Tutt, A.
Tovey, H.
Cheang, M.C.
Kernaghan, S.
Kilburn, L.
Gazinska, P.
Owen, J.
Abraham, J.
Barrett, S.
Barrett-Lee, P.
Brown, R.
Chan, S.
Dowsett, M.
Flanagan, J.M.
Fox, L.
Grigoriadis, A.
Gutin, A.
Harper-Wynne, C.
Hatton, M.Q.
Hoadley, K.A.
Parikh, J.
Parker, P.
Perou, C.M.
Roylance, R.
Shah, V.
Shaw, A.
Smith, I.E.
Timms, K.M.
Wardley, A.M.
Wilson, G.
Gillett, C.
Lanchbury, J.S.
Ashworth, A.
Rahman, N.
Harries, M.
Ellis, P.
Pinder, S.E.
Bliss, J.M.
(2018). Carboplatin in BRCA1/2-mutated and triple-negative breast cancer BRCAness subgroups: the TNT Trial. Nature medicine,
Vol.24
(5),
pp. 628-637.
show abstract
Germline mutations in BRCA1/2 predispose individuals to breast cancer (termed germline-mutated BRCA1/2 breast cancer, gBRCA-BC) by impairing homologous recombination (HR) and causing genomic instability. HR also repairs DNA lesions caused by platinum agents and PARP inhibitors. Triple-negative breast cancers (TNBCs) harbor subpopulations with BRCA1/2 mutations, hypothesized to be especially platinum-sensitive. Cancers in putative 'BRCAness' subgroups-tumors with BRCA1 methylation; low levels of BRCA1 mRNA (BRCA1 mRNA-low); or mutational signatures for HR deficiency and those with basal phenotypes-may also be sensitive to platinum. We assessed the efficacy of carboplatin and another mechanistically distinct therapy, docetaxel, in a phase 3 trial in subjects with unselected advanced TNBC. A prespecified protocol enabled biomarker-treatment interaction analyses in gBRCA-BC and BRCAness subgroups. The primary endpoint was objective response rate (ORR). In the unselected population (376 subjects; 188 carboplatin, 188 docetaxel), carboplatin was not more active than docetaxel (ORR, 31.4% versus 34.0%, respectively; P = 0.66). In contrast, in subjects with gBRCA-BC, carboplatin had double the ORR of docetaxel (68% versus 33%, respectively; biomarker, treatment interaction P = 0.01). Such benefit was not observed for subjects with BRCA1 methylation, BRCA1 mRNA-low tumors or a high score in a Myriad HRD assay. Significant interaction between treatment and the basal-like subtype was driven by high docetaxel response in the nonbasal subgroup. We conclude that patients with advanced TNBC benefit from characterization of BRCA1/2 mutations, but not BRCA1 methylation or Myriad HRD analyses, to inform choices on platinum-based chemotherapy. Additionally, gene expression analysis of basal-like cancers may also influence treatment selection..
Perez, E.A.
Ballman, K.V.
Mashadi-Hossein, A.
Tenner, K.S.
Kachergus, J.M.
Norton, N.
Necela, B.M.
Carr, J.M.
Ferree, S.
Perou, C.M.
Baehner, F.
Cheang, M.C.
Thompson, E.A.
(2017). Intrinsic Subtype and Therapeutic Response Among HER2-Positive Breast Tumors from the NCCTG (Alliance) N9831 Trial. J natl cancer inst,
Vol.109
(2),
pp. 1-8.
show abstract
Background: Genomic data from human epidermal growth factor receptor 2-positive (HER2+) tumors were analyzed to assess the association between intrinsic subtype and clinical outcome in a large, well-annotated patient cohort. Methods: Samples from the NCCTG (Alliance) N9831 trial were analyzed using the Prosigna algorithm on the NanoString platform to define intrinsic subtype, risk of recurrence scores, and risk categories for 1392 HER2+ tumors. Subtypes were evaluated for recurrence-free survival (RFS) using Kaplan-Meier and Cox model analysis following adjuvant chemotherapy (n = 484) or chemotherapy plus trastuzumab (n = 908). All statistical tests were two-sided. Results: Patients with HER2+ tumors from N9831 were primarily scored as HER2-enriched (72.1%). These individuals received statistically significant benefit from trastuzumab (hazard ratio [HR] = 0.68, 95% confidence interval [CI] = 0.52 to 0.89, P = .005), as did the patients (291 of 1392) with luminal-type tumors (HR = 0.52, 95% CI = 0.32 to 0.85, P = .01). Patients with basal-like tumors (97 of 1392) did not have statistically significantly better RFS when treated with trastuzumab and chemotherapy compared with chemotherapy alone (HR = 1.06, 95% CI = 0.53 to 2.13, P = .87). Conclusions: The majority of clinically defined HER2-positive tumors were classified as HER2-enriched or luminal using the Prosigna algorithm. Intrinsic subtype alone cannot replace conventional histopathological evaluation of HER2 status because many tumors that are classified as luminal A or luminal B will benefit from adjuvant trastuzumab if that subtype is accompanied by HER2 overexpression. However, among tumors that overexpress HER2, we speculate that assessment of intrinsic subtype may influence treatment, particularly with respect to evaluating alternative therapeutic approaches for that subset of HER2-positive tumors of the basal-like subtype..
Prat, A.
Cheang, M.C.
Galván, P.
Nuciforo, P.
Paré, L.
Adamo, B.
Muñoz, M.
Viladot, M.
Press, M.F.
Gagnon, R.
Ellis, C.
Johnston, S.
(2016). Prognostic Value of Intrinsic Subtypes in Hormone Receptor-Positive Metastatic Breast Cancer Treated With Letrozole With or Without Lapatinib. Jama oncol,
Vol.2
(10),
pp. 1287-1294.
show abstract
Importance: The value of the intrinsic subtypes of breast cancer (luminal A, luminal B, human epidermal growth factor receptor 2 [currently known as ERBB2, but referred to as HER2 in this study]-enriched, and basal-like) in the metastatic setting is currently unknown. Objective: To evaluate the association of the intrinsic subtypes of breast cancer with outcome and/or benefit in hormone receptor (HR)-positive metastatic breast cancer. Design, Setting, and Participants: Unplanned retrospective analysis of 821 tumor samples (85.7% primary and 14.3% metastatic) from the EGF30008 phase 3 clinical trial (NCT00073528), in which postmenopausal women with HR-positive invasive breast cancer and no prior therapy for advanced or metastatic disease were randomized to letrozole with or without lapatinib, an epidermal growth factor receptor (EGFR)/HER2 tyrosine kinase inhibitor. Tumor samples were classified into each subtype using the research-based PAM50 classifier. Prior neoadjuvant/adjuvant antiestrogen therapy was allowed. Patients with extensive symptomatic visceral disease were excluded. Treatment effects were evaluated using interaction tests. Main Outcomes and Measures: Primary and secondary end points were progression-free survival and overall survival. Results: The median (range) age was 62 (31-94) years. Intrinsic subtype was the strongest prognostic factor independently associated with progression-free survival and overall survival in all patients, and in patients with HER2-negative (n = 644) or HER2-positive (n = 157) diseases. Median progression-free survival differed across the intrinsic subtypes of clinically HER2-negative disease: luminal A (16.9 [95% CI, 14.1-19.9] months), luminal B (11.0 [95% CI, 9.6-13.6] months), HER2-enriched (4.7 [95% CI, 2.7-10.8] months), and basal-like (4.1 [95% CI, 2.5-13.8] months). Median OS also differed across the intrinsic subtypes: luminal A (45 [95% CI, 41-not applicable {NA}] months), luminal B (37 [95% CI, 31-42] months), HER2-enriched (16 [95% CI, 10-NA] months), and basal-like (23 [95% CI, 12-NA] months). Patients with HER2-negative/HER2-enriched disease benefited from lapatinib therapy (median PFS, 6.49 vs 2.60 months; progression-free survival hazard ratio, 0.238 [95% CI, 0.066-0.863]; interaction P = .02). Conclusions and Relevance: This is the first study to reveal an association between intrinsic subtype and outcome in first-line HR-positive metastatic breast cancer. Patients with HR-positive/HER2-negative disease with a HER2-enriched profile may benefit from lapatinib in combination with endocrine therapy. The clinical value of intrinsic subtyping in hormone receptor-positive metastatic breast cancer warrants further investigation, but patients with luminal A/HER2-negative metastatic breast cancer might be good candidates for letrozole monotherapy in the first-line setting regardless of visceral disease and number of metastases..
Sun, Z.
Prat, A.
Cheang, M.C.
Gelber, R.D.
Perou, C.M.
(2015). Chemotherapy benefit for 'ER-positive' breast cancer and contamination of nonluminal subtypes—waiting for TAILORx and RxPONDER. Ann oncol,
Vol.26
(1),
pp. 70-74.
show abstract
BACKGROUND: Retrospective analyses of NSABP B20 and SWOG 8814 showed a large benefit of chemotherapy in patients with ER-positive tumors and high OncotypeDX Recurrence Score (RS≥31). However, it might be possible that both studies may be contaminated by non-luminal tumors, especially in high-risk RS group. METHODS: We conducted simulations in order to obtain a better understanding of how the NSABP B20 and SWOG 8814 results would have been if non-luminal breast cancer would have been excluded. Simulations were done separately for the node-negative and node-positive cohorts. RESULTS AND CONCLUSION: The results of the simulations suggest that the non-luminal tumors are augmenting the apparent benefit of chemotherapy, but do not appear to be responsible for the entire effect. These simulations could provide information about the potential influence of contamination by unexpected tumor subtypes on the future results of TAILORx and RxPONDER clinical trials..
Cheang, M.C.
Martin, M.
Nielsen, T.O.
Prat, A.
Voduc, D.
Rodriguez-Lescure, A.
Ruiz, A.
Chia, S.
Shepherd, L.
Ruiz-Borrego, M.
Calvo, L.
Alba, E.
Carrasco, E.
Caballero, R.
Tu, D.
Pritchard, K.I.
Levine, M.N.
Bramwell, V.H.
Parker, J.
Bernard, P.S.
Ellis, M.J.
Perou, C.M.
Di Leo, A.
Carey, L.A.
(2015). Defining Breast Cancer Intrinsic Subtypes by Quantitative Receptor Expression. The oncologist,
Vol.20
(5),
pp. 474-482.
show abstract
Abstract
Purpose.
To determine intrinsic breast cancer subtypes represented within categories defined by quantitative hormone receptor (HR) and HER2 expression.
Methods.
We merged 1,557 cases from three randomized phase III trials into a single data set. These breast tumors were centrally reviewed in each trial for quantitative ER, PR, and HER2 expression by immunohistochemistry (IHC) stain and by reverse transcription-quantitative polymerase chain reaction (RT-qPCR), with intrinsic subtyping by research-based PAM50 RT-qPCR assay.
Results.
Among 283 HER2-negative tumors with <1% HR expression by IHC, 207 (73%) were basal-like; other subtypes, particularly HER2-enriched (48, 17%), were present. Among the 1,298 HER2-negative tumors, borderline HR (1%–9% staining) was uncommon (n = 39), and these tumors were heterogeneous: 17 (44%) luminal A/B, 12 (31%) HER2-enriched, and only 7 (18%) basal-like. Including them in the definition of triple-negative breast cancer significantly diminished enrichment for basal-like cancer (p < .05). Among 106 HER2-positive tumors with <1% HR expression by IHC, the HER2-enriched subtype was the most frequent (87, 82%), whereas among 127 HER2-positive tumors with strong HR (>10%) expression, only 69 (54%) were HER2-enriched and 55 (43%) were luminal (39 luminal B, 16 luminal A). Quantitative HR expression by RT-qPCR gave similar results. Regardless of methodology, basal-like cases seldom expressed ER/ESR1 or PR/PGR and were associated with the lowest expression level of HER2/ERBB2 relative to other subtypes.
Conclusion.
Significant discordance remains between clinical assay-defined subsets and intrinsic subtype. For identifying basal-like breast cancer, the optimal HR IHC cut point was <1%, matching the American Society of Clinical Oncology and College of American Pathologists guidelines. Tumors with borderline HR staining are molecularly diverse and may require additional assays to clarify underlying biology.
.
Garcia-Murillas, I.
Schiavon, G.
Weigelt, B.
Ng, C.
Hrebien, S.
Cutts, R.J.
Cheang, M.
Osin, P.
Nerurkar, A.
Kozarewa, I.
Garrido, J.A.
Dowsett, M.
Reis-Filho, J.S.
Smith, I.E.
Turner, N.C.
(2015). Mutation tracking in circulating tumor DNA predicts relapse in early breast cancer. Science translational medicine,
Vol.7
(302),
pp. 302ra133-?.
show abstract
The identification of early-stage breast cancer patients at high risk of relapse would allow tailoring of adjuvant therapy approaches. We assessed whether analysis of circulating tumor DNA (ctDNA) in plasma can be used to monitor for minimal residual disease (MRD) in breast cancer. In a prospective cohort of 55 early breast cancer patients receiving neoadjuvant chemotherapy, detection of ctDNA in plasma after completion of apparently curative treatment-either at a single postsurgical time point or with serial follow-up plasma samples-predicted metastatic relapse with high accuracy [hazard ratio, 25.1 (confidence interval, 4.08 to 130.5; log-rank P < 0.0001) or 12.0 (confidence interval, 3.36 to 43.07; log-rank P < 0.0001), respectively]. Mutation tracking in serial samples increased sensitivity for the prediction of relapse, with a median lead time of 7.9 months over clinical relapse. We further demonstrated that targeted capture sequencing analysis of ctDNA could define the genetic events of MRD, and that MRD sequencing predicted the genetic events of the subsequent metastatic relapse more accurately than sequencing of the primary cancer. Mutation tracking can therefore identify early breast cancer patients at high risk of relapse. Subsequent adjuvant therapeutic interventions could be tailored to the genetic events present in the MRD, a therapeutic approach that could in part combat the challenge posed by intratumor genetic heterogeneity..
Voduc, K.D.
Nielsen, T.O.
Perou, C.M.
Harrell, J.C.
Fan, C.
Kennecke, H.
Minn, A.J.
Cryns, V.L.
Cheang, M.C.
(2015). αB-crystallin Expression in Breast Cancer is Associated with Brain Metastasis. Npj breast cancer,
Vol.1.
show abstract
Background/objectives The molecular chaperone αB-crystallin is expressed in estrogen receptor, progesterone receptor and human epidermal growth factor receptor-2 "triple-negative" breast carcinomas and promotes brain and lung metastasis. We examined αB-crystallin expression in primary breast carcinomas with metastatic data to evaluate its association with prognosis and site-specific metastases.Methods αB-crystallin gene ( CRYAB ) expression was examined using publically available global-gene expression data (n=855 breast tumors) with first site of distant metastasis information ("855Met"). αB-crystallin protein expression was determined by immunohistochemistry using the clinically annotated tissue microarray (n=3987 breast tumors) from British Columbia Cancer Agency (BCCA). Kaplan-Meier and multivariable Cox regression analyses were used to evaluate the prognostic value of αB-crystallin. Multivariable logistic regression analysis was used to evaluate risks of αB-crystallin and other markers for site of metastasis.Results In the 855Met dataset, αB-crystallin gene ( CRYAB) expression was an independent predictor of brain as the first distant site of relapse (HR = 1.2, (95% CI 1.0-1.4), P = 0.021). In the BCCA series, αB-crystallin protein expression was an independent prognostic marker of poor breast cancer specific survival (HR = 1.3, (95% CI 1.1-1.6), P = 0.014). Among patients with metastases, αB-crystallin was the strongest independent predictor of brain metastasis (OR = 2.99 (95% CI 1.83-4.89), P < 0.0001) and the only independent predictor of brain as the first site of distant metastasis (OR = 3.15 (95% CI1.43-6.95), P = 0.005). αB-crystallin was also associated with worse survival (3.0 versus 4.7 months, P = 0.007).Conclusions αB-crystallin is a promising biomarker to identify breast cancer patients at high risk for early relapse in the brain, independent of ER and HER2 status..
Prat, A.
Cheang, M.C.
Martin, M.
Parker, J.S.
Carrasco, E.
Caballero, R.
Tyldesley, S.
Gelmon, K.
Bernard, P.S.
Nielsen, T.O.
Perou, C.M.
(2013). Prognostic Significance of Progesterone Receptor-Positive Tumor Cells Within Immunohistochemically Defined Luminal A Breast Cancer. Journal of clinical oncology,
Vol.31
(2),
pp. 203-209.
Cheang, M.C.
Voduc, K.D.
Tu, D.
Jiang, S.
Leung, S.
Chia, S.K.
Shepherd, L.E.
Levine, M.N.
Pritchard, K.I.
Davies, S.
Stijleman, I.J.
Davis, C.
Ebbert, M.T.
Parker, J.S.
Ellis, M.J.
Bernard, P.S.
Perou, C.M.
Nielsen, T.O.
(2012). Responsiveness of Intrinsic Subtypes to Adjuvant Anthracycline Substitution in the NCIC CTG MA 5 Randomized Trial. Clinical cancer research,
Vol.18
(8),
pp. 2402-2412.
Cheang, M.C.
Chia, S.K.
Voduc, D.
Gao, D.
Leung, S.
Snider, J.
Watson, M.
Davies, S.
Bernard, P.S.
Parker, J.S.
Perou, C.M.
Ellis, M.J.
Nielsen, T.O.
(2009). Ki67 Index, HER2 Status, and Prognosis of Patients With Luminal B Breast Cancer. Jnci-journal of the national cancer institute,
Vol.101
(10),
pp. 736-750.
Cheang, M.C.
Voduc, D.
Bajdik, C.
Leung, S.
McKinney, S.
Chia, S.K.
Perou, C.M.
Nielsen, T.O.
(2008). Basal-like breast cancer defined by five biomarkers has superior prognostic value then triple-negative phenotype. Clinical cancer research,
Vol.14
(5),
pp. 1368-1376.
Parker, J.
Mullins, M.
Cheang, M.C.
Davies, S.
Mardis, E.
Nielsen, T.O.
Ellis, M.J.
Marron, S.
Perou, C.M.
Bernard, P.S.
(2008). A supervised risk predictor of breast cancer based on biological subtypes. Journal of clinical oncology,
Vol.26
(15).
Cheang, M.C.
van de Rijn, M.
Nielsen, T.O.
(2008). Gene expression profiling of breast cancer. Annual review of pathology-mechanisms of disease,
Vol.3,
pp. 67-97.