Molecular analyses of triple-negative breast cancer in the young and elderly

Mattias Aine, Ceren Boyaci, Johan Hartman, Jari Häkkinen, Shamik Mitra, Ana Bosch Campos, Emma Nimeus, Anna Ehinger, Johan Vallon-Christersson, Åke Borg, Johan Staaf, Mattias Aine, Ceren Boyaci, Johan Hartman, Jari Häkkinen, Shamik Mitra, Ana Bosch Campos, Emma Nimeus, Anna Ehinger, Johan Vallon-Christersson, Åke Borg, Johan Staaf

Abstract

Background: Breast cancer in young adults has been implicated with a worse outcome. Analyses of genomic traits associated with age have been heterogenous, likely because of an incomplete accounting for underlying molecular subtypes. We aimed to resolve whether triple-negative breast cancer (TNBC) in younger versus older patients represent similar or different molecular diseases in the context of genetic and transcriptional subtypes and immune cell infiltration.

Patients and methods: In total, 237 patients from a reported population-based south Swedish TNBC cohort profiled by RNA sequencing and whole-genome sequencing (WGS) were included. Patients were binned in 10-year intervals. Complimentary PD-L1 and CD20 immunohistochemistry and estimation of tumor-infiltrating lymphocytes (TILs) were performed. Cases were analyzed for differences in patient outcome, genomic, transcriptional, and immune landscape features versus age at diagnosis. Additionally, 560 public WGS breast cancer profiles were used for validation.

Results: Median age at diagnosis was 62 years (range 26-91). Age was not associated with invasive disease-free survival or overall survival after adjuvant chemotherapy. Among the BRCA1-deficient cases (82/237), 90% were diagnosed before the age of 70 and were predominantly of the basal-like subtype. In the full TNBC cohort, reported associations of patient age with changes in Ki67 expression, PIK3CA mutations, and a luminal androgen receptor subtype were confirmed. Within DNA repair deficiency or gene expression defined molecular subgroups, age-related alterations in, e.g., overall gene expression, immune cell marker gene expression, genetic mutational and rearrangement signatures, amount of copy number alterations, and tumor mutational burden did, however, not appear distinct. Similar non-significant associations for genetic alterations with age were obtained for other breast cancer subgroups in public WGS data. Consistent with age-related immunosenescence, TIL counts decreased linearly with patient age across different genetic TNBC subtypes.

Conclusions: Age-related alterations in TNBC, as well as breast cancer in general, need to be viewed in the context of underlying genomic phenotypes. Based on this notion, age at diagnosis alone does not appear to provide an additional layer of biological complexity above that of proposed genetic and transcriptional phenotypes of TNBC. Consequently, treatment decisions should be less influenced by age and more driven by tumor biology.

Trial registration: ClinicalTrials.gov NCT02306096.

Keywords: Age at diagnosis; Gene expression; Mutational signatures; Mutations; PD-L1; Patient outcome; TILs; Triple-negative breast cancer.

Conflict of interest statement

The authors declare that they have no competing interests with exception of Johan Hartman who has received speakers’ honoraria and travel support from Roche, advisory board fees from MSD, Novartis, and Roche, and institutional research grants from Cepheid and Novartis, and is the co-founder and shareholder of Stratipath AB.

Figures

Fig. 1
Fig. 1
Study scheme. a Analyses performed in the SCAN-B TNBC cohort together with investigated main sample groups. b Analyses performed in the external Nik-Zainal et al. [3] cohort together with investigated main sample groups. In both panels, sample size numbers for patient groups refer to the largest set of patients available for at least one of the specified analyses. Specific sample size numbers are provided in the detailed results and the “Methods” section. References to the main figures and tables presenting results are provided for each analysis. HRD+: HRDetect-high, HRD-: HRDetect-low, TILs: tumor-infiltrating lymphocytes, TMB: tumor mutational burden, CNA: copy number alteration, Lum A: Luminal A, Lum B: Luminal B
Fig. 2
Fig. 2
Patient age versus BRCA1 deficiency and gene expression subtypes in TNBC. a Cumulative proportion of patients with BRCA1 deficiency (BRCA1 hypermethylation or BRCA1-null tumors) and non-BRCA1-deficient patients versus age at diagnosis. Red triangles indicate age at diagnosis for 50%, 80%, and 90% of BRCA1-deficient patients. b Cumulative proportions of patients in HRDetect groups versus age at diagnosis. c Principal component analysis of gene expression data for 232 SCAN-B cases using 19,102 RefSeq genes and different molecular and clinicopathological factors, including age at diagnosis (years: Age) and stratified age groups (10-year intervals: Age groups). d Cumulative proportions of PAM50 subtypes versus age at diagnosis. e Cumulative proportions of IntClust 10 subtypes versus age at diagnosis. f Cumulative proportions of TNBCtype subtypes versus age at diagnosis. g Heatmap of 1179 genes differentially expressed between six 10-year interval age groups in 232 SCAN-B cases. Hierarchical clustering of cases (columns) and genes (rows) were performed using Pearson correlation as distance metric and ward.D linkage on mean-centered log2 transformed data with an offset of 0.1. The six top clusters were identified and labeled. MKI67: Ki67. Steroid response: Scores according to the steroid response metagene [40]. h From left to right: Estimations of tumor cell content from WGS (ASCAT method), epithelial, stromal, B cell lymphocyte, and endothelial cell proportions from CIBERSORTx versus the hierarchical clusters in g. For age group definitions, “[” equals ≥, “)” equals <, and “]” equals ≤ for the value specified next to it
Fig. 3
Fig. 3
Immune cell landscape of TNBC with respect to age at diagnosis. a CIBERSORTx estimated B cell proportions per sample versus stratified age groups in all cases (left), HRDetect-high, HRDetect-low, and PAM50 basal-like cases (right). Top axes indicate group sizes. Two-sided p values calculated using Kruskal-Wallis test. Linear regression modeling showing p value and slope coefficient (k) when using B cell proportion and continuous age in the model. b Heatmap of 102 immune cell marker genes in 232 SCAN-B cases using Pearson correlation and ward.D linkage. Hierarchical clustering of cases (columns) and genes (rows) was performed using Pearson correlation as distance metric and ward.D linkage on mean-centered log2 transformed data with an offset of 0.1. c TIL percentage estimated from whole-slide hemotoxylin and eosin-stained sections versus stratified age groups in all cases (left), HRDetect-high, HRDetect-low, and PAM50 basal-like cases (right). Top axes indicate group sizes. Two-sided p values calculated using Kruskal-Wallis test. Linear regression modeling showing p value and slope coefficient (k) when using TIL percentage and continuous age in the model. For age group definitions, “[” equals ≥, “)” equals <, and “]” equals ≤ for the value specified next to it. In panels a and c, separate results of a sensitivity analysis for trend are reported in red due to very small sample numbers across the full stratified age range
Fig. 4
Fig. 4
Copy number alterations versus age at diagnosis in TNBC. a Copy number landscape of HRDetect-high patients < 50 years at diagnosis versus > 70 years at diagnosis. b Difference in amplification frequency of CCND1, CCNE1, EGFR, and MCL1 with age groups when analyzed in the total SCAN-B cohort. Two-sided p values calculated using chi-square test for trends in proportions. c Difference in mutation frequency of PIK3CA and TP53 with age groups when analyzed in the total SCAN-B cohort. Two-sided p values calculated using chi-square test for trends in proportions. d Proportions of amplified cases for CCND1, CCNE1, EGFR, and MCL1 according to HRDetect classification. e Proportions of mutated cases for PIK3CA and TP53 according to HRDetect (left), PAM50 (center), and TNBCtype (right) classifications. For age group definitions, “[” equals ≥, “)” equals <, and “]” equals ≤ for the value specified next to it
Fig. 5
Fig. 5
Composite view of molecular and genetic phenotypes versus age at diagnosis in TNBC. a Integrative view of gene expression subtypes (PAM50 basal-like, LAR, IntClust 4, 10), HRD classification, BRCA1, BRCA2, PALB2, RAD51C, MCL1, CCND1, CCNE1, EGFR, PIK3CA, and TP53 alterations, mutational signatures (S1-S26), rearrangement signatures (RS1-RS6), patterns of insertion, and deletions versus age groups stratified by an underlying BRCA1 deficiency, BRCA2 deficiency, and HRDetect classification. BRCA1-null, BRCA2-null, and PALB2-null imply biallelic loss of the gene based on WGS. b Illustration of signature patterns in BRCA1-deficient tumors (light gray) and HRDetect-low/intermediate cases (white) stratified by age for proportion of insertions (left), rearrangement signature 6 (RS6, center), and mutational substitution signature 3 (S3, right, refitted by SigFit). RS6 is characterized by clustered rearrangements typically found in cases with driver amplifications, while S3 is associated with BRCA1/2 deficiency [41]. Top axes indicate group sizes. Top axes indicate group sizes. Two-sided p values are calculated by Kruskal-Wallis test per group. For age group definitions, “[” equals ≥, “)” equals <, and “]” equals ≤ for the value specified next to it

References

    1. Gluz O, Liedtke C, Gottschalk N, Pusztai L, Nitz U, Harbeck N. Triple-negative breast cancer--current status and future directions. Ann Oncol. 2009;20(12):1913–1927. doi: 10.1093/annonc/mdp492.
    1. Foulkes WD, Smith IE, Reis-Filho JS. Triple-negative breast cancer. N Engl J Med. 2010;363(20):1938–1948. doi: 10.1056/NEJMra1001389.
    1. Nik-Zainal S, Davies H, Staaf J, Ramakrishna M, Glodzik D, Zou X, Martincorena I, Alexandrov LB, Martin S, Wedge DC, et al. Landscape of somatic mutations in 560 breast cancer whole-genome sequences. Nature. 2016;534(7605):47–54. doi: 10.1038/nature17676.
    1. Jiang YZ, Ma D, Suo C, Shi J, Xue M, Hu X, Xiao Y, Yu KD, Liu YR, Yu Y, et al. Genomic and transcriptomic landscape of triple-negative breast cancers: subtypes and treatment strategies. Cancer Cell. 2019;35(3):428–440. doi: 10.1016/j.ccell.2019.02.001.
    1. Bareche Y, Venet D, Ignatiadis M, Aftimos P, Piccart M, Rothe F, Sotiriou C. Unravelling triple-negative breast cancer molecular heterogeneity using an integrative multiomic analysis. Ann Oncol. 2018;29(4):895–902. doi: 10.1093/annonc/mdy024.
    1. Lehmann BD, Bauer JA, Chen X, Sanders ME, Chakravarthy AB, Shyr Y, Pietenpol JA. Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest. 2011;121(7):2750–2767. doi: 10.1172/JCI45014.
    1. Pareja F, Reis-Filho JS. Triple-negative breast cancers - a panoply of cancer types. Nat Rev Clin Oncol. 2018;15(6):347–348. doi: 10.1038/s41571-018-0001-7.
    1. Turner NC, Reis-Filho JS. Tackling the diversity of triple-negative breast cancer. Clin Cancer Res. 2013;19(23):6380–6388. doi: 10.1158/1078-0432.CCR-13-0915.
    1. Staaf J, Glodzik D, Bosch A, Vallon-Christersson J, Reutersward C, Hakkinen J, Degasperi A, Amarante TD, Saal LH, Hegardt C, et al. Whole-genome sequencing of triple-negative breast cancers in a population-based clinical study. Nat Med. 2019;25(10):1526–1533. doi: 10.1038/s41591-019-0582-4.
    1. Radosa JC, Eaton A, Stempel M, Khander A, Liedtke C, Solomayer EF, Karsten M, Pilewskie M, Morrow M, King TA. Evaluation of local and distant recurrence patterns in patients with triple-negative breast cancer according to age. Ann Surg Oncol. 2017;24(3):698–704. doi: 10.1245/s10434-016-5631-3.
    1. Kim EK, Noh WC, Han W, Noh DY. Prognostic significance of young age (<35 years) by subtype based on ER, PR, and HER2 status in breast cancer: a nationwide registry-based study. World J Surg. 2011;35(6):1244–1253. doi: 10.1007/s00268-011-1071-1.
    1. Liedtke C, Hess KR, Karn T, Rody A, Kiesel L, Hortobagyi GN, Pusztai L, Gonzalez-Angulo AM. The prognostic impact of age in patients with triple-negative breast cancer. Breast Cancer Res Treat. 2013;138(2):591–599. doi: 10.1007/s10549-013-2461-x.
    1. Partridge AH, Hughes ME, Warner ET, Ottesen RA, Wong YN, Edge SB, Theriault RL, Blayney DW, Niland JC, Winer EP, et al. Subtype-dependent relationship between young age at diagnosis and breast cancer survival. J Clin Oncol. 2016;34(27):3308–3314. doi: 10.1200/JCO.2015.65.8013.
    1. Syed BM, Green AR, Nolan CC, Morgan DA, Ellis IO, Cheung KL. Biological characteristics and clinical outcome of triple negative primary breast cancer in older women - comparison with their younger counterparts. Plos One. 2014;9(7):e100573. doi: 10.1371/journal.pone.0100573.
    1. Tichy JR, Lim E, Anders CK. Breast cancer in adolescents and young adults: a review with a focus on biology. J Natl Compr Cancer Netw. 2013;11(9):1060–1069. doi: 10.6004/jnccn.2013.0128.
    1. Liao S, Hartmaier RJ, McGuire KP, Puhalla SL, Luthra S, Chandran UR, Ma T, Bhargava R, Modugno F, Davidson NE, et al. The molecular landscape of premenopausal breast cancer. Breast Cancer Res. 2015;17:104. doi: 10.1186/s13058-015-0618-8.
    1. Anders CK, Fan C, Parker JS, Carey LA, Blackwell KL, Klauber-DeMore N, Perou CM. Breast carcinomas arising at a young age: unique biology or a surrogate for aggressive intrinsic subtypes? J Clin Oncol. 2011;29(1):e18–e20. doi: 10.1200/JCO.2010.28.9199.
    1. Yau C, Fedele V, Roydasgupta R, Fridlyand J, Hubbard A, Gray JW, Chew K, Dairkee SH, Moore DH, Schittulli F, et al. Aging impacts transcriptomes but not genomes of hormone-dependent breast cancers. Breast Cancer Res. 2007;9(5):R59. doi: 10.1186/bcr1765.
    1. Ma D, Jiang YZ, Xiao Y, Xie MD, Zhao S, Jin X, Xu XE, Shao ZM. Integrated molecular profiling of young and elderly patients with triple-negative breast cancer indicates different biological bases and clinical management strategies. Cancer. 2020;126(14):3209–3218. doi: 10.1002/cncr.32922.
    1. Azim HA, Jr, Nguyen B, Brohee S, Zoppoli G, Sotiriou C. Genomic aberrations in young and elderly breast cancer patients. BMC Med. 2015;13:266. doi: 10.1186/s12916-015-0504-3.
    1. Anders CK, Hsu DS, Broadwater G, Acharya CR, Foekens JA, Zhang Y, Wang Y, Marcom PK, Marks JR, Febbo PG, et al. Young age at diagnosis correlates with worse prognosis and defines a subset of breast cancers with shared patterns of gene expression. J Clin Oncol. 2008;26(20):3324–3330. doi: 10.1200/JCO.2007.14.2471.
    1. Saal LH, Vallon-Christersson J, Hakkinen J, Hegardt C, Grabau D, Winter C, Brueffer C, Tang MH, Reutersward C, Schulz R, et al. The Sweden Cancerome Analysis Network - Breast (SCAN-B) Initiative: a large-scale multicenter infrastructure towards implementation of breast cancer genomic analyses in the clinical routine. Genome Med. 2015;7(1):20. doi: 10.1186/s13073-015-0131-9.
    1. SCAN-B. . Accessed 19 Oct 2020.
    1. Ryden L, Loman N, Larsson C, Hegardt C, Vallon-Christersson J, Malmberg M, Lindman H, Ehinger A, Saal LH, Borg A. Minimizing inequality in access to precision medicine in breast cancer by real-time population-based molecular analysis in the SCAN-B initiative. Br J Surg. 2018;105(2):e158–e168. doi: 10.1002/bjs.10741.
    1. Glodzik D, Bosch A, Hartman J, Aine M, Vallon-Christersson J, Reutersward C, Karlsson A, Mitra S, Nimeus E, Holm K, et al. Comprehensive molecular comparison of BRCA1 hypermethylated and BRCA1 mutated triple negative breast cancers. Nat Commun. 2020;11(1):3747. doi: 10.1038/s41467-020-17537-2.
    1. Davies H, Glodzik D, Morganella S, Yates LR, Staaf J, Zou X, Ramakrishna M, Martin S, Boyault S, Sieuwerts AM, et al. HRDetect is a predictor of BRCA1 and BRCA2 deficiency based on mutational signatures. Nat Med. 2017;23(4):517–525. doi: 10.1038/nm.4292.
    1. Telli ML, Timms KM, Reid J, Hennessy B, Mills GB, Jensen KC, Szallasi Z, Barry WT, Winer EP, Tung NM, 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(15):3764–3773. doi: 10.1158/1078-0432.CCR-15-2477.
    1. Burrell RA, McClelland SE, Endesfelder D, Groth P, Weller MC, Shaikh N, Domingo E, Kanu N, Dewhurst SM, Gronroos E, et al. Replication stress links structural and numerical cancer chromosomal instability. Nature. 2013;494(7438):492–496. doi: 10.1038/nature11935.
    1. Ma D, Jiang YZ, Liu XY, Liu YR, Shao ZM. Clinical and molecular relevance of mutant-allele tumor heterogeneity in breast cancer. Breast Cancer Res Treat. 2017;162(1):39–48. doi: 10.1007/s10549-017-4113-z.
    1. Gori K, Baez-Ortega A: sigfit: flexible Bayesian inference of mutational signatures bioRxiv 2018, doi:10.1101/372896.
    1. Brueffer C, Vallon-Christersson J, Grabau D, Ehinger A, Häkkinen J, Hegardt C, Malina J, Chen Y, Bendahl P-O, Manjer J, et al. Clinical value of RNA sequencing–based classifiers for prediction of the five conventional breast cancer biomarkers: a report from the population-based multicenter Sweden Cancerome Analysis Network—Breast Initiative. JCO Precision Oncol. 2018;2(2):1–18.
    1. Curtis C, Shah SP, Chin SF, Turashvili G, Rueda OM, Dunning MJ, Speed D, Lynch AG, Samarajiwa S, Yuan Y, et al. The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature. 2012;486(7403):346–352. doi: 10.1038/nature10983.
    1. Ali HR, Rueda OM, Chin SF, Curtis C, Dunning MJ, Aparicio SA, Caldas C. Genome-driven integrated classification of breast cancer validated in over 7,500 samples. Genome Biol. 2014;15(8):431. doi: 10.1186/s13059-014-0431-1.
    1. Chen X, Li J, Gray WH, Lehmann BD, Bauer JA, Shyr Y, Pietenpol JA. TNBCtype: a subtyping tool for triple-negative breast cancer. Cancer Inform. 2012;11:147–156. doi: 10.4137/CIN.S9983.
    1. Lauss M, Visne I, Kriegner A, Ringner M, Jonsson G, Hoglund M. Monitoring of technical variation in quantitative high-throughput datasets. Cancer Inform. 2013;12:193–201. doi: 10.4137/CIN.S12862.
    1. Newman AM, Steen CB, Liu CL, Gentles AJ, Chaudhuri AA, Scherer F, Khodadoust MS, Esfahani MS, Luca BA, Steiner D, et al. Determining cell type abundance and expression from bulk tissues with digital cytometry. Nat Biotechnol. 2019;37(7):773–782. doi: 10.1038/s41587-019-0114-2.
    1. Schenck RO, Lakatos E, Gatenbee C, Graham TA, Anderson ARA. NeoPredPipe: high-throughput neoantigen prediction and recognition potential pipeline. BMC Bioinformatics. 2019;20(1):264. doi: 10.1186/s12859-019-2876-4.
    1. Shukla SA, Rooney MS, Rajasagi M, Tiao G, Dixon PM, Lawrence MS, Stevens J, Lane WJ, Dellagatta JL, Steelman S, et al. Comprehensive analysis of cancer-associated somatic mutations in class I HLA genes. Nat Biotechnol. 2015;33(11):1152–1158. doi: 10.1038/nbt.3344.
    1. Hudis CA, Barlow WE, Costantino JP, Gray RJ, Pritchard KI, Chapman JA, Sparano JA, Hunsberger S, Enos RA, Gelber RD, et al. Proposal for standardized definitions for efficacy end points in adjuvant breast cancer trials: the STEEP system. J Clin Oncol. 2007;25(15):2127–2132. doi: 10.1200/JCO.2006.10.3523.
    1. Fredlund E, Staaf J, Rantala JK, Kallioniemi O, Borg A, Ringner M. The gene expression landscape of breast cancer is shaped by tumor protein p53 status and epithelial-mesenchymal transition. Breast Cancer Res. 2012;14(4):R113. doi: 10.1186/bcr3236.
    1. Nik-Zainal S, Morganella S. Mutational signatures in breast cancer: the problem at the DNA level. Clin Cancer Res. 2017;23(11):2617–2629. doi: 10.1158/1078-0432.CCR-16-2810.
    1. Alexandrov LB, Jones PH, Wedge DC, Sale JE, Campbell PJ, Nik-Zainal S, Stratton MR. Clock-like mutational processes in human somatic cells. Nat Genet. 2015;47(12):1402–1407. doi: 10.1038/ng.3441.
    1. Quist J, Mirza H, Cheang MCU, Telli ML, O'Shaughnessy JA, Lord CJ, Tutt ANJ, Grigoriadis A. A four-gene decision tree signature classification of triple-negative breast cancer: implications for targeted therapeutics. Mol Cancer Ther. 2019;18(1):204–212. doi: 10.1158/1535-7163.MCT-18-0243.
    1. Burstein MD, Tsimelzon A, Poage GM, Covington KR, Contreras A, Fuqua SA, Savage MI, Osborne CK, Hilsenbeck SG, Chang JC, et al. Comprehensive genomic analysis identifies novel subtypes and targets of triple-negative breast cancer. Clin Cancer Res. 2015;21(7):1688–1698. doi: 10.1158/1078-0432.CCR-14-0432.
    1. Bareche Y, Buisseret L, Gruosso T, Girard E, Venet D, Dupont F, Desmedt C, Larsimont D, Park M, Rothe F, et al. Unraveling triple-negative breast cancer tumor microenvironment heterogeneity: towards an optimized treatment approach. J Natl Cancer Inst. 2020;112(7):708–719. doi: 10.1093/jnci/djz208.
    1. Desmedt C, Salgado R, Fornili M, Pruneri G, Van den Eynden G, Zoppoli G, Rothe F, Buisseret L, Garaud S, Willard-Gallo K, et al. Immune infiltration in invasive lobular breast cancer. J Natl Cancer Inst. 2018;110(7):768–776. doi: 10.1093/jnci/djx268.
    1. Loi S, Drubay D, Adams S, Pruneri G, Francis PA, Lacroix-Triki M, Joensuu H, Dieci MV, Badve S, Demaria S, et al. Tumor-infiltrating lymphocytes and prognosis: a pooled individual patient analysis of early-stage triple-negative breast cancers. J Clin Oncol. 2019;37(7):559–569. doi: 10.1200/JCO.18.01010.
    1. Ruffell B, Au A, Rugo HS, Esserman LJ, Hwang ES, Coussens LM. Leukocyte composition of human breast cancer. Proc Natl Acad Sci U S A. 2012;109(8):2796–2801. doi: 10.1073/pnas.1104303108.
    1. Ali HR, Provenzano E, Dawson SJ, Blows FM, Liu B, Shah M, Earl HM, Poole CJ, Hiller L, Dunn JA, et al. Association between CD8+ T-cell infiltration and breast cancer survival in 12,439 patients. Ann Oncol. 2014;25(8):1536–1543. doi: 10.1093/annonc/mdu191.
    1. Woo SR, Fuertes MB, Corrales L, Spranger S, Furdyna MJ, Leung MY, Duggan R, Wang Y, Barber GN, Fitzgerald KA, et al. STING-dependent cytosolic DNA sensing mediates innate immune recognition of immunogenic tumors. Immunity. 2014;41(5):830–842. doi: 10.1016/j.immuni.2014.10.017.
    1. Weiskopf D, Weinberger B, Grubeck-Loebenstein B. The aging of the immune system. Transpl Int. 2009;22(11):1041–1050. doi: 10.1111/j.1432-2277.2009.00927.x.
    1. Lehmann BD, Abramson VG, Sanders ME, Mayer EL, Haddad TC, Nanda R, Van Poznak C, Storniolo AM, Nangia JR, Gonzalez-Ericsson PI, et al. TBCRC 032 IB/II multicenter study: molecular insights to AR antagonist and PI3K inhibitor efficacy in patients with AR(+) metastatic triple-negative breast cancer. Clin Cancer Res. 2020;26(9):2111–2123. doi: 10.1158/1078-0432.CCR-19-2170.
    1. Gerratana L, Basile D, Buono G, De Placido S, Giuliano M, Minichillo S, Coinu A, Martorana F, De Santo I, Del Mastro L, et al. Androgen receptor in triple negative breast cancer: a potential target for the targetless subtype. Cancer Treat Rev. 2018;68:102–110. doi: 10.1016/j.ctrv.2018.06.005.

Source: PubMed

3
Abonnieren