adapter-ligated libraries were constructed using the AmpliSeq Library Kit 2.0 with amplicons designed targeting BRCA1/2 and TP53. Generation of sequence reads, trimming adapter sequences, filtering, and removal of poor signal-profile reads was performed via the Ion Torrent platform-specific pipeline software Torrent Suite v5.2.2. Initial variant calling was performed by comparison to the reference genome hg19 (build 37) using the “Torrent Variant Caller v5.2.0.34″ plug-in from the Torrent Suite Software. All BRCA2 variants were validated by Sanger sequencing and pathogenicity was evaluated using interactive Biosoftware Alamut Visual v.2.7.2. BRCA1 promoter methylation was analyzed as previously described (15). Multiplex ligation-dependent probe amplification (MLPA) analysis of BRCA1 and BRCA2 was undertaken to identify large rearrangements using the SALSA MLPA Kit P002B, and for confirmation of observed abnormalities, the SALSA MLPA Kit P087 was used (MRC Holland). Analyses were performed according to the manufacturer's instruction; products were run on an ABI automated sequencer (ABI 3730XL), and the data were analyzed by Genemarker version 2.7.0 (Softgenetics).

In situ detection of BRCA1 RNA

In situ detection of BRCA1 mRNA was performed using RNAScope (Advanced Cell Diagnostics) on the automated Ventana Discovery Ultra system (Ventana Medical Systems). BRCA1 and positive control peptidylprolyl isomerase B (PPIB) probes (product codes: 485479 and 313909) were purchased from the same company. RNAscope analysis was performed according to manufacturer's instructions using the reagent kit (VS Reagent Kit 320600; Advanced Cell Diagnostics) on proteinase K (0.1%, 5 min at 37 °C)-treated paraffin sections (4 μm).

Exome sequencing

DNA libraries for Illumina sequencing were generated using standard protocols (Illumina) and subsequently sequenced in an Illumina HiSeq 2500 system by GATC-biotech. Exome-targeting was performed using the Sureselect v5 (v6 for tumors M077, M209, M211) methods using standard protocols (Agilent Technologies). DNA libraries were whole-exome sequenced (2 × 125bp) using the HiSeq v4 paired-end sequencing protocol to a minimum depth base coverage of 90× for tumor samples and 60× for matched normal. Sequence reads were mapped against human reference genome GRCh37 using Burrows–Wheeler Aligner (v0.7.12) with default settings (16). Sequence reads originating from multiple lanes were merged after alignment using Samtools (v1.5) prior to further analysis (17). Sequence duplicates were marked using PicardTools (v1.129; ref. 18). Somatic variant calling was performed by Mutect2 (v3.7) using a matched-normal design while utilizing the dbSNP (v149, hg19) and COSMIC (v80, hg19) databases and using default settings (19–21). Variant annotation was performed by ANNOVAR (22). Heuristic filtering removed variants not passing all standard Mutect2 post-calling filters. Sequence data have been deposited at the European Genome-phenome Archive (EGA, http://www.ebi.ac.uk/ega/), which is hosted by the EBI, under accession number EGAD00001003929.

Mutational signatures

For each somatic variant, its trinucleotide context was derived from the human reference genome GRCh37 and enumerated into a mutational spectrum matrix Mij (i = 96; number of trinucleotide contexts; j = number of samples) using the MutationalPatterns R package (v1.4.0) in the R statistical platform (23). Multi-allelic and InDel variants were not included in this analysis. The 30 consensus mutational signatures, as established by Alexandrov and colleagues, (matrix Sij; i = 96; number of trinucleotide motifs; j = number of signatures) were downloaded from COSMIC (as visited on 8-11-2017; ref. 24). Per sample, a constrained linear combination of the 30 validated mutational signatures was constructed, which reconstructs the sample-specific mutational spectrum, using nonnegative least squares regression implemented in the R package pracma (v1.9.3). Signatures with lower relative contribution than 3% were summarized into a “Filtered” category.

MSI analysis, MMR protein IHC, and MLH1 promotor methylation assay

These analyses were performed as previously described (25).

Tumor-infiltrating lymphocytes

Tumor-infiltrating lymphocytes (TIL) were scored on HE stained sections, according to the consensus by the International TILs Working Group 2014 (26).

Statistical analysis

Statistical analyses were all two-sided and performed using IBM SPSS statistics v21. Significance was calculated by Fisher exact test for categorical data, by Mann–Whitney test for continuous data, and by exact binomial test for the incidence of MSI. P-values of <0.05 were considered significant.

Ex vivo functional RECAP test

A total of 170 samples were subjected to the RECAP test (Supplementary Fig. S1; ref. 14). In 125 of 170 (74%) primary breast cancer tissues RECAP was completed successfully (Fig. 1). In all cases, the reason for failure (n = 45) was lack of proliferating tumor cells. No differences in clinicopathologic characteristics between tumors that yielded successful versus nonsuccessful tests were observed (Supplementary Table S1). The first 41 patients were also included in our previous cohort. Here, we show that execution of a functional assay in a pseudo-diagnostic setting is achievable and validate the findings from the earlier cohort (11% HRD in cohort 1 vs. 19% HRD in cohort 2, P = 0.339). In total, we identified 95 (76%) HRP, 24 (19%) HRD, and 6 (5%) HRi samples (Fig. 1). Both HRi and HRD tumors were more frequently TN (P < 0.001) and Bloom and Richardson (B&R) grade 3 (P < 0.001) than HRP tumors. Also, HRD tumors had a larger size (P = 0.050) and were never B&R grade 1 (Supplementary Table S2).

Figure 1.

RECAP test results: 19% of primary breast cancers showed HRD. A, Schematic representation of the primary breast cancers obtained for RECAP testing. B, Percentage of RAD51 foci positive tumor cells among geminin-expressing nuclei in the 125 successful tests. A total of 95 breast cancer samples were HRP (>50% Geminin positive cells with RAD51 foci), 24 were HRD (<20% Geminin positive cells with RAD51 foci), and six samples were HRi (>20%/<50% Geminin positive cells with RAD51 foci). Black dots indicate TNBCs.

Figure 1.

RECAP test results: 19% of primary breast cancers showed HRD. A, Schematic representation of the primary breast cancers obtained for RECAP testing. B, Percentage of RAD51 foci positive tumor cells among geminin-expressing nuclei in the 125 successful tests. A total of 95 breast cancer samples were HRP (>50% Geminin positive cells with RAD51 foci), 24 were HRD (<20% Geminin positive cells with RAD51 foci), and six samples were HRi (>20%/<50% Geminin positive cells with RAD51 foci). Black dots indicate TNBCs.

Close modal

Identification of BRCA defects in HRD breast cancers

Pathogenic BRCA2 mutations were found in six (6/23 = 26%) HRD samples, but not in the tested HRP samples (Table 1). In six tumors (two HRD, one HRi, and three HRP), BRCA2 variants were detected that were classified as benign. We did not identify any BRCA1 point mutations. Next, BRCA1 promoter hypermethylation was detected in six (6/23 = 26%) HRD tumors and in one HRi tumor, but not in tested HRP samples (Table 1). Interestingly, all six HRD tumors displaying BRCA1 promoter hypermethylation were TNBCs (Table 1; Supplementary Table S3). Vice versa, of the six HRD tumors harboring a pathogenic BRCA2 mutation, five showed hormone receptor positivity.

Table 1.

Overview of samples with BRCA defects

BRCA2 mutationBRCA1 promotor hypermethylation (score)BRCA1 mRNA RNAscopeBRCA LGRsTumor %RECAP status
SampleMutationPathogenicity
M057 c.517G>C p.G173R Pathogenic $    >50% Negative (HRD) 
P002 c.7617+1G>T Pathogenic    60% Negative (HRD) 
M096 c.7285G>T p.E2429X Pathogenic $    50–70% Negative (HRD) 
M188 c.3846_3847del p.T1282fs Pathogenic    Macrodissected (FFPE) Negative (HRD) 
M231 c.755_758del p.D252fs Pathogenic    65% Negative (HRD) 
M275 c.3269delT p.M1090fs Pathogenic    33% Negative (HRD) 
M213 c.1708A>C p.N570H Benign    >50% Positive (HRP) 
M114 c.6829T>C p.L2277L Benign    40% Positive (HRP) 
P9 c.3445A>G p.M1149V Benign    50% Positive (HRP) 
M211 c.5054C>T p.S1685L Benign − (0.02) Positive  >50 Negative (HRD) 
M106 c.6347A>G p.H2116R Benign − (0.01) Positive  50–70 Negative (HRD) 
P001   + (NA) Negative  — Negative (HRD) 
M028   + (NA) Negative  — Negative (HRD) 
M119   + (0.56) Negative  50–70 Negative (HRD) 
M131   + (0.82) Heterogeneous  >70 Negative (HRD) 
M182   + (0.45) Negative  >50 Negative (HRD) 
M277   + (0.24) Heterogeneous  20 Negative (HRD) 
M141   + (0.29) Negative  50 Intermediate (HRi) 
M094   − (0.01) Negative BRCA1 deletion 50–70 Negative (HRD) 
M232   − (0.02) Positive BRCA 1 + 2 deletion >70 Negative (HRD) 
M248   − (0.01) Positive Mosaic BRCA1 deletion, BRCA2 duplication 50–70 Negative (HRD) 
M112 c.6935A>T p.D2312V Benign − (0.01) Positive Mosaic BRCA1 deletion, BRCA2 duplication 50 Intermediate (HRi) 
M156   − (0.01) Positive BRCA2 deletion >70 Negative (HRD) 
M093   − (0.01) Positive WT >70 Negative (HRD) 
M260   − (0.01) Positive WT 30 Negative (HRD) 
M270   − (0.01) Positive WT 50–70 Negative (HRD) 
M271   − (0.01) Positive WT 30 Negative (HRD) 
M077   − (0.01) Positive WT >50 Negative (HRD) 
M253   − (0.01) Positive WT 50 Intermediate (HRi) 
M209   − (0.01) Positive WT 50 Intermediate (HRi) 
M055   − (0.01) Positive WT 50–70 Intermediate (HRi) 
M278   − (0.01)   10 Intermediate (HRi) 
BRCA2 mutationBRCA1 promotor hypermethylation (score)BRCA1 mRNA RNAscopeBRCA LGRsTumor %RECAP status
SampleMutationPathogenicity
M057 c.517G>C p.G173R Pathogenic $    >50% Negative (HRD) 
P002 c.7617+1G>T Pathogenic    60% Negative (HRD) 
M096 c.7285G>T p.E2429X Pathogenic $    50–70% Negative (HRD) 
M188 c.3846_3847del p.T1282fs Pathogenic    Macrodissected (FFPE) Negative (HRD) 
M231 c.755_758del p.D252fs Pathogenic    65% Negative (HRD) 
M275 c.3269delT p.M1090fs Pathogenic    33% Negative (HRD) 
M213 c.1708A>C p.N570H Benign    >50% Positive (HRP) 
M114 c.6829T>C p.L2277L Benign    40% Positive (HRP) 
P9 c.3445A>G p.M1149V Benign    50% Positive (HRP) 
M211 c.5054C>T p.S1685L Benign − (0.02) Positive  >50 Negative (HRD) 
M106 c.6347A>G p.H2116R Benign − (0.01) Positive  50–70 Negative (HRD) 
P001   + (NA) Negative  — Negative (HRD) 
M028   + (NA) Negative  — Negative (HRD) 
M119   + (0.56) Negative  50–70 Negative (HRD) 
M131   + (0.82) Heterogeneous  >70 Negative (HRD) 
M182   + (0.45) Negative  >50 Negative (HRD) 
M277   + (0.24) Heterogeneous  20 Negative (HRD) 
M141   + (0.29) Negative  50 Intermediate (HRi) 
M094   − (0.01) Negative BRCA1 deletion 50–70 Negative (HRD) 
M232   − (0.02) Positive BRCA 1 + 2 deletion >70 Negative (HRD) 
M248   − (0.01) Positive Mosaic BRCA1 deletion, BRCA2 duplication 50–70 Negative (HRD) 
M112 c.6935A>T p.D2312V Benign − (0.01) Positive Mosaic BRCA1 deletion, BRCA2 duplication 50 Intermediate (HRi) 
M156   − (0.01) Positive BRCA2 deletion >70 Negative (HRD) 
M093   − (0.01) Positive WT >70 Negative (HRD) 
M260   − (0.01) Positive WT 30 Negative (HRD) 
M270   − (0.01) Positive WT 50–70 Negative (HRD) 
M271   − (0.01) Positive WT 30 Negative (HRD) 
M077   − (0.01) Positive WT >50 Negative (HRD) 
M253   − (0.01) Positive WT 50 Intermediate (HRi) 
M209   − (0.01) Positive WT 50 Intermediate (HRi) 
M055   − (0.01) Positive WT 50–70 Intermediate (HRi) 
M278   − (0.01)   10 Intermediate (HRi) 

NOTE: Methylation score between 0.0 and 1.0, cut-off for presence of promotor hypermethylation is >0.2. $ Mutations were somatic.

Thus, 12/23 HRD and 1/6 HRi samples were explained by a BRCA2 mutation or BRCA1 promoter hypermethylation. Subsequently, we proceeded with an MLPA analysis for BRCA1 and BRCA2 on the 16 HRD and HRi samples that remained unexplained to identify possible LGRs. Large BRCA1 deletions were found in four samples and BRCA2 deletions in two samples (Table 1), of which one tumor harbored both a BRCA1 and BRCA2 deletion. Two tumors [M248 (HRD) and M112 (HRi)] showed extensive chromosomal instability as they contained a mosaic BRCA1 deletion (meaning the deletion was present in a subclone of the tumor) and a BRCA2 duplication (Table 1). In total, BRCA defects have thus been identified in 16/23 HRD and 2/6 HRi samples.

Silencing of BRCA1 was validated in tumors with BRCA1 promoter hypermethylation (n = 7) and BRCA1 LGRs (n = 4) by RNAscope in situ RNA hybridization (Supplementary Fig. S2; Table 1). All tumors displaying BRCA1 promoter hypermethylation showed absence of BRCA1 RNA, except for two heterogeneous samples that contained BRCA1 positive and negative areas (Supplementary Fig. S2). As expected, the BRCA1 deletion in tumor M094 led to a total absence of BRCA1 mRNA (Supplementary Fig. S2). The mosaic BRCA1 deletions did not result in complete BRCA1 silencing (Supplementary Fig. S2). Neither did the BRCA1 deletion in M232, however this tumor also harbored a BRCA2 deletion that can explain the HRD phenotype.

After thorough analysis of the BRCA genes using several techniques, 13/23 HRD and 1/6 HRi samples could be explained by deficiencies in BRCA. Moreover, 3/23 HRD (M131, M277, and M248) and 1/6 HRi (M112) tumors also harbored BRCA deficiencies (BRCA1 promoter methylation or mosaic BRCA1 deletions), which explain the HRi and partially the HRD phenotype, as these tumors showed heterogeneous BRCA1 mRNA expression. Finally, 7/23 HRD and 4/6 HRi tumors did not show any BRCA defects and therefore remained unexplained (Fig. 2). To further characterize these HRD tumors, functional features correlating with the HRD phenotype were determined.

Figure 2.

Characteristics of HRD and HRi tumors. Overview of molecular alterations and TIL counts in each HRD or HRi tumor (NB, data for one HRD tumor is absent, because DNA was unavailable). ER, estrogen receptor. High TILs were defined as >10% TILs.

Figure 2.

Characteristics of HRD and HRi tumors. Overview of molecular alterations and TIL counts in each HRD or HRi tumor (NB, data for one HRD tumor is absent, because DNA was unavailable). ER, estrogen receptor. High TILs were defined as >10% TILs.

Close modal

HRD tumors show more tumor infiltrating lymphocytes than HRP tumors

Recently, a subgroup of patients with TNBC was identified who showed good response to immune checkpoint inhibition through programmed death-ligand 1 (PD-L1) blockade (27, 28). This subgroup was characterized by having >10% TILs and high CD8 lymphocyte counts in the tumor centers (28). Because 11/17 TNBCs in our study showed HRD, we hypothesized that the RECAP test might select for a specific subgroup of patients with TNBC who might benefit from PD-L1 therapy. We found that significantly more HRD tumors (6/22) had >10% TILs than HRP tumors (0/28) (P = 0.004; Fig. 3). Also, tumors with BRCA defects showed more frequently >10% TILs compared with non-BRCA tumors (P = 0.001), which was also true for TNBCs compared with ER/PR+ tumors (P = 0.026).

Figure 3.

HRD tumors more frequently showed >10% tumor infiltrating lymphocytes than HRP tumors. A, HE sections were scored for stromal TILs. Examples of samples with <1%, 15%, and 70% TILs are shown respectively. B, More HRD tumors (6/22) had >10% TILs than HRP tumors (0/28; P = 0.004). Also, tumors with BRCA defects showed more frequently >10% TILs compared with non-BRCA tumors (P = 0.001), which was also true for TNBCs compared to ER/PR+ tumors (P = 0.026). Significance was calculated by Fisher exact test.

Figure 3.

HRD tumors more frequently showed >10% tumor infiltrating lymphocytes than HRP tumors. A, HE sections were scored for stromal TILs. Examples of samples with <1%, 15%, and 70% TILs are shown respectively. B, More HRD tumors (6/22) had >10% TILs than HRP tumors (0/28; P = 0.004). Also, tumors with BRCA defects showed more frequently >10% TILs compared with non-BRCA tumors (P = 0.001), which was also true for TNBCs compared to ER/PR+ tumors (P = 0.026). Significance was calculated by Fisher exact test.

Close modal

HRD tumors show mutational signatures related to BRCA deficiencies and microsatellite instability

Next, WES was performed to determine the molecular landscape of HRD tumors. A selection of HRD (n = 8) and HRi (n = 3) tumors with BRCA1/2 mutations/deletions, BRCA1 promoter hypermethylation, and BRCA WT tumors and one HRP tumor were subjected to WES. First, mutational load was determined in these tumors and high mutational load did not correlate with high numbers of TILs. Second, we did not identify commonly mutated genes other than BRCA1/2, which might explain the functional HR defect. Third, WES data were used to identify mutational signatures which are specific combinations of mutations that arise due to a certain underlying mutational or DNA repair processes (29).

Mutational signatures were derived from the WES data from HRD/HRi tumors of which matching normal DNA was available to filter out germline variants (n = 10) to explore novel mechanisms related to or underlying the HRD phenotype. Because discussion in the field exists that mutational signatures can only be faithfully obtained from WGS instead of WES data, we first carried out a pilot experiment comparing mutational signature analysis using all somatic mutations in five BrC WGS datasets (30) and filtered these WGS datasets to only contain somatic mutations on exonic regions. Both methods resulted in similar distributions of the mutational signatures (Supplementary Fig. S3).

Mutational signature 3 is related to failure of DSB repair by HR and associated with germline and somatic BRCA1/2 defects in breast, pancreatic, and ovarian cancers (31). Signature 3 was present in 6 of 10 analyzed samples (M94, M95, M119, M131, M141, and M221; Fig. 4). APOBEC-related mutagenesis (predominantly C>G or C>T substitutions in TCA or TCT motifs) is captured in signatures 2 and 13, which arise through activity of the AID/APOBEC family. BRCA-related signatures could also be identified to a lesser extent in samples (M211 and M094) having a high mutational burden of signatures 2 and 13.

Figure 4.

HRD and HRi tumors showed mutational signatures related to BRCA and MSI. Relative and absolute contribution of the mutational signatures in WES datasets is depicted, and total numbers of single-nucleotide mutations and percentage of TILs in the sample.

Figure 4.

HRD and HRi tumors showed mutational signatures related to BRCA and MSI. Relative and absolute contribution of the mutational signatures in WES datasets is depicted, and total numbers of single-nucleotide mutations and percentage of TILs in the sample.

Close modal

Microsatellite instability in HRD breast cancers

One tumor (M077) showed a high mutational load and high contributions of signatures 15, 20, and 26, which are related to microsatellite instability (MSI) and mismatch repair (MMR) deficiency (http://cancer.sanger.ac.uk/cosmic/signatures). MSI is a common feature in endometrial and gastro-intestinal cancers, caused by either germline (Lynch syndrome) or somatic mutations in one of the MMR genes and/or promoter hypermethylation (32, 33). Presence of MSI in tumor M077 was confirmed using pentaplex PCR and IHC for the MMR proteins, which showed absence of MLH1 and PMS2, caused by MLH1 promoter methylation (Supplementary Fig. S4).

A set of 44 tumors (20 HRD, 6 HRi, 18 HRP) was subjected to MSI analysis by pentaplex PCR and IHC for the MMR proteins. One HRD tumor (M188) of which mutational signatures were not available, also showed MSI. MSI was never detected in any HRP tumors (Fig. 5). Tumor M188 showed absence of MSH2 and MSH6, caused by a homozygous deletion of MSH2 (Supplementary Fig. S4). The two MSI BrCs (M077 and M188) were TN and BRCA WT and ER positive and BRCA2 mutated, respectively (Fig. 2). The incidence of MSI within the HRD group (2/20, 10%) is significantly higher than the incidence of MSI in the unselected breast cancer population (1%; P = 0.017; refs. 34, 35).

Figure 5.

Two HRD breast tumors showed MSI. A, MSI was found in two HRD tumors, but not in any HRP or HRi tumors. B, IHC staining of MLH1, PMS2, MSH2, and MSH6.

Figure 5.

Two HRD breast tumors showed MSI. A, MSI was found in two HRD tumors, but not in any HRP or HRi tumors. B, IHC staining of MLH1, PMS2, MSH2, and MSH6.

Close modal

Here, a unique series of fresh primary breast cancer tissues (n = 125) has been analyzed for HRD using the functional RECAP test. This first large validation study describes that functional assessment of HR in a pseudo-diagnostic setting is achievable and produces robust and interpretable results for most patients (74%). We found that the percentage of HRD tumors detected by the RECAP test is similar in this larger cohort, as compared with our previous report. Therefore, both cohorts were combined to achieve more power to thoroughly investigate the molecular mechanism underlying the HRD phenotype. Sixteen HRD samples showed deficiencies in BRCA1/2 (BRCA mutations, deletions, or promoter hypermethylation), whereas seven HRD tumors were non-BRCA related, demonstrating that HRD tumors encompass more than only BRCA-deficient tumors.

Several different HRD tests have been designed to identify HRD tumors in addition to the BRCA mutated or promoter methylated tumors to enlarge the population of patients with breast cancer that could benefit from treatments targeting the HR pathway. For example, the BRCAness classifier, which is based on specific genomic patterns derived from copy number data of BRCA1/2 mutated breast cancers that also occur in sporadic cancers (10, 11) and the Myriad MyChoice HRD test, which is a combined score of three different structural chromosomal aberrations [telomeric allelic imbalance (TAI), large-scale transition (LST), and loss of heterozygosity (LOH); ref. 36]. Both the BRCAness classifier and MyChoice are robust, easily applicable in the clinic, and have also been validated to predict in vivo response to high dose chemotherapy and neo-adjuvant platinum-based therapy, respectively, in patients with TNBC (37, 38). As opposed to the neo-adjuvant setting, the MyChoice HRD test did not predict response to PARPi therapy in platinum-sensitive recurrent ovarian cancer (39). These genomic HRD tests have the drawback that they do not determine the real-time HR status, also it remains unclear whether all HRD cases are identified. In theory, the functional RECAP test can also detect reversion of the HRD phenotype in BRCA-deficient tumors, that have been treated with various DNA damaging chemotherapies that may have induced resistance. Moreover, the BRCAness classifier focuses on TNBC only, whereas the RECAP test identifies HRD independent of hormonal status. More recently, a HRD test based on several genomic signatures has been published, HRDetect (40). This test has recently been shown to predict in vivo response to platinum-based therapies in advanced breast cancer (41). HRDetect relies on whole genome sequencing and is therefore more expensive and has a longer turnaround time for biopsy results, hampering its clinical implementation. Furthermore, the tumor cell percentage needs to be above 50% for reliable results, which is not a prerequisite for the RECAP test. However, HRDetect has the advantage that it can be performed on frozen material, whereas the RECAP test requires fresh material. The percentage of non-BRCA HRD tumors (approximately 33%) detected by the HRDetect and the RECAP test are quite comparable, although it remains elusive whether these tests identify the same tumors, therefore comparison of several HRD tests within the same patient cohort is required.

The major strength of this study is that functional diagnostics have been applied to a unprecedented large collection of tumors. The advantage of the RECAP test over genetic tests, is its functional character for exploring the HR phenotype rather than the static nature of genomic tests. Also, the RECAP test is feasible in samples with low tumor percentage, because the microscopic read-out allows differentiation between tumor and stromal cells. The RECAP test has a high success rate and results are available within one week after the biopsy procedure. In this study, reproducibility and robustness of the RECAP test is validated in an independent set of 129 tumors. For the molecular analyses to unravel the mechanisms underlying the HRD phenotype, the 41 samples included in our previous publication were also included, to achieve more power. The main limitation of this study is that although prospective trials evaluating the predictive value of RECAP for in vivo patient response to PARPi have been initiated, results are not yet available. However, previously a functional RAD51 test performed on biopsies obtained from patients 24 hours after start of therapy correlated with response to anthracycline-based therapies, indicating that functional assessment of HR can have predictive value for therapy response (13).

Among the spectrum of BRCA defects, we have not identified any BRCA1 mutations. This is somewhat remarkable but could be due to a selection bias, as the Erasmus Medical Center is specialized in hereditary BrC and all patients with TNBC are tested, therefore most families with hereditary BRCA1 mutations have been identified and carriers are offered strict screening programs or undergo prophylactic surgery in The Netherlands (42). Also, in this study there is a selection bias for tumor size, as the tumor should be large enough to provide residual material without compromising standard diagnostic procedures. Since BRCA mutation carriers are offered strict screening programs, tumors are often identified at an early stage and residual material is not available for the RECAP test. Also, many BRCA1 mutation carriers with TNBC are treated with neoadjuvant chemotherapy, which was an exclusion criterion for this study.

Clinical consequences of BRCA1 promoter methylation are unclear (43). In the current study, BRCA1 promoter methylation resulted in absence of BRCA1 RNA in four samples, but in two tumors there was still heterogeneous expression of BRCA1 RNA (Supplementary Fig. S2). In these tumors, the percentage of cells with RAD51 foci was 1% and 2%, respectively. The discrepancy between the very low HRD score and heterogeneous BRCA1 RNA could be explained by sampling from different areas of the tumor, since the tumor sample for the RECAP test was irradiated and an unirradiated tumor sample was used for molecular analyses. This sampling error is not limited to this study, but also occurs in regular diagnostics when biopsies are obtained from a certain region of a heterogeneous tumor. Tumor heterogeneity in BRCA1 promoter methylated tumors is very important for clinical decisions on PARPi use. If subsequent studies reveal that this phenomenon is observed in a large fraction of these tumors, PARPi may not be very effective in tumors with BRCA1 promoter methylation.

We identified 6 HRi BrCs in the current cohort. Only in one of the HRi tumors, distinct areas of RAD51 negative and RAD51 positive tumor cells were observed, suggesting clonal heterogeneity. The other HRi tumors all showed interspersed RAD51 negative as well as RAD51 positive tumor cells. As a BRCA defect was found in 2/6 HRi tumors, they biologically resemble HRD tumors. However, whether HRi tumors benefit from PARPi treatment remains to be elucidated.

Mutational signature analyses were performed to explore novel mechanisms related to or underlying the HRD phenotype. One HRD tumor that showed a large contribution of three signatures related to MSI and MMR deficiency proved to be truly MSI. Using pentaplex PCR and IHC, MSI was discovered in two HRD (2/20, 10%) but not in HRP or HRi tumors. This incidence is much higher than in the unselected breast cancer population (1%; refs. 34, 35), suggesting that the RECAP test may also identify MSI tumors. The relation between MSI and HRD as well as the order in which tumors develop these deficiencies remains unclear and future research is required. We hypothesize that either MSI tumors acquire HRD over time due to accumulation of mutations in genes involved in HR (44), or HRD tumors acquire MSI at a later stage of tumor development as a compensatory mechanism, to lower replication fork instability by not repairing mismatches but rather continuing DNA replication. As MSI tumors have many neo-antigens, PD-L1 blockade therapy showed antitumor activity in phase I trials (45). Recently, a first report of a patient with MSI BrC showing a profound response to PD-L1 blockade was published (46). Moreover, tumors with high numbers of TILs are generally more sensitive to immunotherapy (47). Interestingly, in our cohort, the two MSI tumors comprise a different subset of HRD tumors than the ones with high TILs. Thus, the RECAP test identifies not only tumors with BRCA defects (n = 16), but also a subgroup of breast cancers that might respond well to immunotherapy due to either MSI (n = 2) or high TIL counts (n = 6).

The RECAP test is a robust functional HR assay, detecting both BRCA1/2-deficient and BRCA1/2-proficient HRD tumors. Functional assessment of HR in a pseudo-diagnostic setting is achievable and produces robust and interpretable results. Clinical trials evaluating the predictive value of the RECAP test for in vivo response to PARPi have been initiated.

H.J. Dubbink reports receiving commercial research grants from AstraZeneca. W.N.M. Dinjens reports receiving speakers bureau honoraria from Roche, and is a consultant/advisory board member for Amgen. No potential conflicts of interest were disclosed by the other authors.

Conception and design: T.G. Meijer, N.S. Verkaik, K.A.T. Naipal, A. Jager, D.C. van Gent

Development of methodology: W.N.M. Dinjens, P.M. Nederlof, H.J G. van de Werken, J.W.M. Martens, A. Jager, D.C. van Gent

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): T.G. Meijer, N.S. Verkaik, A.M. Sieuwerts, K.A.T. Naipal, C.H.M. van Deurzen, M.A. den Bakker, H.-J. Dubbink, T.D. den Toom, W.N.M. Dinjens, E. Lips, P.M. Nederlof, A. Jager

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): T.G. Meijer, J. van Riet, H.-J. Dubbink, T.D. den Toom, W.N.M. Dinjens, E. Lips, P.M. Nederlof, M. Smid, H.J G. van de Werken, J.W.M. Martens, A. Jager, D.C. van Gent

Writing, review, and/or revision of the manuscript: T.G. Meijer, N.S. Verkaik, A.M. Sieuwerts, K.A.T. Naipal, C.H.M. van Deurzen, H.-J. Dubbink, W.N.M. Dinjens, E. Lips, P.M. Nederlof, M. Smid, H.J G. van de Werken, R. Kanaar, J.W.M. Martens, A. Jager, D.C. van Gent

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): A.M. Sieuwerts, J. van Riet, H.F.B.M. Sleddens, A. Jager

Study supervision: R. Kanaar, A. Jager, D.C. van Gent

The authors thank many colleagues from the Departments of Molecular Genetics, Medical Oncology, and Pathology at Erasmus MC as well as from the Maasstad Hospital, who contributed to the collection of patient material. We thank Lindsey Oudijk for her assistance with the TILs scoring. We thank Ronald van Marion for expert technical assistance. D.C. van Gent, A. Jager, and R. Kanaar have received funding from the Dutch Cancer Society (Alpe d’Huzes grant number EMCR 2014-7048 and grant number EMCR 2008-4045). This work is part of the Oncode Institute, which is partly financed by the Dutch Cancer Society and was funded by the gravitation program CancerGenomiCs.nl from the Netherlands Organisation for Scientific Research (NWO).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Siegel
RL
,
Miller
KD
,
Jemal
A
. 
Cancer Statistics, 2017
.
CA Cancer J Clin
2017
;
67
:
7
30
.
2.
Nelson
HD
,
Fu
R
,
Goddard
K
,
Mitchell
JP
,
Okinaka-Hu
L
,
Pappas
M
, et al
Risk assessment, genetic counseling, and genetic testing for BRCA-Related Cancer: systematic review to update the U.S. Preventive Services Task Force Recommendation
.
Pacific Northwest Evidence-based Practice Center
,
Rockville (MD)
:
Agency for Healthcare Research and Quality (US)
; 
2013
Dec.
Report No.: 12-05164-EF-1
.
3.
Hartman
AR
,
Kaldate
RR
,
Sailer
LM
,
Painter
L
,
Grier
CE
,
Endsley
RR
, et al
Prevalence of BRCA mutations in an unselected population of triple-negative breast cancer
.
Cancer
2012
;
118
:
2787
95
.
4.
Konecny
GE
,
Kristeleit
RS
. 
PARP inhibitors for BRCA1/2-mutated and sporadic ovarian cancer: current practice and future directions
.
Br J Cancer
2016
;
115
:
1157
73
.
5.
Lord
CJ
,
Ashworth
A
. 
The DNA damage response and cancer therapy
.
Nature
2012
;
481
:
287
94
.
6.
Robson
M
,
Im
SA
,
Senkus
E
,
Xu
B
,
Domchek
SM
,
Masuda
N
, et al
Olaparib for metastatic breast cancer in patients with a germline BRCA mutation
.
N Engl J Med
2017
;
377
:
523
33
.
7.
Wendie
DdB
,
Kasmintan
AS
,
Sophie
S
,
Kasaian
K
,
Leelakumari
S
,
Jones
M
, et al
Homologous recombination deficiency in breast cancer: a clinical review
.
JCO Prec Oncol
2017
:
1
13
.
8.
Abkevich
V
,
Timms
KM
,
Hennessy
BT
,
Potter
J
,
Carey
MS
,
Meyer
LA
, et al
Patterns of genomic loss of heterozygosity predict homologous recombination repair defects in epithelial ovarian cancer
.
Br J Cancer
2012
;
107
:
1776
82
.
9.
Birkbak
NJ
,
Wang
ZC
,
Kim
JY
,
Eklund
AC
,
Li
Q
,
Tian
R
, et al
Telomeric allelic imbalance indicates defective DNA repair and sensitivity to DNA-damaging agents
.
Cancer Discov
2012
;
2
:
366
75
.
10.
Joosse
SA
,
Brandwijk
KI
,
Devilee
P
,
Wesseling
J
,
Hogervorst
FB
,
Verhoef
S
, et al
Prediction of BRCA2-association in hereditary breast carcinomas using array-CGH
.
Breast Cancer Res Treat
2012
;
132
:
379
89
.
11.
Joosse
SA
,
van Beers
EH
,
Tielen
IH
,
Horlings
H
,
Peterse
JL
,
Hoogerbrugge
N
, et al
Prediction of BRCA1-association in hereditary non-BRCA1/2 breast carcinomas with array-CGH
.
Breast Cancer Res Treat
2009
;
116
:
479
89
.
12.
Konstantinopoulos
PA
,
Spentzos
D
,
Karlan
BY
,
Taniguchi
T
,
Fountzilas
E
,
Francoeur
N
, et al
Gene expression profile of BRCAness that correlates with responsiveness to chemotherapy and with outcome in patients with epithelial ovarian cancer
.
J Clin Oncol
2010
;
28
:
3555
61
.
13.
Graeser
M
,
McCarthy
A
,
Lord
CJ
,
Savage
K
,
Hills
M
,
Salter
J
, et al
A marker of homologous recombination predicts pathologic complete response to neoadjuvant chemotherapy in primary breast cancer
.
Clin Cancer Res
2010
;
16
:
6159
68
.
14.
Naipal
KA
,
Verkaik
NS
,
Ameziane
N
,
van Deurzen
CH
,
Ter Brugge
P
,
Meijers
M
, et al
Functional ex vivo assay to select homologous recombination-deficient breast tumors for PARP inhibitor treatment
.
Clin Cancer Res
2014
;
20
:
4816
26
.
15.
Gross
E
,
van Tinteren
H
,
Li
Z
,
Raab
S
,
Meul
C
,
Avril
S
, et al
Identification of BRCA1-like triple-negative breast cancers by quantitative multiplex-ligation-dependent probe amplification (MLPA) analysis of BRCA1-associated chromosomal regions: a validation study
.
BMC Cancer
2016
;
16
:
811
.
16.
Li
H
,
Durbin
R
. 
Fast and accurate short read alignment with Burrows-Wheeler transform
.
Bioinformatics
2009
;
25
:
1754
60
.
17.
Li
H
,
Handsaker
B
,
Wysoker
A
,
Fennell
T
,
Ruan
J
,
Homer
N
, et al
The Sequence Alignment/Map format and SAMtools
.
Bioinformatics
2009
;
25
:
2078
9
.
19.
Forbes
SA
,
Beare
D
,
Boutselakis
H
,
Bamford
S
,
Bindal
N
,
Tate
J
, et al
COSMIC: somatic cancer genetics at high-resolution
.
Nucleic Acids Res
2017
;
45
:
D777
83
.
20.
McKenna
A
,
Hanna
M
,
Banks
E
,
Sivachenko
A
,
Cibulskis
K
,
Kernytsky
A
, et al
The genome analysis toolkit: a MapReduce framework for analyzing next-generation DNA sequencing data
.
Genome Res
2010
;
20
:
1297
303
.
21.
Sherry
ST
,
Ward
MH
,
Kholodov
M
,
Baker
J
,
Phan
L
,
Smigielski
EM
, et al
dbSNP: the NCBI database of genetic variation
.
Nucleic Acids Res
2001
;
29
:
308
11
.
22.
Wang
K
,
Li
M
,
Hakonarson
H
. 
ANNOVAR: functional annotation of genetic variants from high-throughput sequencing data
.
Nucleic Acids Res
2010
;
38
:
e164
.
23.
Blokzijl
F
,
Janssen
R
,
van Boxtel
R
,
Cuppen
E
. 
MutationalPatterns: an integrative R package for studying patterns in base substitution catalogues Francis
.
bioRxiv Prepr
2017
.
doi
: .
24.
Alexandrov
LB
,
Nik-Zainal
S
,
Wedge
DC
,
Campbell
PJ
,
Stratton
MR
. 
Deciphering signatures of mutational processes operative in human cancer
.
Cell Rep
2013
;
3
:
246
59
.
25.
van Lier
MG
,
Wagner
A
,
van Leerdam
ME
,
Biermann
K
,
Kuipers
EJ
,
Steyerberg
EW
, et al
A review on the molecular diagnostics of Lynch syndrome: a central role for the pathology laboratory
.
J Cell Mol Med
2010
;
14
:
181
97
.
26.
Salgado
R
,
Denkert
C
,
Demaria
S
,
Sirtaine
N
,
Klauschen
F
,
Pruneri
G
, et al
The evaluation of tumor-infiltrating lymphocytes (TILs) in breast cancer: recommendations by an International TILs Working Group 2014
.
Ann Oncol
2015
;
26
:
259
71
.
27.
Kurozumi
S
,
Fujii
T
,
Matsumoto
H
,
Inoue
K
,
Kurosumi
M
,
Horiguchi
J
, et al
Significance of evaluating tumor-infiltrating lymphocytes (TILs) and programmed cell death-ligand 1 (PD-L1) expression in breast cancer
.
Med Mol Morphol
2017
;
50
:
185
94
.
28.
Schmid
P
,
Cruz
C
,
Braiteh
FS
,
Eder
JP
,
Tolaney
S
,
Kuter
I
, et al
Atezolizumab in metastatic TNBC (mTNBC): Long-term clinical outcomes and biomarker analyses
.
Abstract #2986, AACR
2017
.
29.
Nik-Zainal
S
,
Alexandrov
LB
,
Wedge
DC
,
Van Loo
P
,
Greenman
CD
,
Raine
K
, et al
Mutational processes molding the genomes of 21 breast cancers
.
Cell
2012
;
149
:
979
93
.
30.
Nik-Zainal
S
,
Davies
H
,
Staaf
J
,
Ramakrishna
M
,
Glodzik
D
,
Zou
X
, et al
Landscape of somatic mutations in 560 breast cancer whole-genome sequences
.
Nature
2016
;
534
:
47
54
.
31.
Alexandrov
LB
,
Nik-Zainal
S
,
Wedge
DC
,
Aparicio
SAJR
,
Behjati
S
,
Biankin
AV
, et al
Signatures of mutational processes in human cancer
.
Nature
2013
;
500
:
415
21
.
32.
Geurts-Giele
WR
,
Leenen
CH
,
Dubbink
HJ
,
Meijssen
IC
,
Post
E
,
Sleddens
HF
, et al
Somatic aberrations of mismatch repair genes as a cause of microsatellite-unstable cancers
.
J Pathol
2014
;
234
:
548
59
.
33.
Lynch
HT
,
Snyder
CL
,
Shaw
TG
,
Heinen
CD
,
Hitchins
MP
. 
Milestones of Lynch syndrome: 1895–2015
.
Nat Rev Cancer
2015
;
15
:
181
94
.
34.
Dudley
JC
,
Lin
MT
,
Le
DT
,
Eshleman
JR
. 
Microsatellite instability as a biomarker for PD-1 blockade
.
Clin Cancer Res
2016
;
22
:
813
20
.
35.
Le
DT
,
Durham
JN
,
Smith
KN
,
Wang
H
,
Bartlett
BR
,
Aulakh
LK
, et al
Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade
.
Science
2017
;
357
:
409
13
.
36.
Timms
KM
,
Abkevich
V
,
Hughes
E
,
Neff
C
,
Reid
J
,
Morris
B
, et al
Association of BRCA1/2 defects with genomic scores predictive of DNA damage repair deficiency among breast cancer subtypes
.
Breast Cancer Res
2014
;
16
:
475
.
37.
Vollebergh
MA
,
Lips
EH
,
Nederlof
PM
,
Wessels
LF
,
Schmidt
MK
,
van Beers
EH
, et al
An aCGH classifier derived from BRCA1-mutated breast cancer and benefit of high-dose platinum-based chemotherapy in HER2-negative breast cancer patients
.
Ann Oncol
2011
;
22
:
1561
70
.
38.
Telli
ML
,
Timms
KM
,
Reid
J
,
Hennessy
B
,
Mills
GB
,
Jensen
KC
, et al
Homologous recombination deficiency (HRD) score predicts response to platinum-containing neoadjuvant chemotherapy in patients with triple-negative breast cancer
.
Clin Cancer Res
2016
;
22
:
3764
73
.
39.
Mirza
MR
,
Monk
BJ
,
Herrstedt
J
,
Oza
AM
,
Mahner
S
,
Redondo
A
, et al
Niraparib maintenance therapy in platinum-sensitive, recurrent ovarian cancer
.
N Engl J Med
2016
;
375
:
2154
64
.
40.
Davies
H
,
Glodzik
D
,
Morganella
S
,
Yates
LR
,
Staaf
J
,
Zou
X
, et al
HRDetect is a predictor of BRCA1 and BRCA2 deficiency based on mutational signatures
.
Nat Med
2017
;
23
:
517
25
.
41.
Zhao
EY
,
Shen
Y
,
Pleasance
E
,
Kasaian
K
,
Leelakumari
S
,
Jones
M
, et al
Homologous recombination deficiency and platinum-based therapy outcomes in advanced breast cancer
.
Clin Cancer Res
2017
;
23
:
7521
30
.
42.
Drooger
JC
,
Hooning
MJ
,
Seynaeve
CM
,
Baaijens
MH
,
Obdeijn
IM
,
Sleijfer
S
, et al
Diagnostic and therapeutic ionizing radiation and the risk of a first and second primary breast cancer, with special attention for BRCA1 and BRCA2 mutation carriers: a critical review of the literature
.
Cancer Treat Rev
2015
;
41
:
187
96
.
43.
Sun
T
,
Ruscito
I
,
Dimitrova
D
,
Chekerov
R
,
Kulbe
H
,
Baron
U
, et al
Genetic Versus Epigenetic BRCA1 silencing pathways: clinical effects in primary ovarian cancer patients: a study of the tumor bank ovarian cancer consortium
.
Int J Gynecol Cancer
2017
;
27
:
1658
65
.
44.
Zhao
H
,
Thienpont
B
,
Yesilyurt
BT
,
Moisse
M
,
Reumers
J
,
Coenegrachts
L
, et al
Mismatch repair deficiency endows tumors with a unique mutation signature and sensitivity to DNA double-strand breaks
.
Elife
2014
;
3
:
e02725
.
45.
Westdorp
H
,
Fennemann
FL
,
Weren
RD
,
Bisseling
TM
,
Ligtenberg
MJ
,
Figdor
CG
, et al
Opportunities for immunotherapy in microsatellite instable colorectal cancer
.
Cancer Immunol Immunother
2016
;
65
:
1249
59
.
46.
Marleen
K
,
Hugo
MH
,
Petur
S
,
Chalabi
M
,
Schumacher
TN
,
Blank
CU
, et al
Profound immunotherapy response in mismatch repair-deficient breast cancer
.
JCO Prec Oncol
2017
:
1
3
.
47.
Gibney
GT
,
Weiner
LM
,
Atkins
MB
. 
Predictive biomarkers for checkpoint inhibitor-based immunotherapy
.
Lancet Oncol
2016
;
17
:
e542
51
.