Hormone Receptor–Positive Breast Cancer Sensitive to Pembrolizumab: Evidence of the Pathogenicity of the MLH1 Variant 1835del3

Authors:
Henry G. Kaplan Swedish Cancer Institute, Seattle, Washington

Search for other papers by Henry G. Kaplan in
Current site
Google Scholar
PubMed
Close
 MD
,
Jeffrey R. Whiteaker Fred Hutchinson Cancer Center, Seattle, Washington

Search for other papers by Jeffrey R. Whiteaker in
Current site
Google Scholar
PubMed
Close
 PhD
,
Brianna Nelson Swedish Cancer Institute, Seattle, Washington

Search for other papers by Brianna Nelson in
Current site
Google Scholar
PubMed
Close
 MS, CGC
,
Richard G. Ivey Fred Hutchinson Cancer Center, Seattle, Washington

Search for other papers by Richard G. Ivey in
Current site
Google Scholar
PubMed
Close
 BS
,
Travis D. Lorentzen Fred Hutchinson Cancer Center, Seattle, Washington

Search for other papers by Travis D. Lorentzen in
Current site
Google Scholar
PubMed
Close
 BS
,
Uliana Voytovich Fred Hutchinson Cancer Center, Seattle, Washington

Search for other papers by Uliana Voytovich in
Current site
Google Scholar
PubMed
Close
 BS
,
Lei Zhao Fred Hutchinson Cancer Center, Seattle, Washington

Search for other papers by Lei Zhao in
Current site
Google Scholar
PubMed
Close
 BS
,
David J. Corwin CellNetix, Tukwila, Washington

Search for other papers by David J. Corwin in
Current site
Google Scholar
PubMed
Close
 MD
,
Robert Resta Swedish Cancer Institute, Seattle, Washington

Search for other papers by Robert Resta in
Current site
Google Scholar
PubMed
Close
 MS, CGC
, and
Amanda G. Paulovich Fred Hutchinson Cancer Center, Seattle, Washington

Search for other papers by Amanda G. Paulovich in
Current site
Google Scholar
PubMed
Close
 MD, PhD
Restricted access

A woman with estrogen/progesterone receptor–positive, ERBB2-negative metastatic breast cancer developed progressive disease despite treatment with multiple hormonal and chemotherapeutic modalities. She carried a germline variant of MLH1 (1835del3), also known as c.1835_1837del and v612del, the pathogenicity of which has not been conclusively determined. MLH1 staining was not seen on immunohistochemical staining of her tumor tissue. The patient experienced a >5-year dramatic response to 4 doses of pembrolizumab. Family studies revealed multiple other relatives with the MLH1 1835del3 variant, as well as multiple relatives with colon cancer. The one relative with colon cancer who underwent genetic testing demonstrated the same variant. Laboratory studies revealed that the patient’s tumor showed loss of heterozygosity (LOH) in the MLH1 region, high levels of microsatellite instability, and a high tumor mutational burden. LOH in the MLH1 region, along with the remarkable clinical response to pembrolizumab treatment and the presence of the same MLH1 variant in affected relatives, supports the hypothesis that the MLH1 1835del3 variant is pathogenic. Given the patient’s family history, this likely represents an uncommon presentation of Lynch syndrome. Physicians should be alert to evaluate patients for targetable genetic variants even in unlikely clinical situations such as the one described here.

Submitted January 10, 2023; final revision received April 11, 2023; accepted for publication May 11, 2023. Published online August 29, 2023.

Disclosures: J.R. Whiteaker has disclosed serving as a consultant for CellCarta. A.G. Paulovich has disclosed serving as a consultant for CellCarta and having an ownership interest in Precision Assays LLC. The remaining authors have disclosed that they have not received any financial considerations from any person or organization to support the preparation, analysis, results, or discussion of this article.

Funding: This work was funded by Kaplan Cancer Research Fund and in part by the National Cancer Institute of the National Institutes of Health, under the NCI Beau Biden National Cancer Moonshot (Task Order No. HHSN26100025, Applied Proteogenomics Organizational Learning and Outcomes, under Contract No. HHSN261201500003I), the NCI Clinical Proteomic Tumor Analysis Consortium (grant numbers U01CA214114 and U01CA271407), and the NCI Research Specialist program (grant number R50CA211499).

Disclaimer: The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

Correspondence: Henry G. Kaplan, MD, Swedish Cancer Institute, 1221 Madison Street, Suite 920, Seattle, WA 98104. Email: hank.kaplan@swedish.org
  • Collapse
  • Expand
  • 1.

    Cortes J, Rugo HS, Cescon DW, et al. Pembrolizumab plus chemotherapy in advanced triple negative breast cancer. N Engl J Med 2022;387:217226.

  • 2.

    Schmid P, Cortes J, Dent R, et al. Event-free survival with pembrolizumab in early triple-negative breast cancer. N Engl J Med 2022;386:556567.

  • 3.

    Dorling L, Carvalho S, Allen J, et al. Breast cancer risk genes – association analysis in more than 113,000 women. N Engl J Med 2021;384:428439.

  • 4.

    Lemery S, Keegan P, Pazdur R. First FDA approval agnostic of cancer site - when a biomarker defines the indication. N Engl J Med 2017;377:14091412.

  • 5.

    Petrelli F, Ghidini M, Ghidini A, et al. Outcomes following immune checkpoint inhibitor treatment of patients with microsatellite instability-high cancers: a systematic review and meta-analysis. JAMA Oncol 2020;6:10681071.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 6.

    Le DT, Uram JN, Wang H, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med 2015;372:25092520.

  • 7.

    Le DT, Durham JN, Smith KN, et al. Mismatch-repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017;357:409413.

  • 8.

    Marabelle A, Le DT, Ascierto PA, et al. Efficacy of pembrolizumab in patients with noncolorectal high microsatellite instability/mismatch repair-deficient cancer: results from the phase II KEYNOTE-158 study. J Clin Oncol 2020;38:110.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 9.

    Alva AS, Mangat PK, Garrett-Mayer E, et al. Pembrolizumab in patient with metastatic breast cancer with high tumor mutational burden: results from the targeted agent and profiling utilization registry (TAPUR) study. J Clin Oncol 2021;39:24432451.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 10.

    Stadler ZK, Battaglin F, Middha S, et al. Reliable detection of mismatch repair deficiency in colorectal cancers using mutational load in next generation sequencing panels. J Clin Oncol 2016;34:21412147.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 11.

    Nikitin AG, Chudakova DA, Enikeev RF, et al. Lynch syndrome germline mutations in breast cancer: next generation sequencing case-control study of 1263 participants. Front Oncol 2020;10:666.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 12.

    Win AK, Lindor NM, Jenkins MA. Risk of breast cancer in Lynch syndrome: a systematic review. Breast Cancer Res 2013;15:R27.

  • 13.

    Kohlmann W. Lynch syndrome and breast cancer risk: weighing the data. JCO Precis Oncol 2020;4:PO.19.00376.

  • 14.

    Stoll J, Rosenthal E, Cummings S, et al. No evidence of increased risk of breast cancer in women with Lynch syndrome identified by multigene panel testing. JCO Precis Oncol 2020;4:5160.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 15.

    Sorscher S. Identifying Lynch syndrome-related breast cancers. JCO Precis Oncol 2020;4:579580.

  • 16.

    Porkka NK, Olkinuora A, Kuopio T, et al. Does breast carcinoma belong to the Lynch syndrome tumor spectrum? – Somatic mutational profiles vs. ovarian and colorectal carcinomas. Oncotarget 2020;11:12441256.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 17.

    Walsh MD, Buchanan DD, Cummings MC, et al. Lynch syndrome- associated breast cancers: clinicopathologic characteristics of a case series from the colon cancer family registry. Clin Cancer Res 2010;16:22142224.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 18.

    Harkness EF, Barrow E, Newton K, et al. Lynch syndrome caused by MLH1 mutations is associated with an increased risk of breast cancer: a cohort study. J Med Genet 2015;52:553556.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 19.

    Malik SS, Masood N, Asif M, et al. Expressional analysis of MLH1 and MSH2 in breast cancer. Curr Probl Cancer 2019;43:97105.

  • 20.

    Chalmers ZR, Connelly CF, Fabrizio D, et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med 2017;9:34.

  • 21.

    Schwartz CJ, da Silva EM, Marra A, et al. Morphologic and genomic characteristics of breast cancers occurring in individuals with Lynch syndrome. Clin Cancer Res 2022;28:404413.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 22.

    Grandval P, Barouk-Simonet E, Bronner M, et al. Is the controversy on breast cancer as part of the Lynch-related tumor spectrum still open? Fam Cancer 2012;11:681683.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 23.

    Roberts ME, Jackson SA, Susswein LR, et al. MSH6 and PMS2 germ-line pathogenic variants implicated in Lynch syndrome are associated with breast cancer. Genet Med 2018;20:11671174.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 24.

    Risinger JI, Barrett JC, Watson P, et al. Molecular genetic evidence of the occurrence of breast cancer as an integral tumor in patients with the hereditary nonpolyposis colorectal carcinoma syndrome. Cancer 1996;77:18361843.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 25.

    Bergthorsson JT, Egilsson V, Gudmundsson J, et al. Identification of a breast tumor with microsatellite instability in a potential carrier of the hereditary non-polyposis colon cancer trait. Clin Genet 1995;47:305310.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 26.

    Ford JM. Is breast cancer a part of Lynch syndrome? Breast Cancer Res 2012;14:110.

  • 27.

    Lotsari JE, Gylling A, Abdel-Rahman WM, 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.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 28.

    Müller A, Edmonston TB, Corao DA, et al. Exclusion of breast cancer as an integral tumor of hereditary nonpolyposis colorectal cancer. Cancer Res 2002;62:10141019.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 29.

    Latham A, Srinivasan P, Kernel Y, et al. Microsatellite instability is associated with the presence of Lynch syndrome pan-cancer. J Clin Oncol 2019;37:286295.

  • 30.

    Whiteaker JR, Lundeen RA, Zhao L, et al. Targeted mass spectrometry enables multiplexed quantification of immunomodulatory proteins in clinical biospecimens. Front Immunol 2021;12:765898.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 31.

    Whiteaker JR, Wang T, Zhao L, et al. Targeted mass spectrometry enables quantification of novel pharmacodynamic biomarkers of ATM kinase inhibition. Cancers (Basel) 2021;13:3843.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 32.

    Burke W, Parens E, Chung WK, et al. The challenge of genetic variants of unknown clinical significance: a narrative review. Ann Intern Med 2022;175:9941000.

  • 33.

    Horimoto Y, Thinzar Hlaing M, Saeki H, et al. Microsatellite instability and mismatch repair protein expressions in lymphocyte-predominant breast cancer. Cancer Sci 2020;111:26472654.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 34.

    Ren XY, Song Y, Wang J, et al. Mismatch repair deficiency and microsatellite instability in triple-negative breast cancer: a retrospective study of 440 patients. Front Oncol 2021;11:570623.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 35.

    Vidula N, Lipman A, Kato S, et al. Detection of microsatellite instability high (MSI-H) status by targeted plasma-based genotyping in metastatic breast cancer. NPJ Breast Cancer 2022;8:117.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 36.

    Wen YH, Brogi E, Zeng Z, 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;36:17001708.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 37.

    Roberts SA, Lawrence MS, Klimczak LJ, et al. An APOBEC cytidine deaminase mutagenesis pattern is widespread in human cancers. Nat Genet 2013;45:970976.

  • 38.

    Davies H, Morganella S, Purdie CA, et al. Whole-genome sequencing reveals breast cancers with mismatch repair deficiency. Cancer Res 2017;77:47554762.

  • 39.

    Chumsri S, Sokol ES, Soyano-Muller AE, et al. Durable complete response with immune checkpoint inhibitor in breast cancer with high tumor mutational burden and APOBEC signature. J Natl Compr Canc Netw 2020;18:517521.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 40.

    Niv Y. Microsatellite instability and MLH1 promoter hypermethylation in colorectal cancer. World J Gastroenterol 2007;13:17671769.

  • 41.

    Dawood S, Schuster K, Feldman R. Comparison of tumor mutation burden and PD-L1 between 2797 primary and metastatic breast cancer samples. Ann Oncol 2018;29(Suppl 9):Abstract 382P.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 42.

    Cho YA, Lee H, Kim DG, et al. PD-L1 expression is significantly associated with tumor mutation burden and microsatellite instability score. Cancers (Basel) 2021;13:4659.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 43.

    Thompson BA, Spurdle AB, Plazzer JP, et al. Application of a 5-tiered scheme for standardized classification of 2,360 unique mismatch repair gene variants in the InSiGHT locus-specific database. Nat Genet 2014;46:107115.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 44.

    Raevaara TE, Korhonen MK, Lohi H, et al. Functional significance and clinical phenotype of nontruncating mismatch repair variants of MLH1. Gastroenterology 2005;129:537549.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 45.

    Mangold E, Pagenstecher C, Friedl W, et al. Tumours from MSH2 mutation carriers show loss of MSH2 expression but many tumours from MLH1 mutation carriers exhibit weak positive MLH1 staining. J Pathol 2005;207:385395.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 46.

    Hardt K, Heick SB, Betz B, et al. Missense variants in hMLH1 identified in patients from the German HNPCC consortium and functional studies. Fam Cancer 2011;10:273284.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 47.

    Dowty JG, Win AK, Buchanan DD, et al. Cancer risks for MLH1 and MSH2 mutation carriers. Hum Mutat 2013;34:490497.

  • 48.

    Das S, Johnson DB. Immune-related adverse events and anti-tumor efficacy of immune checkpoint inhibitors. J Immunother Cancer 2019;7:306.

  • 49.

    Lynch HT, de la Chapelle A. Hereditary colorectal cancer. N Engl J Med 2003;348:919932.

Metrics

All Time Past Year Past 30 Days
Abstract Views 0 0 0
Full Text Views 3470 2867 43
PDF Downloads 1997 1558 23
EPUB Downloads 0 0 0