Purpose:

Lynch syndrome is defined by germline pathogenic mutations involving DNA mismatch repair (MMR) genes and linked with the development of MMR-deficient colon and endometrial cancers. Whether breast cancers developing in the context of Lynch syndrome are causally related to MMR deficiency (MMRd), remains controversial. Thus, we explored the morphologic and genomic characteristics of breast cancers occurring in Lynch syndrome individuals.

Experimental Design:

A retrospective analysis of 20,110 patients with cancer who underwent multigene panel genetic testing was performed to identify individuals with a likely pathogenic/pathogenic germline variant in MLH1, MSH2, MSH6, or PMS2 who developed breast cancers. The histologic characteristics and IHC assessment of breast cancers for MMR proteins and programmed death-ligand 1 (PD-L1) expression were assessed on cases with available materials. DNA samples from paired tumors and blood were sequenced with Memorial Sloan Kettering–Integrated Mutation Profiling of Actionable Cancer Targets (≥468 key cancer genes). Microsatellite instability (MSI) status was assessed utilizing MSISensor. Mutational signatures were defined using SigMA.

Results:

A total of 272 individuals with Lynch syndrome were identified, 13 (5%) of whom had primary breast cancers. The majority of breast cancers (92%) were hormone receptor–positive tumors. Five (42%) of 12 breast cancers displayed loss of MMR proteins by IHC. Four (36%) of 11 breast cancers subjected to tumor-normal sequencing showed dominant MSI mutational signatures, high tumor mutational burden, and indeterminate (27%) or high MSISensor scores (9%). One patient with metastatic MMRd breast cancer received anti-PD1 therapy and achieved a robust and durable response.

Conclusions:

A subset of breast cancers developing in individuals with Lynch syndrome are etiologically linked to MMRd and may benefit from anti-PD1/PD-L1 immunotherapy.

Translational Relevance

The association of Lynch syndrome and breast cancer has been a widely debated and controversial topic in past decades. Here we characterized the pathologic and genomic features of breast cancers occurring in patients with Lynch syndrome and investigated whether these breast cancers are casually linked to the mismatch repair deficiency (MMRd) that defines Lynch syndrome–related cancers. Of 20,110 patients with cancer who underwent multigene genetic testing on tumor and matched normal DNA samples, 272 individuals with Lynch syndrome were identified, 13 of whom had primary breast cancers. Five of these 13 breast cancers displayed MMRd by IHC and/or dominant microsatellite instability–related mutational signatures as defined by tumor-normal sequencing. One patient with widely metastatic estrogen receptor (ER)-positive/HER2-negative MMRd breast cancer achieved a robust and durable response upon treatment with anti-PD1 immunotherapy. Although Lynch syndrome–associated breast cancers are rare, this study highlights a subset that are etiologically linked to underlying MMRd and may be candidates for immunotherapy.

Lynch syndrome is a cancer predisposition syndrome characterized by germline mutations in major DNA mismatch repair (MMR) genes (MLH1, MSH2, MSH6, or PMS2) and associated with a significantly increased risk of developing colorectal, endometrial, ovarian, small bowel, and ureteral cancers (1–3). The hallmark of Lynch syndrome spectrum cancers is defective DNA mismatch repair deficiency (MMRd) and high levels of microsatellite instability (MSI-H), which occur upon loss of the expression of MMR genes through a germline mutation and inactivation of the second allele through somatic mutation or loss of heterozygosity, and, more rarely, due to MLH1 gene promoter methylation.

Whether individuals with Lynch syndrome are at increased risk of developing breast cancer remains controversial (2, 4–12). Although previous studies have suggested a lack of association between pathogenic germline variants affecting MMR genes and breast cancer risk (7, 11, 12), whether breast cancers occurring in Lynch syndrome individuals are causally related to the genetic alteration of MMR genes is unclear (2). Loss of MMR protein expression has been reported in up to 50% of breast cancers from patients with Lynch syndrome (8, 9). Davies and colleagues reported 2 of 11 patients with MMRd breast cancers identified by whole-genome sequencing occurring in patients with Lynch syndrome (10). Other studies have found no evidence of MMRd in breast tumors derived from patients with Lynch syndrome or individuals with synchronous or metachronous breast plus colorectal cancer (11). In fact, a recent pan-cancer survey of over 15,000 tumors subjected to Memorial Sloan Kettering–Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT) found no evidence of MMRd breast cancer occurring in patients with Lynch syndrome (12).

Defining whether breast cancers developing in the context of Lynch syndrome display MMRd is not a mere academic exercise, given that MSI-H has been approved as a pan-cancer biomarker of clinical benefit from immune checkpoint inhibitors (13). Hence, the identification of MMRd/MSI-H breast cancers in patients with Lynch syndrome has a direct therapeutic implication. Here, we sought to characterize the pathologic and genomic features of breast cancers occurring in patients with Lynch syndrome and to explore whether these breast cancers would be causally linked to the MMRd characteristic of Lynch syndrome–related cancers. By expanding on the analysis by Latham and colleagues (12), including ≥20,000 patients subjected to multigene germline genetic testing to identify primary breast cancers in patients with Lynch syndrome and focusing on the histologic, IHC, and genomic features of these cancers, we provide direct evidence supporting MMRd/MSI-H in a substantial subset of breast cancers in patients with Lynch syndrome.

Study population

The cohort consisted of 20,110 patients with cancer, who underwent multigene panel genetic testing for germline mutations at Memorial Sloan Kettering Cancer Center (MSKCC, New York, NY) from 2015 to present day, as described previously (14). The cohort includes 3,583 patients reported previously from January 2014 to June 2017 by Latham and colleagues (12), and additional 16,527 unique patients derived from the germline sequencing pool. The tumor somatic alterations by MSK-IMPACT with > 468 gene panels of the identified breast cancers were also analyzed.

Individuals with likely pathogenic/pathogenic germline variant in MLH1, MSH2, MSH6, or PMS2 who developed breast cancers were retrospectively identified. The genetic testing was performed in a Clinical Laboratory Improvement Amendments–certified laboratory using tumor/normal sequencing. All germline Lynch syndrome variants for patients in this cohort were reviewed by a board-certified molecular pathologist (D. Mandelker) and classified according to the American College of Medical Genetics and Genomics criteria (15) as likely pathogenic/pathogenic.

The study was conducted in accordance with the Declaration of Helsinki, and the protocol was approved by the Institutional Review Board (IRB) of MSKCC. Every patient included in this study signed a written informed consent form according to the protocol approved by the MSKCC IRB.

Study design and IHC analysis

Histology of all breast cancers were reviewed by two pathologists (C.J. Schwartz and H. Zhang) and graded according to the Nottingham grading index (16). Clinicopathologic data were retrieved from the electronic medical records and curated by C.J. Schwartz.

Representative formalin-fixed, paraffin-embedded (FFPE) tissue blocks of invasive carcinomas from patients with Lynch syndrome were retrieved from the archive at MSKCC. Estrogen receptor (ER) and human epidermal growth factor receptor 2 (HER2) status were assessed by IHC and/or FISH as described previously (17), following the American Society of Clinical Oncology/College of American Pathologists guideline recommendations (18, 19). IHC staining for MMR proteins (MLH1, PMS2, MSH2, and MSH6) were performed according to methods described previously (20). Positive and negative controls, including internal positive controls, were assessed in every run and every case, respectively. Tumors with positive nuclear expression were classified as normal/retained MMR protein expression. All IHC reactions were reviewed by three pathologists (C.J. Schwartz., J. Shia, and H. Zhang). PD-L1 (SP142) IHC was performed using Ventana Benchmark Ultra System (Ventana Medical Systems) with antibody detection using the OptiView DAB IHC Detection Kit (Ventana Medical Systems), according to the manufacturer's manual (21). PD-L1 was assessed by two pathologists (C.J. Schwartz and H. Zhang) and scored as expression on stromal tumor-infiltrating immune cells occupying ≥1% of the tumor area as described previously (22, 23).

Tumor-infiltrating lymphocytes

All Lynch syndrome breast cancers were evaluated for the presence of stromal tumor-infiltrating lymphocytes (TIL) following the recommendations by the International TILs Working Group 2014 (24). Three pathologists (C.J. Schwartz, H.Y. Wen, and H. Zhang) quantified the percentage of stromal area within the borders of the invasive tumor covered by TILs, and the average was reported. TILs outside of the tumor border, around ductal carcinoma in situ (DCIS) and normal lobules, and in tumor zones with crush artifacts, necrosis and regressive hyalinization were excluded.

Massively parallel sequencing

Adequate tumor and normal tissue samples were available from 11 patients and underwent targeted massively parallel sequencing (n = 11) using the FDA-cleared MSK-IMPACT assay, which targets all exons and selected introns of 468 (n = 7) or 505 (n = 4) cancer genes as described previously (25). Of the 11 cases, eight had sequencing performed at the time of diagnosis, and three were among the patients identified from the germline mutation database and were sequenced retrospectively at the time of this study.

In brief, somatic single-nucleotide variants (SNV) were detected using MuTect (v1.17; ref. 26) and small insertions and deletions were identified using a combination of Strelka (v1.0.15; ref. 27), VarScan2 (v2.3.7; ref. 28), Lancet (v1.0.0; ref. 29); and/or Scalpel (v0.5.3; ref. 30) with further curation using visual inspection as described previously (31). Copy-number alterations in the tumors were identified using FACETS (32) as described previously (31). Mutational hotspots were classified according to Chang and colleagues (33).

Analysis of MSI by PCR

Analysis of MSI status by PCR in tumors (n = 11) was performed using the MSI Analysis System, version 1.2 kit (Promega). In brief, this assay analyzes five mononucleotide microsatellite loci, including NR-21, BAT-25, MONO-27, NR-24, and BAT-26, in both tumor and normal DNA. A ≥ 3 bp shift in the tumor DNA relative to the matched normal tissue constituted instability at one locus. Instability at ≥2 of the five microsatellite loci defines MSI-H status, with fewer than two unstable loci classified as microsatellite stable (MSS), according to methods described previously (34). One microliter of amplified PCR product was applied to the Applied Biosystems 3730 DNA Analyzer. Automatic fragment analysis was carried out with GeneMapper 6.0 Software (Applied Biosystems).

Mutational signatures

The mutational signatures exposures were obtained using Signature Multivariate Analysis (SigMA), a tool for the decomposition of mutational signatures that can be optimized for the analysis of targeted capture sequencing panel results obtained from FFPE tissue samples. It has been demonstrated that when only a few SNVs per sample are available, SigMA outperforms other state-of-the-art methods (35). SigMA computes signature exposures for samples with at least five SNVs, while other tools (e.g., DeconstructSig) require ≥10 variants (36). The SigMA was set with customized settings (i.e., data type: msk, cancer type: breast, check msi: true). The exposures obtained were converted using the NNLS algorithm by SigMA into percentages per signature as described previously (36). Additional analysis was performed using the tool SigProfiler, another state-of-the-art tool that identifies the probability for each signature to cause a specific mutation type in a cancer sample. The python package SigProfilerPlotting was used for the visualization of the mutational spectrum (Fig. 2G; ref. 37).

Comparison of Lynch syndrome sequenced breast cancers to matched sequenced breast cancers

ER-positive/HER2-negative (n = 9), ER-positive/HER2-positive (n = 1), and ER-negative/HER2-negative (n = 1) Lynch syndrome breast cancers were compared with 1,918 breast cancers subjected to MSK-IMPACT targeted sequencing from the study by Razavi and colleagues (38). Tumors were matched by hormone receptor (HR) status, tumor type [invasive ductal (n = 9) or lobular carcinoma (n = 2), respectively], age (10-year intervals), and menopausal status at a 1:3 ratio (n = 33), as described previously (31). Comparison of frequencies of genes altered by somatic genetic alterations between Lynch syndrome breast cancers (n = 11) and control-matched breast cancers (n = 33) was performed using Fisher exact test. Multiple testing correction using the Benjamini–Hochberg method was applied to control for the FDR whenever appropriate. Tumor mutational burdens (TMB) and MSISensor scores were compared between Lynch syndrome breast cancers (n = 11) and control-matched breast cancers (n = 33) by the Mann–Whitney U test. Dominant mutational signatures of sequenced tumors with at least five SNVs were compared between Lynch syndrome breast cancers (n = 9) and control-matched breast cancers (n = 16) by Fisher exact test. P values < 0.05 were considered significant. Statistical analysis was performed using R (version 3.1.2).

Data availability statement

Data are available in a repository (cBioPortal for Cancer Genomics) and can be accessed via a DOI link.

Clinical characteristics of study cohort

Two hundred seventy-two (1.4%) MMR gene pathogenic variants indicating Lynch syndrome were identified from the total cohort of 20,110 patients. This included 57 (21%), 96 (35%), 63 (23%), and 56 (21%) variants affecting MLH1, MSH2, MSH6, and PMS2, respectively. Thirteen patients in this cohort (5%) had been diagnosed with primary breast cancers (Table 1), all of whom had confirmatory germline testing using a multigene inherited cancer susceptibility panel comprised of 76 (n = 1) or 88 genes (n = 12). Two of these patients (cases 2 and 3) overlapped with a previous study (14).

Table 1.

Summary of germline alterations and cancer history in Lynch syndrome cohort.

CaseAgeGermline alterationType of mutationFamily historyOther cancer history
46 MSH6 c.3103C>T (p.R1035*) Nonsense (F, CC, 66), (M, BC, 73), (MGM, BC, 63), (MA, OC, 66) None 
43 PMS2 c.137G.T(p. Ser46IIe) Missense (PGM, UC, 73) None 
36 MSH6 c.3959_3962delCAAG (p. Ala1320Glufs*6) Frameshift deletion (MGM, UC, 63) None 
33 MLH1 c.954delC (p. His318Glnfs*49) Deletion (F, CC, 58), (PA, OC, 41; CC, 62) (UC, 33) 
47 PMS2 c.248T>G (p. Leu83*) Nonsense (MGM, BC, 74) (BC, 38) 
64 MSH6 c.3984_3987dup (p. Leu1330Valfs*12) Frameshift deletion (M, BC, 43), (D, BC, 35), (MA, BC 54, 65), (MA, UC, 63) (BC, 46), (FGP with LGD, 66) 
59 MSH6 c.3513+3514delTA (p. Asp117Glufs*5) Frameshift deletion None (UC, 58) 
38 PMS2 exon 11–12 duplication Rearrangement (M, BC, 51) None 
47 MSH6 c.2906A>G (p. Tyr969Cys) Missense None None 
10 63 MSH2 c.484G>A (p. Gly162Arg) Missense (M, CC, 40), (MA, CC, 59), (S, CC, 56), (D, SBC, 51) (SBC, 56), (LC, 66), (RC, 73) 
11 74 MSH2 c.942+3A>T Missense (F, CC, 71), (PGM, CC, 66) (UC, 46), (UrC, 81) (DCIS, 75) 
12 56 MSH6 c.3485_3487delCTG (p. Ala1162del) Deletion (M, UC, 63), (MA, CC, 70), (PGM, BC, 63) (UC, 53) 
13 42 MLH1 c.1852–1854delAAG (p. Lys1852del3) Deletion (PA, CC, 49), (PA, OC, 50), (PA, UrC, 70) None 
CaseAgeGermline alterationType of mutationFamily historyOther cancer history
46 MSH6 c.3103C>T (p.R1035*) Nonsense (F, CC, 66), (M, BC, 73), (MGM, BC, 63), (MA, OC, 66) None 
43 PMS2 c.137G.T(p. Ser46IIe) Missense (PGM, UC, 73) None 
36 MSH6 c.3959_3962delCAAG (p. Ala1320Glufs*6) Frameshift deletion (MGM, UC, 63) None 
33 MLH1 c.954delC (p. His318Glnfs*49) Deletion (F, CC, 58), (PA, OC, 41; CC, 62) (UC, 33) 
47 PMS2 c.248T>G (p. Leu83*) Nonsense (MGM, BC, 74) (BC, 38) 
64 MSH6 c.3984_3987dup (p. Leu1330Valfs*12) Frameshift deletion (M, BC, 43), (D, BC, 35), (MA, BC 54, 65), (MA, UC, 63) (BC, 46), (FGP with LGD, 66) 
59 MSH6 c.3513+3514delTA (p. Asp117Glufs*5) Frameshift deletion None (UC, 58) 
38 PMS2 exon 11–12 duplication Rearrangement (M, BC, 51) None 
47 MSH6 c.2906A>G (p. Tyr969Cys) Missense None None 
10 63 MSH2 c.484G>A (p. Gly162Arg) Missense (M, CC, 40), (MA, CC, 59), (S, CC, 56), (D, SBC, 51) (SBC, 56), (LC, 66), (RC, 73) 
11 74 MSH2 c.942+3A>T Missense (F, CC, 71), (PGM, CC, 66) (UC, 46), (UrC, 81) (DCIS, 75) 
12 56 MSH6 c.3485_3487delCTG (p. Ala1162del) Deletion (M, UC, 63), (MA, CC, 70), (PGM, BC, 63) (UC, 53) 
13 42 MLH1 c.1852–1854delAAG (p. Lys1852del3) Deletion (PA, CC, 49), (PA, OC, 50), (PA, UrC, 70) None 

Abbreviations: BC, breast cancer; CC, colon cancer; D, daughter; DCIS, ductal carcinoma in situ; F, father; FGP, fundic gland polyp; LC, lung cancer; LGD, low-grade dysplasia; M, mother; MA, maternal aunt; MGM, maternal grandmother; OC, ovarian cancer; PA, paternal aunt; PGM, paternal grandmother; RC, rectal cancer; S, son; SBC, small bowel cancer; UC, uterine cancer; UrC, urothelial cancer.

Of the 13 patients, pathogenic germline variants were identified in MLH1 (n = 2, 15%), MSH2 (n = 2, 15%), MSH6 (n = 6, 46%), and PMS2 (n = 3, 23%). Seven patients (54%) had breast cancers as their sole malignancy, 5 patients (38%) had other Lynch syndrome–related cancers, and 1 patient was diagnosed with synchronous uterine and breast carcinomas (8%; Table 1). Eleven patients (84%) had a family history of Lynch syndrome–related neoplasia. The median age of breast cancer diagnosis was 47 years (range, 33–74).

Tumor characteristics and clinical follow-up

Histologically, 10 breast cancers (77%) were invasive ductal carcinomas and three (23%) were invasive lobular carcinomas (Table 2). All breast cancers were of histologic grade 2 (38%) or 3 (62%). The median tumor size was 2.6 cm (range, 1.0–10.0). Of the 13 tumors, 11 tumors (84%) were ER-positive/HER2-negative, one tumor (case 8) was ER-positive/HER2-positive, and the remaining one tumor (case 7) was ER-negative/HER2-negative. Stromal TILs ranged from 1% to 25%. PD-L1 (SP142) staining was positive (>1%) in five of nine tumors (55%; Table 2).

Table 2.

Clinicopathologic characteristics of breast cancers in Lynch syndrome cohort.

CaseAgePresentationTumor typeGradeSize (cm)LVIER (%)PR (%)HER2aStromal TILS (%)PD-L1 (SP142)SurgeryMetastasisTTP (months)Follow-up (months)Outcome
46 Self-palpated Ductal 3.5 Yes 97 99 2+ 2% N/A TM Yes 43 171 AWD 
43 Self-palpated Ductal 3.2 Yes 80 90 0/1+ 20% Negative TM Yes 55 67 DOD 
36 Palpated on physical exam Ductal 3.5 Yes 90 95 10% Negative TM Yes 47 AWD 
33 Screening mammography Ductal 3.3 Yes 99 25% Positive TM No N/A 31 NED 
47 Screening mammography Lobular N/A N/A 100 95 1+ N/A N/A TM Yes 108 240 DOD 
64 Screening mammography Ductal 1.3 Yes 99 30 10% Positive TM Yes 12 13 AWD 
59 Screening mammography Ductal N/A N/A 2+ 10% Positive N/A Yes 11 AWD 
38 Self-palpated Ductal N/A 99 99 3+ 10% N/A N/A Yes N/A 15 AWD 
47 Self-palpated Ductal 2,2 1.0,1.0 Yes 100 70 1+ 10% Positive TM No N/A NED 
10 63 Palpated on physical exam Ductal No 90 1/2+ 2% Negative BCS No N/a 144 NED 
11 74 Screening mammography Lobular 1.15 No 95 90 1/2+ N/A N/A TM No N/a 118 NED 
12 56 Screening mammography Lobular 10 Suspicious 90 60 1% Negative TM No N/a 27 NED 
13 42 Screening mammography Ductal 1.9 Yes 70 80 20% Positive BCS Yes 34 36 AWD 
CaseAgePresentationTumor typeGradeSize (cm)LVIER (%)PR (%)HER2aStromal TILS (%)PD-L1 (SP142)SurgeryMetastasisTTP (months)Follow-up (months)Outcome
46 Self-palpated Ductal 3.5 Yes 97 99 2+ 2% N/A TM Yes 43 171 AWD 
43 Self-palpated Ductal 3.2 Yes 80 90 0/1+ 20% Negative TM Yes 55 67 DOD 
36 Palpated on physical exam Ductal 3.5 Yes 90 95 10% Negative TM Yes 47 AWD 
33 Screening mammography Ductal 3.3 Yes 99 25% Positive TM No N/A 31 NED 
47 Screening mammography Lobular N/A N/A 100 95 1+ N/A N/A TM Yes 108 240 DOD 
64 Screening mammography Ductal 1.3 Yes 99 30 10% Positive TM Yes 12 13 AWD 
59 Screening mammography Ductal N/A N/A 2+ 10% Positive N/A Yes 11 AWD 
38 Self-palpated Ductal N/A 99 99 3+ 10% N/A N/A Yes N/A 15 AWD 
47 Self-palpated Ductal 2,2 1.0,1.0 Yes 100 70 1+ 10% Positive TM No N/A NED 
10 63 Palpated on physical exam Ductal No 90 1/2+ 2% Negative BCS No N/a 144 NED 
11 74 Screening mammography Lobular 1.15 No 95 90 1/2+ N/A N/A TM No N/a 118 NED 
12 56 Screening mammography Lobular 10 Suspicious 90 60 1% Negative TM No N/a 27 NED 
13 42 Screening mammography Ductal 1.9 Yes 70 80 20% Positive BCS Yes 34 36 AWD 

Abbreviations: AWD, alive with disease; DOD, died of disease; ER, estrogen receptor; HER2, human epidermal growth factor receptor 2; LVI, lymphovascular invasion; N/A, not available; NED, no evidence of disease; PR, progesterone receptor; TILs, tumor-infiltrating lymphocytes; TTP, time to disease progression.

aAll tumors with 2+ or 1/2+ staining for HER2 IHC were negative for amplification by fluorescent in situ hybridization.

In terms of locoregional disease control, 69% of patients received total mastectomy and 15.5% were treated with breast conservation therapy. The remaining 15.5% had metastatic disease at presentation. The median follow-up is 36 months (range, 3–240; Table 2). At the time of study, the majority of patients were either alive with disease (46%) or dead of disease (15%). The remaining cohort showed no evidence of disease (39%; Table 2).

IHC evidence of MMRd

We next sought to define whether breast cancers developing in the context of Lynch syndrome germline variants would harbor the hallmark features of MMRd. IHC analysis of 12 breast cancers (Table 1) with available material revealed loss of MMR proteins in five cases (42%), all of which are ER-positive/HER-2 negative breast cancers. Case 4, harboring an MLH1 c.954delC frameshift deletion, demonstrated loss of MLH1 and PMS2 with partial loss of MSH6 protein (Fig. 1AF). Loss of MLH1 and PMS2 protein was also observed in case 13, harboring an MLH1 deletion. Deficiency of MSH2 and MSH6 protein was seen in two cases (cases 10 and 11), with missense mutations in MSH2. Isolated protein loss of MSH6 was observed in case 6, harboring a frameshift mutation resulting in a premature termination codon 12 amino acids downstream of p. Leu1330Valfs*12.

Figure 1.

Clinicopathologic and genomic characterization of breast cancers in patients with Lynch syndrome germline pathogenic variants. A, Representative photomicrographs of case 4, including IHC analysis of MLH1 (B), PMS2 (C), MSH2 (D), MSH6 (E), and PD-L1 (SP142; F). Scale bars = 100 μmol/L. G, Non-synonymous somatic mutations and gene copy-number alterations identified by MSK-IMPACT targeted sequencing. Cases are shown in columns and genes in rows. Phenobars (top) provide information about DNA MMR protein expression, ER/PR/HER2 status, histologic type, histologic grade, stromal TILs, TMB, and dominant mutational signatures. Germline and somatic mutations are color coded according to the legend (right).

Figure 1.

Clinicopathologic and genomic characterization of breast cancers in patients with Lynch syndrome germline pathogenic variants. A, Representative photomicrographs of case 4, including IHC analysis of MLH1 (B), PMS2 (C), MSH2 (D), MSH6 (E), and PD-L1 (SP142; F). Scale bars = 100 μmol/L. G, Non-synonymous somatic mutations and gene copy-number alterations identified by MSK-IMPACT targeted sequencing. Cases are shown in columns and genes in rows. Phenobars (top) provide information about DNA MMR protein expression, ER/PR/HER2 status, histologic type, histologic grade, stromal TILs, TMB, and dominant mutational signatures. Germline and somatic mutations are color coded according to the legend (right).

Close modal

Genomic evidence of MMRd

MSK-IMPACT analysis of 11 breast cancers revealed a repertoire of somatic mutations consistent with that reported for invasive breast cancers, including TP53 (54%), PIK3CA (45%), and GATA3 (27%; Fig. 1G; Supplementary Table S1). Three of four breast cancers that demonstrated MMR protein loss by IHC showed two hits in the proposed Lynch syndrome gene. Two cases (cases 4 and 13) showed loss of heterozygosity in an MMR gene, whereas case 6 harbored a second somatic inactivating mutation in MSH6 (p. Phe1088Leufs; Fig. 1G). Case 10 (germline MSH2) lacked evidence of biallelic inactivation despite loss of MSH2 and MSH6 proteins by IHC. Thus, inactivation of the gene may be the result of another inactivating regulatory process such as somatic hypermethylation, which has been reported to occur in Lynch syndrome individuals with MSH2 germline variants (39).

Of the four cases demonstrating MMR protein loss by IHC with adequate materials for sequencing, three showed evidence of MSI by PCR analysis of five microsatellite loci and one yielded an inconclusive result (Fig. 1G; Table 3). Three of four cases displayed high TMB (i.e., > 10 mutations per Mb; ref. 40) and either indeterminate (three cases) or high (one case) MSISensor scores (Fig. 1G; Table 3). The sole case (case 10) deemed MSI-H (score of > 10 by MSISensor) was a 63-year-old female with a missense mutation in the MSH2 gene. The tumor was also MSI by PCR (Table 3). Both patient and family members had history of Lynch syndrome–related cancers. The tumor was a moderately differentiated ER-positive/HER2-negative invasive ductal carcinoma with low TILs (2%). IHC analysis for PD-L1 was negative. The patient underwent breast conservation surgery followed by radiotherapy and hormonal therapy. The patient currently has no evidence of disease after 144 months of clinical follow-up.

Table 3.

Summary of MMR IHC, MSI PCR, TMB, and MSI Score/Sensor in Lynch syndrome cohort.

CaseMMR status (IHC)Second hit in MMR genes (MSK-IMPACT)MSI status (PCR)Signature category by Exposure_SigMATMB score (mut/Mb)MSI scoreMSI sensor
Retained No MSS Aging 5.3 1.07 Stable (liver metastasis) 
Retained No MSS HRD 3.5 0.05 Stable 
Retained Yes (LOH) MSS N/A 2.6 0.58 Stable 
Loss of MLH1 and PMS2, partial loss of MSH6 Yes (LOH) MSI MSI 16.7 7.24 Indeterminate 
Retained No MSS APOBEC 3.5 N/A 
Loss of MSH6 Yes (Somatic) Indeterminate MSI 20.2 3.56 Indeterminate 
N/A N/A MSS Sig 17 3.5 0.25 Stable 
Retained No MSS Aging 3.5 0.12 Stable 
Retained N/A N/A N/A N/A N/A N/A 
10 Loss of MSH2 and MSH6 No MSI MSI 24.84 11.5 High 
11 Loss of MSH2 and MSH6 N/A N/A N/A N/A N/A N/A 
12 Retained No MSS N/A 1.05 0.57 Stable 
13 Loss of MLH1 and PMS2 Yes (LOH) MSI MSI 8.39 7.68 Indeterminate 
CaseMMR status (IHC)Second hit in MMR genes (MSK-IMPACT)MSI status (PCR)Signature category by Exposure_SigMATMB score (mut/Mb)MSI scoreMSI sensor
Retained No MSS Aging 5.3 1.07 Stable (liver metastasis) 
Retained No MSS HRD 3.5 0.05 Stable 
Retained Yes (LOH) MSS N/A 2.6 0.58 Stable 
Loss of MLH1 and PMS2, partial loss of MSH6 Yes (LOH) MSI MSI 16.7 7.24 Indeterminate 
Retained No MSS APOBEC 3.5 N/A 
Loss of MSH6 Yes (Somatic) Indeterminate MSI 20.2 3.56 Indeterminate 
N/A N/A MSS Sig 17 3.5 0.25 Stable 
Retained No MSS Aging 3.5 0.12 Stable 
Retained N/A N/A N/A N/A N/A N/A 
10 Loss of MSH2 and MSH6 No MSI MSI 24.84 11.5 High 
11 Loss of MSH2 and MSH6 N/A N/A N/A N/A N/A N/A 
12 Retained No MSS N/A 1.05 0.57 Stable 
13 Loss of MLH1 and PMS2 Yes (LOH) MSI MSI 8.39 7.68 Indeterminate 

Abbreviations: HRD, homologous repair deficiency; IHC, immunohistochemistry; LOH, loss of heterozygosity; MMR, mismatch repair; MSI, microsatellite instability; MSS, microsatellite stable; N/A, not available; PCR, polymerase chain reaction; TMB, tumor mutational burden [somatic mutations per Megabase (mut/Mb)].

Nine cases were found to have a least five SNVs (81%) and were subjected to mutational signature analysis with SigMA. All cases with MMR protein loss and indeterminate or high MSISensor scores demonstrated dominant microsatellite instability signatures. Notably, three of these MMRd breast cancers displayed expression of PD-L1 (>1%) in the tumor-associated inflammatory cells with variable percentages of tumor stromal infiltrating lymphocytes (mean, 14.3; range, 2–25%; Fig. 1G).

Given the higher frequency of MMRd/MSI (4/11, 36%) observed in the Lynch syndrome breast cancer cohort, and prior observations of MMRd in <2% of all breast cancers (10), we compared the TMBs, MSISensor scores, and dominant mutational signatures to a 1:3 cohort of breast cancers (n = 33) matched according to age, tumor type, HR profile, and menopausal status, respectively (Supplementary Table S2). Breast cancers in the Lynch syndrome cohort (n = 11) displayed a higher TMB (P = 0.003, Mann–Whitney U test; Supplementary Fig. S1A) and had MSI scores showing a positive trend toward significance (P = 0.069, Mann–Whitney U test; Supplementary Fig. S1B) when compared with control-matched breast cancers (n = 33). We also observed an enrichment in dominant MSI mutational signatures in tumors harboring at least five SNVs were also enriched in Lynch syndrome breast cancers (n = 11) relative to control-matched breast cancers (n = 16, P = 0.040, Fisher exact test; Supplementary Fig. S1C). We next compared the genomic characteristics of Lynch syndrome breast cancers relative to the control-matched breast cancer cohort (Supplementary Fig. S2). No statistically significant differences in single-gene comparisons were identified. Given the small sample size of Lynch syndrome breast cancers included in this study, further analyses to define differences in mutational frequencies between Lynch syndrome breast cancers and non–Lynch syndrome breast cancers are warranted.

In summary, five of 13 cases (38.5%) with tissue available for IHC and/or MSK-IMPACT displayed features of MMRd or MSI-H (summarized in Table 3).

Immunotherapy in patient with Lynch syndrome with metastatic MMRd breast cancer

Given that MSI-H has been approved as a pan-cancer biomarker of benefit from checkpoint inhibitors (41, 42) we queried whether any patients with Lynch syndrome with MMRd tumors had received immunotherapy. A 64-year-old female (case 6), with a germline MSH6 frameshift mutation and remote history of an ER-positive breast cancer 18 years prior, developed a contralateral ER-positive/HER2-negative/MMRd/PD-L1–positive breast cancer with synchronous lymph node and extensive bone metastases involving both the pelvis and lumbar spine (Fig. 2AF). Anti-PD1 therapy (pembrolizumab) was initiated in the palliative setting with complete pathologic response at 5 months on PET imaging (Fig. 2G). The patient has no radiologic evidence of disease 13 months after initiation of therapy.

Figure 2.

Metastatic breast cancer (case 6) in Lynch syndrome patient treated with immune checkpoint inhibitor. A, Timeline of disease progression, including initial breast cancer diagnosis, development of contralateral breast cancer/metastatic disease and diagnosis of Lynch syndrome, and treatment interval with anti-PD1 therapy. Representative hematoxylin-and-eosin photomicrographs of the tumor (B), ER (C), MSH6 (D), and PD-L1 (SP142; E). Scale bars = 100 μmol/L. F, Representation of the mutational spectrum using the conventional 96 codon-context mutation type classification. This classification is based on the six substitution subtypes: C>A, C>G, C>T, T>A, T>C, and T>G, as well as the nucleotides immediately 5′ and 3′ to the mutation. G, PET scan of lymph node (top left) and pelvic metastasis (bottom left) before treatment. Complete PET response of lymph node (top right) and pelvis (bottom right) after treatment.

Figure 2.

Metastatic breast cancer (case 6) in Lynch syndrome patient treated with immune checkpoint inhibitor. A, Timeline of disease progression, including initial breast cancer diagnosis, development of contralateral breast cancer/metastatic disease and diagnosis of Lynch syndrome, and treatment interval with anti-PD1 therapy. Representative hematoxylin-and-eosin photomicrographs of the tumor (B), ER (C), MSH6 (D), and PD-L1 (SP142; E). Scale bars = 100 μmol/L. F, Representation of the mutational spectrum using the conventional 96 codon-context mutation type classification. This classification is based on the six substitution subtypes: C>A, C>G, C>T, T>A, T>C, and T>G, as well as the nucleotides immediately 5′ and 3′ to the mutation. G, PET scan of lymph node (top left) and pelvic metastasis (bottom left) before treatment. Complete PET response of lymph node (top right) and pelvis (bottom right) after treatment.

Close modal

We explored the clinical, morphologic, and genomic characteristics of breast cancers occurring in individuals with Lynch syndrome from a single tertiary cancer center. Our study revealed 5 of 13 breast cancers in individuals with Lynch syndrome demonstrating MMRd by either IHC, PCR, and/or multigene panel studies. We were able to identify these breast cancers using a large sequencing pool from >20,000 patients who underwent germline testing at MSKCC (New York, NY). The majority were ER-positive/HER2-negative (85%), moderately to poorly differentiated breast cancers (100%). At a median follow-up of 36 months, the patients were AWD (2/5) or NED (3/5). One patient with advanced MMRd breast cancer and distant metastasis demonstrated durable and complete clinical response to immunotherapy administered in the palliative setting and currently is NED after 13 months of the initiation of immunotherapy.

Our findings provide direct evidence that a subset of primary breast cancers developing in the context of a Lynch syndrome germline pathogenic variant harbors features of MMRd, including MSI-H, in line with the previous observations showing MMRd breast cancers in the context of Lynch syndrome (8, 10). MSI-H has been approved as a pan-cancer biomarker of benefit from immune checkpoint inhibitors (41, 42). Consistent with the notion that some breast cancers developing in the context of Lynch syndrome germline pathogenic variants are bona fide MSI-H cancers, benefit from immune checkpoint inhibition was observed in case 6, as expected. Further studies beyond the case highlighted here are warranted.

MSISensor scores of ≥10 have been shown to reliably identify MSI-H status in solid tumors from various primary sites (34, 43). Applying this criterion in identifying MSI-H breast cancers has been explored. Latham and colleagues (12) identified the Lynch syndrome in 15,045 patients who had matched tumor/normal DNA sequencing encompassing over 50 cancer types through MSK-IMPACT platform in MSKCC from January 1, 2014 to June 30, 2017. They identified Lynch syndrome in 0.3% (7/2,371) of patients with breast cancer and all seven cases were MSS tumors based on MSISensor scores of <3. In our current study, we identified 272 individuals with Lynch syndrome among 20,110 patients with cancer who had consecutive genetic testing performed through MSKCC clinical pipeline with a cut-off date of November 30, 2020. Only 3,583 patients included in our study had the clinical genetic testing performed before the cut-off date of June 30, 2017 employed in Latham's study, representing the overlapping patient population between these two cohorts (up to 3,583 of 20,110; 18%). Two patients in the population included in both studies had developed primary breast cancers (cases 2 and 3 in our study) showing no evidence of MMRd. Including the overlapping 2 patients included in Latham's and additionally identified 11 patients unique to our study, 5 patients were identified to have MMRd breast cancers based on loss of MMR proteins by IHC (5/13) and/or tumor-normal sequencing studies on 11 cases with available tissue materials (4/11) showing dominant MSI mutational signatures, high TMB, and indeterminate or high MSISensor scores. Of the nine cases subjected to mutational signature analysis by SigMA, the four MMRd breast cancers harbored 57, 52, 45, and 17 SNVs, well above the five SNV threshold required for analysis (35). These cases were found to harbor a dominant MSI mutational signature and a mutational spectrum consistent with that of MSI-H cancers (i.e., mutational signatures 20 and 21). Only one MMRd breast cancer (case 10) in our study had a MSISensor score of >10, suggesting that MMRd breast cancers may have a lower MSISensor threshold than other MSI/MMRd cancers, such as colorectal and endometrial cancers, for which the assay was initially intended to identify (34). In line with the study from Latham and colleagues (12), our results suggest that further evaluation might be indicated to determine whether current MSISensor cut-off values can faithfully identify MMRd/MSI-H breast cancers.

This study has limitations. Despite the large cohort of patients subjected to tumor-normal MSK-IMPACT sequencing, the sample size of breast cancers developing in the context of Lynch syndrome was small, consistent with the rarity of MMRd and/or MSI-H in primary breast cancers. MMRd breast cancer is reported to represent around 1.7% of all breast cancers, with a much smaller fraction involving individuals with Lynch syndrome (10). Signature analysis could only be performed in 9 of 11 patients, given that only MSK-IMPACT sequencing was performed. Mutational signature decomposition when the number of SNVs is low (e.g., in the case of cancers subjected to MSK-IMPACT containing 5–10 somatic SNVs) may have suboptimal robustness. Although a substantial proportion of breast cancers occurring in patients with Lynch syndrome likely constitute sporadic breast cancers developing in the context of Lynch syndrome, whole-genome sequencing, to ascertain the frequency of MMRd mutational signatures, as well as MMR IHC analysis, to define the loss of MMR proteins, of consecutive breast cancers from patients with Lynch syndrome is warranted.

Although our study is not intended to address whether breast cancer belongs in the spectrum of Lynch syndrome–related cancers, it is important to note that at the population level, there appears to be no increased risk for breast cancer in Lynch syndrome individuals with MLH1, MSH2, and PMS2 variants (44, 45). Individuals with MSH6 variants may confer a slightly higher risk of developing breast cancer; however, significance was not achieved in a large case–control study (44). Currently, breast cancers are not considered Lynch syndrome–associated cancers per current NCCN guidelines due to confounding data regarding risk in patients with Lynch syndrome (46), and breast cancers developing in the context of Lynch syndrome are currently not included in the special management of hereditary breast cancers (47). It is plausible that the remaining tumors in our cohort likely correspond to sporadic breast cancers arising in Lynch syndrome individuals, as reported previously (48). With recent studies supporting universal genetic testing for patients with breast cancer ≤60 (49), it remains uncertain whether MMR genes should be added to the testing panel. Our results emphasize, however, that in up to 4 in 10 patients with Lynch syndrome who develop breast cancers, their tumors may be etiologically linked with MMRd caused by the pathogenic germline variant affecting MLH1, MSH2, MSH6, or PMS2 genes. These findings support the contention that MMR IHC analysis and/or MSI assessment by PCR or multigene sequencing should be performed in the tumor specimens from patients with Lynch syndrome who develop breast cancer.

In conclusion, we demonstrated pathologic and genomic features of MMRd/MSI-H in a subset of breast cancers arising in patients with Lynch syndrome, establishing a likely etiologic link between the germline alteration and hallmark features of MMRd/MSI in this context. Breast cancers occurring in Lynch syndrome should be tested for MMRd by either IHC or genomic sequencing to identify the patients who may benefit from immunotherapy.

F. Pareja reports grants from NCI/NIH outside the submitted work. P. Razavi reports grants from Grail, ArcherDx, Illumina, and Epic Sciences; grants and personal fees from Novartis; personal fees from AstraZeneca, Foundation Medicine, Tempus Labs, and Natera outside the submitted work. M.E. Robson reports personal fees from Change Healthcare outside the submitted work. J.Z. Drago reports personal fees from Biotheranostics outside the submitted work. H.Y. Wen reports personal fees from Merck and AstraZeneca outside the submitted work. L. Zhang reports other support from Decipher Medicine outside the submitted work. B. Weigelt reports personal fees from Repare Therapeutics outside the submitted work. J.S. Reis-Filho reports personal fees from Paige, Repare Therapeutics, Grupo Oncoclinicas, Roche Tissue Diagnostics, Novartis, Genentech, Roche, In Vicro, and Eli-Lilly outside the submitted work. H. Zhang reports personal fees from Roche/Genentech outside the submitted work. No disclosures were reported by the other authors.

C.J. Schwartz: Conceptualization, resources, formal analysis, methodology, writing–original draft, writing–review and editing. E.M. da Silva: Conceptualization, data curation, methodology. A. Marra: Conceptualization, data curation, formal analysis. A.M. Gazzo: Data curation, methodology. P. Selenica: Data curation, methodology. V.K. Rai: Formal analysis. D. Mandelker: Conceptualization, data curation, supervision, methodology. F. Pareja: Conceptualization, writing–review and editing. M. Misyura: Data curation, formal analysis. T.M. D'Alfonso: Conceptualization, writing–review and editing. E. Brogi: Conceptualization, writing–review and editing. P. Drullinsky: Conceptualization. P. Razavi: Conceptualization, writing–review and editing. M.E. Robson: Conceptualization. J.Z. Drago: Conceptualization, writing–review and editing. H.Y. Wen: Conceptualization, formal analysis, writing–review and editing. L. Zhang: Conceptualization. B. Weigelt: Conceptualization, writing–review and editing. J. Shia: Conceptualization, supervision, writing–review and editing. J.S. Reis-Filho: Conceptualization, resources, data curation, writing–review and editing. H. Zhang: Conceptualization, data curation, formal analysis, supervision, writing–review and editing.

This work was funded in part by the Marie-Josée and Henry R. Kravis Center for Molecular Oncology and the NCI Cancer Center Core grant #P30-CA008748. F. Pareja is partially funded by an NIH K12 CA184746 grant. F. Pareja, B. Weigelt, and J.S. Reis-Filho are funded in part by the NIH/NCI P50 CA247749 01 grant. E.M. da Silva is partially funded by the MSK-MIND grant. J.S. Reis-Filho is funded in part by a grant from the Breast Cancer Research Foundation.

The funders had no role in the design of the study; the collection, analysis, and interpretation of the data; the writing of the article; or the decision to submit the article.

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.
Vasen
HF
,
Watson
P
,
Mecklin
JP
,
Lynch
HT
. 
New clinical criteria for hereditary nonpolyposis colorectal cancer (HNPCC, Lynch syndrome) proposed by the International Collaborative group on HNPCC
.
Gastroenterology
1999
;
116
:
1453
6
.
2.
Win
AK
,
Young
JP
,
Lindor
NM
,
Tucker
KM
,
Ahnen
DJ
,
Young
GP
, et al
Colorectal and other cancer risks for carriers and noncarriers from families with a DNA mismatch repair gene mutation: a prospective cohort study
.
J Clin Oncol
2012
;
30
:
958
64
.
3.
Pande
M
,
Wei
C
,
Chen
J
,
Amos
CI
,
Lynch
PM
,
Lu
KH
, et al
Cancer spectrum in DNA mismatch repair gene mutation carriers: results from a hospital based Lynch syndrome registry
.
Fam Cancer
2012
;
11
:
441
7
.
4.
Risinger
JI
,
Barrett
JC
,
Watson
P
,
Lynch
HT
,
Boyd
J
. 
Molecular genetic evidence of the occurrence of breast cancer as an integral tumor in patients with the hereditary nonpolyposis colorectal carcinoma syndrome
.
Cancer
1996
;
77
:
1836
43
.
5.
Bergthorsson
JT
,
Egilsson
V
,
Gudmundsson
J
,
Arason
A
,
Ingvarsson
S
. 
Identification of a breast tumor with microsatellite instability in a potential carrier of the hereditary non-polyposis colon cancer trait
.
Clin Genet
1995
;
47
:
305
10
.
6.
Ford
JM
. 
Is breast cancer a part of Lynch syndrome?
Breast Cancer Res
2012
;
14
:
110
.
7.
Stoll
J
,
Rosenthal
E
,
Cummings
S
,
Willmott
J
,
Bernhise
R
,
Kupfer
SS
, et al
No evidence of increased risk of breast cancer in women with Lynch syndrome identified by multigene panel testing
.
JCO Precis Oncol
2020
;
4
:
51
60
.
8.
Lotsari
JE
,
Gylling
A
,
Abdel-Rahman
WM
,
Nieminen
TT
,
Aittomäki
K
,
Friman
M
, et al
Breast carcinoma and Lynch syndrome: molecular analysis of tumors arising in mutation carriers, non-carriers, and sporadic cases
.
Breast Cancer Res
2012
;
14
:
R90
.
9.
Walsh
MD
,
Buchanan
DD
,
Cummings
MC
,
Pearson
S-A
,
Arnold
ST
,
Clendenning
M
, et al
Lynch syndrome-associated breast cancers: clinicopathologic characteristics of a case series from the colon cancer family registry
.
Clin Cancer Res
2010
;
16
:
2214
24
.
10.
Davies
H
,
Morganella
S
,
Purdie
CA
,
Jang
SJ
,
Borgen
E
,
Russnes
H
, et al
Whole-genome sequencing reveals breast cancers with mismatch repair deficiency
.
Cancer Res
2017
;
77
:
4755
62
.
11.
Müller
A
,
Edmonston
TB
,
Corao
DA
,
Rose
DG
,
Palazzo
JP
,
Becker
H
, et al
Exclusion of breast cancer as an integral tumor of hereditary nonpolyposis colorectal cancer
.
Cancer Res
2002
;
62
:
1014
9
.
12.
Latham
A
,
Srinivasan
P
,
Kemel
Y
,
Shia
J
,
Bandlamudi
C
,
Mandelker
D
, et al
Microsatellite instability is associated with the presence of lynch syndrome pan-cancer
.
J Clin Oncol
2019
;
37
:
286
95
.
13.
Le
DT
,
Uram
JN
,
Wang
H
,
Bartlett
BR
,
Kemberling
H
,
Eyring
AD
, et al
PD-1 blockade in tumors with mismatch-repair deficiency
.
N Engl J Med
2015
;
372
:
2509
20
.
14.
Mandelker
D
,
Zhang
L
,
Kemel
Y
,
Stadler
ZK
,
Joseph
V
,
Zehir
A
, et al
Mutation detection in patients with advanced cancer by universal sequencing of cancer-related genes in tumor and normal DNA vs guideline-based germline testing
.
JAMA
2017
;
318
:
825
35
.
15.
Richards
S
,
Aziz
N
,
Bale
S
,
Bick
D
,
Das
S
,
Gastier-Foster
J
, et al
Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology
.
Genet Med
2015
;
17
:
405
24
.
16.
Elston
CW
,
Ellis
IO
. 
Pathological prognostic factors in breast cancer. I. The value of histological grade in breast cancer: experience from a large study with long-term follow-up
.
Histopathology
1991
;
19
:
403
10
.
17.
Ng
CK
,
Martelotto
LG
,
Gauthier
A
,
Wen
H-C
,
Piscuoglio
S
,
Lim
RS
, et al
Intra-tumor genetic heterogeneity and alternative driver genetic alterations in breast cancers with heterogeneous HER2 gene amplification
.
Genome Biol
2015
;
16
:
107
.
18.
Hammond
MEH
,
Hayes
DF
,
Dowsett
M
,
Allred
DC
,
Hagerty
KL
,
Badve
S
, et al
American Society of Clinical Oncology/College Of American Pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer
.
J Clin Oncol
2010
;
28
:
2784
95
.
19.
Wolff
AC
,
Hammond
MEH
,
Hicks
DG
,
Dowsett
M
,
McShane
LM
,
Allison
KH
, et al
Recommendations for human epidermal growth factor receptor 2 testing in breast cancer: American Society of Clinical Oncology/College of American Pathologists clinical practice guideline update
.
J Clin Oncol
2013
;
31
:
3997
4013
.
20.
Wen
YH
,
Brogi
E
,
Zeng
Z
,
Akram
M
,
Catalano
J
,
Paty
PB
, et al
DNA mismatch repair deficiency in breast carcinoma: a pilot study of triple-negative and non–triple-negative tumors
.
Am J Surg Pathol
2012
;
36
:
1700
8
.
21.
Reis
H
,
Serrette
R
,
Posada
J
,
Lu
V
,
Chen
Y-B
,
Gopalan
A
, et al
PD-L1 expression in urothelial carcinoma with predominant or pure variant histology: concordance among 3 commonly used and commercially available antibodies
.
Am J Surg Pathol
2019
;
43
:
920
7
.
22.
Hoda
RS
,
Brogi
E
,
Dos Anjos
CH
,
Grabenstetter
A
,
Ventura
K
,
Patil
S
, et al
Clinical and pathologic features associated with PD-L1 (Sp142) expression in stromal tumor-infiltrating immune cells of triple-negative breast carcinoma
.
Mod Pathol
2020
;
33
:
2221
32
.
23.
Hoda
RS
,
Brogi
E
,
D'Alfonso
TM
,
Grabenstetter
A
,
Giri
D
,
Hanna
MG
, et al
Interobserver variation of PD-L1 SP142 immunohistochemistry interpretation in breast carcinoma: a study of 79 cases using whole slide imaging
.
Arch Pathol Lab Med
2021
;
145
:
1132
7
.
24.
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
.
25.
Da Cruz Paula
A
,
da Silva
EM
,
Segura
SE
,
Pareja
F
,
Bi
R
,
Selenica
P
, et al
Genomic profiling of primary and recurrent adult granulosa cell tumors of the ovary
.
Mod Pathol
2020
;
33
:
1606
17
.
26.
Cibulskis
K
,
Lawrence
MS
,
Carter
SL
,
Sivachenko
A
,
Jaffe
D
,
Sougnez
C
, et al
Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples
.
Nat Biotechnol
2013
;
31
:
213
9
.
27.
Saunders
CT
,
Wong
WS
,
Swamy
S
,
Becq
J
,
Murray
LJ
,
Cheetham
RK
. 
Strelka: accurate somatic small-variant calling from sequenced tumor-normal sample pairs
.
Bioinformatics
2012
;
28
:
1811
7
.
28.
Koboldt
DC
,
Zhang
Q
,
Larson
DE
,
Shen
D
,
McLellan
MD
,
Lin
L
, et al
VarScan 2: somatic mutation and copy number alteration discovery in cancer by exome sequencing
.
Genome Res
2012
;
22
:
568
76
.
29.
Narzisi
G
,
Corvelo
A
,
Arora
K
,
Bergmann
EA
,
Shah
M
,
Musunuri
R
, et al
Genome-wide somatic variant calling using localized colored de Bruijn graphs
.
Commun Biol
2018
;
1
:
20
.
30.
Narzisi
G
,
O'Rawe
JA
,
Iossifov
I
,
Fang
H
,
Lee
YH
,
Wang
ZH
, et al
Accurate de novo and transmitted indel detection in exome-capture data using microassembly
.
Nat Methods
2014
;
11
:
1033
6
.
31.
Pareja
F
,
Lee
JY
,
Brown
DN
,
Piscuoglio
S
,
Gularte-Mérida
R
,
Selenica
P
, et al
The genomic landscape of mucinous breast cancer
.
J Natl Cancer Inst
2019
;
111
:
737
41
.
32.
Shen
R
,
Seshan
VE
. 
FACETS: allele-specific copy number and clonal heterogeneity analysis tool for high-throughput DNA sequencing
.
Nucleic Acids Res
2016
;
44
:
e131
.
33.
Chang
MT
,
Bhattarai
TS
,
Schram
AM
,
Bielski
CM
,
Donoghue
MTA
,
Jonsson
P
, et al
Accelerating discovery of functional mutant alleles in cancer
.
Cancer Discov
2018
;
8
:
174
83
.
34.
Middha
S
,
Zhang
L
,
Nafa
K
,
Jayakumaran
G
,
Wong
D
,
Kim
HR
, et al
Reliable pan-cancer microsatellite instability assessment by using targeted next-generation sequencing data
.
JCO Precis Oncol
2017
;
2017
:
PO.17.00084
.
35.
Gulhan
DC
,
Lee
JJ-K
,
Melloni
GEM
,
Cortés-Ciriano
I
,
Park
PJ
. 
Detecting the mutational signature of homologous recombination deficiency in clinical samples
.
Nat Genet
2019
;
51
:
912
9
.
36.
Moukarzel
LA
,
Da Cruz Paula
A
,
Ferrando
L
,
Hoang
T
,
Sebastiao
APM
,
Pareja
F
, et al
Clonal relationship and directionality of progression of synchronous endometrial and ovarian carcinomas in patients with DNA mismatch repair-deficiency associated syndromes
.
Mod Pathol
2021
;
34
:
994
1007
.
37.
Bergstrom
EN
,
Huang
MN
,
Mahto
U
,
Barnes
M
,
Stratton
MR
,
Rozen
SG
, et al
SigProfilerMatrixGenerator: a tool for visualizing and exploring patterns of small mutational events
.
BMC Genomics
2019
;
20
:
685
.
38.
Razavi
P
,
Chang
MT
,
Xu
G
,
Bandlamudi
C
,
Ross
DS
,
Vasan
N
, et al
The genomic landscape of endocrine-resistant advanced breast cancers
.
Cancer Cell
2018
;
34
:
427
38
.
39.
Nagasaka
T
,
Rhees
J
,
Kloor
M
,
Gebert
J
,
Naomoto
Y
,
Boland
CR
, et al
Somatic hypermethylation of MSH2 is a frequent event in Lynch syndrome colorectal cancers
.
Cancer Res
2010
;
70
:
3098
108
.
40.
Shao
C
,
Li
G
,
Huang
L
,
Pruitt
S
,
Castellanos
E
,
Frampton
G
, et al
Prevalence of high tumor mutational burden and association with survival in patients with less common solid tumors
.
JAMA Netw Open
2020
;
3
:
e2025109
.
41.
Schmid
P
,
Adams
S
,
Rugo
HS
,
Schneeweiss
A
,
Barrios
CH
,
Iwata
H
, et al
Atezolizumab and nab-paclitaxel in advanced triple-negative breast cancer
.
N Engl J Med
2018
;
379
:
2108
21
.
42.
Bouffet
E
,
Larouche
V
,
Campbell
BB
,
Merico
D
,
de Borja
R
,
Aronson
M
, et al
Immune checkpoint inhibition for hypermutant glioblastoma multiforme resulting from germline biallelic mismatch repair deficiency
.
J Clin Oncol
2016
;
34
:
2206
11
.
43.
Stadler
ZK
,
Battaglin
F
,
Middha
S
,
Hechtman
JF
,
Tran
C
,
Cercek
A
, et al
Reliable detection of mismatch repair deficiency in colorectal cancers using mutational load in next-generation sequencing panels
.
J Clin Oncol
2016
;
34
:
2141
7
.
44.
Breast Cancer Association Consortium
,
Dorling
L
,
Carvalho
S
,
Allen
J
,
González-Neira
A
,
Luccarini
C
, et al
Breast cancer risk genes — association analysis in more than 113,000 women
.
N Engl J Med
2021
;
384
:
428
39
.
45.
Hu
C
,
Hart
SN
,
Gnanaolivu
R
,
Huang
H
,
Lee
KY
,
Na
J
, et al
A population-based study of genes previously implicated in breast cancer
.
N Engl J Med
2021
;
384
:
440
51
.
46.
Gupta
S
,
Provenzale
D
,
Llor
X
,
Halverson
AL
,
Grady
W
,
Chung
DC
, et al
NCCN guidelines insights: Genetic/familial high-risk assessment: Colorectal, version 2.2019
.
J Natl Compr Canc Netw
2009
;
17
:
1032
41
.
47.
Tung
NM
,
Boughey
JC
,
Pierce
LJ
,
Robson
ME
,
Bedrosian
I
,
Dietz
JR
, et al
Management of hereditary breast cancer: American Society of Clinical Oncology, American Society for Radiation Oncology, and Society of Surgical Oncology guideline
.
J Clin Oncol
2020
;
38
:
2080
106
.
48.
D'Arcy
C
,
Wen
YH
,
Stadler
ZK
,
Brogi
E
,
Shia
J
. 
Synchronous breast cancers with different morphologic and molecular phenotypes occurring in Lynch syndrome: what does the heterogeneity imply?
Am J Surg Pathol
2011
;
35
:
1743
8
.
49.
Desai
NV
,
Yadav
S
,
Batalini
F
,
Couch
FJ
,
Tung
NM
. 
Germline genetic testing in breast cancer: rationale for the testing of all women diagnosed by the age of 60 years and for risk-based testing of those older than 60 years
.
Cancer
2021
;
127
:
828
33
.