760 results on '"Eric P Winer"'
Search Results
2. Survival in male breast cancer over the past 3 decades
- Author
-
José P Leone, Rachel A Freedman, Julieta Leone, Sara M Tolaney, Carlos T Vallejo, Bernardo A Leone, Eric P Winer, Nancy U Lin, and Michael J Hassett
- Subjects
Cancer Research ,Oncology - Abstract
Background Breast cancer mortality in women has declined statistically significantly over the past several years. In men, it is unclear whether survival has changed over time. We evaluated changes in breast cancer–specific survival (BCSS) and overall survival (OS) in male breast cancer over the past 3 decades. Methods We evaluated men diagnosed with breast cancer between 1988 and 2017, reported in the Surveillance, Epidemiology, and End Results registry. Patients were categorized into 3 groups by year of diagnosis: 1988-1997, 1998-2007, and 2008-2017. BCSS and OS were estimated by Kaplan-Meier, and differences between groups were compared by log-rank test. Multivariable Cox regression evaluated the independent association of year of diagnosis with BCSS and OS. All tests were 2-sided. Results We included 8481 men. Overall, BCSS at 5 years was 83.69%, 83.78%, and 84.41% in groups 1988-1997, 1998-2007, and 2008-2017, respectively (P = .86). There was no statistically significant difference in BCSS between the 3 groups within each stage of disease. Among all patients, OS at 5 years was 64.61%, 67.31%, and 69.05% in groups 1988-1997, 1998-2007, and 2008-2017, respectively (P = .01). In adjusted Cox models, each additional year of diagnosis had no statistically significant association with BCSS (hazard ratio = 1.00, 95% confidence interval = 0.99 to 1.01, P = .75), but there was statistically significant improvement in OS (hazard ratio = 0.99, 95% CI = 0.98 to 0.99, P = .009). Conclusions Over the past 3 decades, there has been no statistically significant improvement in BCSS in male breast cancer. Changes in OS over time are consistent with increasing life expectancy. Efforts to improve BCSS in male breast cancer are warranted.
- Published
- 2022
3. Clinical Efficacy and Whole-Exome Sequencing of Liquid Biopsies in a Phase IB/II Study of Bazedoxifene and Palbociclib in Advanced Hormone Receptor–Positive Breast Cancer
- Author
-
Junko Tsuji, Tianyu Li, Albert Grinshpun, Tim Coorens, Douglas Russo, Leilani Anderson, Rebecca Rees, Agostina Nardone, Candace Patterson, Niall J. Lennon, Carrie Cibulskis, Ignaty Leshchiner, Nabihah Tayob, Sara M. Tolaney, Nadine Tung, Donald P. McDonnell, Ian E. Krop, Eric P. Winer, Chip Stewart, Gad Getz, and Rinath Jeselsohn
- Subjects
Cancer Research ,Treatment Outcome ,Receptors, Estrogen ,Oncology ,Receptor, ErbB-2 ,Antineoplastic Combined Chemotherapy Protocols ,Exome Sequencing ,Liquid Biopsy ,Humans ,Female ,Breast Neoplasms ,Article - Abstract
Purpose: Sensitivity to endocrine therapy (ET) is critical for the clinical benefit from the combination of palbociclib plus ET in hormone receptor–positive/HER2-negative (HR+/HER2−) advanced breast cancer. Bazedoxifene is a third-generation selective estrogen receptor (ER) modulator and selective ER degrader with activity in preclinical models of endocrine-resistant breast cancer, including models harboring ESR1 mutations. Clinical trials in healthy women showed that bazedoxifene is well tolerated. Patients and Methods: We conducted a phase Ib/II study of bazedoxifene plus palbociclib in patients with HR+/HER2− advanced breast cancer who progressed on prior ET (N = 36; NCT02448771). Results: The study met its primary endpoint, with a clinical benefit rate of 33.3%, and the safety profile was consistent with what has previously been seen with palbociclib monotherapy. The median progression-free survival (PFS) was 3.6 months [95% confidence interval (CI), 2.0–7.2]. An activating PIK3CA mutation at baseline was associated with a shorter PFS (HR = 4.4; 95% CI, 1.5–13; P = 0.0026), but activating ESR1 mutations did not impact the PFS. Longitudinal plasma circulating tumor DNA whole-exome sequencing (WES; N = 68 plasma samples) provided an overview of the tumor heterogeneity and the subclonal genetic evolution, and identified actionable mutations acquired during treatment. Conclusions: The combination of palbociclib and bazedoxifene has clinical efficacy and an acceptable safety profile in a heavily pretreated patient population with advanced HR+/HER2− breast cancer. These results merit continued investigation of bazedoxifene in breast cancer.
- Published
- 2022
4. Preclinical and Clinical Efficacy of Trastuzumab Deruxtecan in Breast Cancer Brain Metastases
- Author
-
Sheheryar Kabraji, Jing Ni, Sarah Sammons, Tianyu Li, Amanda E.D. Van Swearingen, Yanzhi Wang, Alyssa Pereslete, Liangge Hsu, Pamela J. DiPiro, Chris Lascola, Heather Moore, Melissa Hughes, Akshara S. Raghavendra, Maria Gule-Monroe, Rashmi K. Murthy, Eric P. Winer, Carey K. Anders, Jean J. Zhao, and Nancy U. Lin
- Subjects
Cancer Research ,Immunoconjugates ,Receptor, ErbB-2 ,Brain Neoplasms ,Breast Neoplasms ,Trastuzumab ,Antibodies, Monoclonal, Humanized ,Article ,Cohort Studies ,Treatment Outcome ,Oncology ,Humans ,Female ,Camptothecin ,Prospective Studies ,Retrospective Studies - Abstract
Purpose: Brain metastases can occur in up to 50% of patients with metastatic HER2-positive breast cancer. Because patients with active brain metastases were excluded from previous pivotal clinical trials, the central nervous system (CNS) activity of the antibody–drug conjugate trastuzumab deruxtecan (T-DXd) is not well characterized. Experimental Design: We studied how T-DXd affects growth and overall survival in orthotopic patient-derived xenografts (PDX) of HER2-positive and HER2-low breast cancer brain metastases (BCBM). Separately, we evaluated the effects of T-DXd in a retrospective cohort study of 17 patients with stable or active brain metastases. Results: T-DXd inhibited tumor growth and prolonged survival in orthotopic PDX models of HER2-positive (IHC 3+) and HER2-low (IHC 2+/FISH ratio < 2) BCBMs. T-DXd reduced tumor size and prolonged survival in a T-DM1–resistant HER2-positive BCBM PDX model. In a retrospective multi-institutional cohort study of 17 patients with predominantly HER2-positive BCBMs, the CNS objective response rate (ORR) was 73% (11/15) while extracranial response rate was 45% (5/11). In the subset of patients with untreated or progressive BCBM at baseline, the CNS ORR was 70% (7/10). The median time on treatment with T-DXd was 8.9 (1.3–16.2) months, with 42% (7/17) remaining on treatment at data cutoff. Conclusions: T-DXd demonstrates evidence of CNS activity in HER2-positive and HER2-low PDX models of BCBM and preliminary evidence of clinical efficacy in a multi-institution case series of patients with BCBM. Prospective clinical trials to further evaluate CNS activity of T-DXd in patients with active brain metastases are warranted. See related commentary by Soffietti and Pellerino, p. 8
- Published
- 2022
5. Circulating Tumor DNA and Late Recurrence in High-Risk Hormone Receptor-Positive, Human Epidermal Growth Factor Receptor 2-Negative Breast Cancer
- Author
-
Marla Lipsyc-Sharf, Elza C. de Bruin, Katheryn Santos, Robert McEwen, Daniel Stetson, Ashka Patel, Gregory J. Kirkner, Melissa E. Hughes, Sara M. Tolaney, Ann H. Partridge, Ian E. Krop, Charlene Knape, Ute Feger, Giovanni Marsico, Karen Howarth, Eric P. Winer, Nancy U. Lin, and Heather A. Parsons
- Subjects
Cancer Research ,Neoplasm, Residual ,Oncology ,Receptor, ErbB-2 ,Mutation ,Biomarkers, Tumor ,Humans ,Triple Negative Breast Neoplasms ,Prospective Studies ,Neoplasm Recurrence, Local ,Circulating Tumor DNA - Abstract
PURPOSE To examine the prevalence and dynamics of circulating tumor DNA (ctDNA) and its association with metastatic recurrence in patients with high-risk early-stage hormone receptor–positive breast cancer (HR+ BC) more than 5 years from diagnosis. METHODS We enrolled 103 patients with high-risk stage II-III HR+ BC diagnosed more than 5 years prior without clinical evidence of recurrence. We performed whole-exome sequencing (WES) on primary tumor tissue to identify somatic mutations tracked via a personalized, tumor-informed ctDNA test to detect minimal residual disease (MRD). We collected plasma at the time of consent and at routine visits every 6-12 months. Patients were followed for clinical recurrence. RESULTS In total, 85 of 103 patients had sufficient tumor tissue; of them, 83 of 85 (97.6%) patients had successful whole-exome sequencing. Personalized ctDNA assays were designed targeting a median of 36 variants to test 219 plasma samples. The median time from diagnosis to first sample was 8.4 years. The median follow-up was 10.4 years from diagnosis and 2.0 years from first sample. The median number of plasma samples per patient was two. Eight patients (10%) had positive MRD testing at any time point. Six patients (7.2%) developed distant metastatic recurrence, all of whom were MRD-positive before overt clinical recurrence, with median ctDNA lead time of 12.4 months. MRD was not identified in one patient (1.2%) with local recurrence. Two of eight MRD-positive patients had not had clinical recurrence at last follow-up. CONCLUSION In this prospective study, in patients with high-risk HR+ BC in the late adjuvant setting, ctDNA was identified a median of 1 year before all cases of distant metastasis. Future studies will determine if ctDNA-guided intervention in patients with HR+ BC can alter clinical outcomes.
- Published
- 2023
6. A Distinct Chromatin State Drives Therapeutic Resistance in Invasive Lobular Breast Cancer
- Author
-
Agostina Nardone, Xintao Qiu, Sandor Spisak, Zsuzsanna Nagy, Ariel Feiglin, Avery Feit, Gabriela Cohen Feit, Yingtian Xie, Alba Font-Tello, Cristina Guarducci, Francisco Hermida-Prado, Sudeepa Syamala, Klothilda Lim, Miguel Munoz Gomez, Matthew Pun, MacIntosh Cornwell, Weihan Liu, Aysegul Ors, Hisham Mohammed, Paloma Cejas, Jane B. Brock, Matthew L. Freedman, Eric P. Winer, Xiaoyong Fu, Rachel Schiff, Henry W. Long, Otto Metzger Filho, and Rinath Jeselsohn
- Subjects
Carcinoma, Lobular ,Tamoxifen ,Cancer Research ,Receptors, Estrogen ,Oncology ,Drug Resistance, Neoplasm ,Carcinoma, Ductal, Breast ,Humans ,Breast Neoplasms ,Female ,Prognosis ,Chromatin ,Article - Abstract
Most invasive lobular breast cancers (ILC) are of the luminal A subtype and are strongly hormone receptor–positive. Yet, ILC is relatively resistant to tamoxifen and associated with inferior long-term outcomes compared with invasive ductal cancers (IDC). In this study, we sought to gain mechanistic insights into these clinical findings that are not explained by the genetic landscape of ILC and to identify strategies to improve patient outcomes. A comprehensive analysis of the epigenome of ILC in preclinical models and clinical samples showed that, compared with IDC, ILC harbored a distinct chromatin state linked to gained recruitment of FOXA1, a lineage-defining pioneer transcription factor. This resulted in an ILC-unique FOXA1–estrogen receptor (ER) axis that promoted the transcription of genes associated with tumor progression and poor outcomes. The ILC-unique FOXA1–ER axis led to retained ER chromatin binding after tamoxifen treatment, which facilitated tamoxifen resistance while remaining strongly dependent on ER signaling. Mechanistically, gained FOXA1 binding was associated with the autoinduction of FOXA1 in ILC through an ILC-unique FOXA1 binding site. Targeted silencing of this regulatory site resulted in the disruption of the feed-forward loop and growth inhibition in ILC. In summary, ILC is characterized by a unique chromatin state and FOXA1–ER axis that is associated with tumor progression, offering a novel mechanism of tamoxifen resistance. These results underscore the importance of conducting clinical trials dedicated to patients with ILC in order to optimize treatments in this breast cancer subtype. Significance: A unique FOXA1–ER axis in invasive lobular breast cancer promotes disease progression and tamoxifen resistance, highlighting a potential therapeutic avenue for clinical investigations dedicated to this disease. See related commentary by Blawski and Toska, p. 3668
- Published
- 2022
7. Nodal Positivity in Early-Stage Triple-Negative Breast Cancer: Implications for Preoperative Immunotherapy
- Author
-
Elizabeth A. Mittendorf, Olga Kantor, Anna Weiss, Edward Richardson, Ana Garrido-Castro, Leah H. Portnow, Ian E. Krop, Nancy U. Lin, Eric P. Winer, Sara M. Tolaney, and Tari A. King
- Subjects
Oncology ,Surgery - Published
- 2022
8. Expanding the Staging Criteria for T1-2N0 Hormone-Receptor Positive Breast Cancer Patients Enrolled in TAILORx
- Author
-
Olga Kantor, Harold J. Burstein, Tari A. King, Steven Shak, Christy A. Russell, Armando E. Giuliano, Gabriel N. Hortobagyi, Eric P. Winer, Larissa A. Korde, Joseph A. Sparano, and Elizabeth A. Mittendorf
- Subjects
Oncology ,Surgery - Published
- 2022
9. Extended adjuvant endocrine therapy in a longitudinal cohort of young breast cancer survivors
- Author
-
Tal Sella, Yue Zheng, Shoshana M. Rosenberg, Kathryn J. Ruddy, Shari I. Gelber, Rulla M. Tamimi, Jeffrey M. Peppercorn, Lidia Schapira, Virginia F. Borges, Steven E. Come, Lisa A. Carey, Eric P. Winer, and Ann H. Partridge
- Subjects
Oncology ,Pharmacology (medical) ,Radiology, Nuclear Medicine and imaging - Abstract
Extended adjuvant endocrine therapy (eET) improves outcomes in breast cancer survivors. Most studies however have been limited to postmenopausal women, and optimal eET for young survivors is uncertain. We report eET use among participants in the Young Women’s Breast Cancer Study (YWS), a multicenter prospective cohort of women age ≤40 newly diagnosed with breast cancer enrolled between 2006–2016. Women with stage I–III hormone receptor-positive breast cancer, ≥6 years from diagnosis without recurrence were considered eET candidates. Use of eET was elicited from annual surveys sent years 6–8 after diagnosis, censoring for recurrence/death. 663 women were identified as eET candidates with 73.9% (490/663) having surveys eligible for analysis. Among eligible participants, mean age was 35.5 (±3.9), 85.9% were non-Hispanic white, and 59.6% reported eET use. Tamoxifen monotherapy was the most reported eET (77.4%), followed by aromatase inhibitor (AI) monotherapy (21.9%), AI-ovarian function suppression (AI-OFS) (6.8%) and tamoxifen-OFS (3.1%). In multivariable analysis, increasing age (per year odds ratio [OR]: 1.10, 95% confidence interval [CI]: 1.04–1.16), stage (II v. I: OR: 2.86, 95% CI: 1.81–4.51; III v. I: OR: 3.73, 95%CI: 1.87–7.44) and receipt of chemotherapy (OR: 3.66, 95% CI: 2.16–6.21) were significantly associated with eET use. Many young breast cancer survivors receive eET despite limited data regarding utility in this population. While some factors associated with eET use reflect appropriate risk-based care, potential sociodemographic disparities in uptake warrants further investigation in more diverse populations.
- Published
- 2023
10. Association Between Biomarkers and Clinical Outcomes of Pembrolizumab Monotherapy in Patients With Metastatic Triple-Negative Breast Cancer: KEYNOTE-086 Exploratory Analysis
- Author
-
Sherene Loi, Roberto Salgado, Peter Schmid, Javier Cortes, David W. Cescon, Eric P. Winer, Deborah L. Toppmeyer, Hope S. Rugo, Michelino De Laurentiis, Rita Nanda, Hiroji Iwata, Ahmad Awada, Antoinette R. Tan, Yuan Sun, Vassiliki Karantza, Anran Wang, Lingkang Huang, Assieh Saadatpour, Razvan Cristescu, Jennifer Yearley, Jared Lunceford, Petar Jelinic, and Sylvia Adams
- Subjects
Cancer Research ,Oncology - Abstract
PURPOSE In the two-cohort phase II KEYNOTE-086 study (ClinicalTrials.gov identifier: NCT02447003 ), first-line and second-line or later pembrolizumab monotherapy demonstrated antitumor activity in metastatic triple-negative breast cancer (mTNBC; N = 254). This exploratory analysis evaluates the association between prespecified molecular biomarkers and clinical outcomes. METHODS Cohort A enrolled patients with disease progression after one or more systemic therapies for metastatic disease irrespective of PD-L1 status; Cohort B enrolled patients with previously untreated PD-L1–positive (combined positive score [CPS] ≥ 1) metastatic disease. The association between the following biomarkers as continuous variables and clinical outcomes (objective response rate [ORR], progression-free survival [PFS], and overall survival [OS]) was evaluated: PD-L1 CPS (immunohistochemistry), cluster of differentiation 8 (CD8; immunohistochemistry), stromal tumor-infiltrating lymphocyte (sTIL; hematoxylin and eosin staining), tumor mutational burden (TMB; whole-exome sequencing [WES]), homologous recombination deficiency-loss of heterozygosity, mutational signature 3 (WES), mutational signature 2 (apolipoprotein B mRNA editing catalytic polypeptide–like; WES), T-cell–inflamed gene expression profile (TcellinfGEP; RNA sequencing), and 10 non-TcellinfGEP signatures (RNA sequencing); Wald test P values were calculated, and significance was prespecified at α = 0.05. RESULTS In the combined cohorts (A and B), PD-L1 ( P = .040), CD8 ( P < .001), sTILs ( P = .012), TMB ( P = .007), and TcellinfGEP ( P = .011) were significantly associated with ORR; CD8 ( P < .001), TMB ( P = .034), Signature 3 ( P = .009), and TcellinfGEP ( P = .002) with PFS; and CD8 ( P < .001), sTILs ( P = .004), TMB ( P = .025), and TcellinfGEP ( P = .001) with OS. None of the non-TcellinfGEP signatures were associated with outcomes of pembrolizumab after adjusting for the TcellinfGEP. CONCLUSION In this exploratory biomarker analysis from KEYNOTE-086, baseline tumor PD-L1, CD8, sTILs, TMB, and TcellinfGEP were associated with improved clinical outcomes of pembrolizumab and may help identify patients with mTNBC who are most likely to respond to pembrolizumab monotherapy.
- Published
- 2023
11. Diet and physical activity interventions in Black and Latina women with breast cancer: A scoping review
- Author
-
Margaret S. Pichardo, Tara Sanft, Leah M. Ferrucci, Yaideliz M. Romero-Ramos, Brenda Cartmel, Maura Harrigan, Ana I. Velazquez, Oluwadamilola M. Fayanju, Eric P. Winer, and Melinda L. Irwin
- Subjects
Cancer Research ,Oncology - Abstract
BackgroundA growing number of lifestyle interventions are being developed to promote weight loss and adoption of a healthful lifestyles among breast cancer survivors; yet Black and Latina women remain underrepresented.PurposeWe performed a scoping review of the available peer-reviewed literature to describe and compare the content, design, methods, and primary outcomes of current diet and/or physical activity (PA) interventions after a breast cancer diagnosis among Black and Latina women.MethodsWe queried PubMed, EMBASE, CINAHL, MEDLINE, and Clinicaltrials.gov up to October 1, 2022, to identify all randomized controlled trials of diet and/or PA after diagnosis of breast cancer with a majority (>50%) of Black or Latina participants.ResultsTwenty-two randomized controlled trials were included in this review (five efficacy, twelve pilot, five on-going). Nine trials were among Latinas (two diet, four PA, and three diet/PA), six among Blacks (one PA and five diet/PA) and seven included both populations (five PA and two diet/PA), all of which examined different endpoints. Two of the five efficacy studies achieved their a priori outcome (one diet trial improved short term dietary intake; one PA trial achieved clinically significant improvements in metabolic syndrome score), both in Latinas. Eight pilot trials intervened on both diet and PA and three of them found favorable behavioral changes. Three (two for Latinas and one for Blacks) out of the nine diet and PA trials and three (all for Latinas) efficacy trials incorporated a culturally focused approach (i.e., traditional foods, music, Spanish content, bicultural health coaches, spirituality). Overall, four trials, including one efficacy trial, had one-year follow-up data, with three finding sustained behavior change. Electronic/mobile components were incorporated in five trials and one involved informal care givers. Most of the trials were geographically limited to the Northeast USA (n=8, NY, NC, DC, NJ) and Texas (n=4).ConclusionsMost of the trials we identified were pilot or feasibility studies and of short duration, demonstrating the need for large randomized controlled efficacy lifestyle interventions among Black and Latina breast cancer survivors. Culturally tailored programing was limited but is an important component to incorporate in future trials in these populations.
- Published
- 2023
12. Local Therapy Outcomes and Toxicity From the ATEMPT Trial (TBCRC 033): A Phase II Randomized Trial of Adjuvant Trastuzumab Emtansine Versus Paclitaxel in Combination With Trastuzumab in Women With Stage I HER2-Positive Breast Cancer
- Author
-
Jennifer R. Bellon, Nabihah Tayob, David D. Yang, Jordan Tralins, Chau T. Dang, Steven J. Isakoff, Michelle DeMeo, Harold J. Burstein, Ann H. Partridge, Eric P. Winer, Ian E. Krop, and Sara M. Tolaney
- Subjects
Cancer Research ,Radiation ,Oncology ,Radiology, Nuclear Medicine and imaging - Published
- 2022
13. CALGB 40603 (Alliance): Long-Term Outcomes and Genomic Correlates of Response and Survival After Neoadjuvant Chemotherapy With or Without Carboplatin and Bevacizumab in Triple-Negative Breast Cancer
- Author
-
Jonathan H. Shepherd, Karla Ballman, Mei-Yin C. Polley, Jordan D. Campbell, Cheng Fan, Sara Selitsky, Aranzazu Fernandez-Martinez, Joel S. Parker, Katherine A. Hoadley, Zhiyuan Hu, Yan Li, Matthew G. Soloway, Patricia A. Spears, Baljit Singh, Sara M. Tolaney, George Somlo, Elisa R. Port, Cynthia Ma, Charles Kuzma, Eleftherios Mamounas, Mehra Golshan, Jennifer R. Bellon, Deborah Collyar, Olwen M. Hahn, Clifford A. Hudis, Eric P. Winer, Ann Partridge, Terry Hyslop, Lisa A. Carey, Charles M. Perou, and William M. Sikov
- Subjects
Bevacizumab ,Cancer Research ,Neoplasm, Residual ,Paclitaxel ,Oncology ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Breast Neoplasms ,Female ,Triple Negative Breast Neoplasms ,Neoadjuvant Therapy ,Carboplatin - Abstract
PURPOSE CALGB 40603 ( NCT00861705 ), a 2 × 2 randomized phase II trial, demonstrated that adding carboplatin or bevacizumab to weekly paclitaxel (wP) followed by doxorubicin and cyclophosphamide significantly increased the pathologic complete response (pCR) rate in stage II-III triple-negative breast cancer. We now report long-term outcomes (LTOs) and correlative science end points. PATIENTS AND METHODS The Kaplan-Meier method was used to estimate LTOs in 443 patients who initiated study treatment. Log-rank tests and Cox proportional hazards models evaluated the impact of clinical characteristics, pathologic response, calculated residual cancer burden (RCB) in patients with residual disease (RD), treatment assignment, and dose delivery during wP on LTOs, including event-free survival (EFS). Genomic predictors of treatment response and outcomes were assessed on pretreatment tumor samples by mRNA sequencing. RESULTS Among baseline characteristics, only the clinical stage was associated with LTOs. At a median follow-up of 7.9 years, LTOs were not significantly improved with either carboplatin or bevacizumab, overall or in patients with basal-like subtype cancers by genomic analysis. Patients with pCR (n = 205, 46.3%) had significantly higher 5-year EFS (85.5% v 56.6%, log-rank P < .0001) and overall survival (87.9% v 63.4%, P < .0001) rates compared with patients with RD, even those with RCB class I. Among clinical and genomic features, evidence of immune activation, including tumor-infiltrating lymphocytes and low B-cell receptor evenness, was associated with pCR and improved EFS. CONCLUSION Despite higher pCR rates, neither carboplatin nor bevacizumab appeared to improve LTOs although the study was not powered to assess these secondary end points. pCR was associated with superior LTOs even when compared with minimal RD. Markers of immune activation in pretreatment tumor biopsies were independently associated with higher pCR rates and improved survival.
- Published
- 2022
14. Abstract P2-14-18: A randomized phase II trial of carboplatin with or without nivolumab in metastatic triple-negative breast cancer
- Author
-
Ana C Garrido-Castro, Noah Graham, Kevin Bi, Jihye Park, Jingxin Fu, Tanya Keenan, Edward Thomas Richardson, Ricardo Pastorello, Paulina Lange, Victoria Attaya, Robert Wesolowski, Natalie Sinclair, Zarah Lucas, Steve Lo, Nadine Tung, Meredith Faggen, Peter A Kaufman, Caroline C Block, Fred Briccetti, Madhavi Toke, Wendy Chen, Kai Wucherpfennig, Sascha Marx, Ye Tian, Judith Agudo, Jennifer L Guerriero, Stuart Schnitt, Nancy U Lin, Eric P Winer, Elizabeth A Mittendorf, Nabihah Tayob, Eliezer Van Allen, and Sara M Tolaney
- Subjects
Cancer Research ,Oncology - Abstract
Background: Platinum agents induce DNA crosslinking and cause accumulation of genotoxic stress, which leads to immune activation via IFN-γ signaling, making the combination with nivolumab (PD-1 antibody) an attractive strategy to enhance the benefit of either agent alone in metastatic triple-negative breast cancer (mTNBC). Methods: In this phase II open-label, investigator-initiated, multicenter trial, patients with unresectable locally advanced or mTNBC treated with 0-1 prior lines of chemotherapy in the metastatic setting were randomized 1:1 to carboplatin (AUC 6) with or without nivolumab (360 mg) IV every 3 weeks. Stratification factors included: germline BRCA (gBRCA) status, prior neo/adjuvant platinum, and number of prior lines of metastatic therapy. After approval of PD-L1 inhibition for mTNBC, the study was amended to include first-line mTNBC only and PD-L1 status was added as a stratification factor. Patients randomized to carboplatin alone were allowed to crossover at progression to receive nivolumab (+ nab-paclitaxel post-amendment). The primary objective was to compare progression-free survival (PFS) per RECIST 1.1 criteria of carboplatin with or without nivolumab in first-line mTNBC in the intent-to-treat (ITT) population. Key secondary objectives were objective response rate (ORR), overall survival (OS), clinical benefit rate, and duration and time to objective response. PD-L1 status was confirmed centrally using the SP142 Ventana assay (positive, ≥1% IC). Paired research biopsies at baseline, on-treatment and at progression were performed, if safely accessible. The trial closed to accrual prior to reaching target accrual due to approval of PD-1 inhibition in combination with platinum-based chemotherapy for PD-L1+ mTNBC. Results: Between 1/30/2018 and 12/9/2020, 78 patients enrolled. Three patients did not receive protocol treatment, and the safety analysis was conducted among the 75 that received any treatment; 37 received carboplatin + nivolumab (Arm A), 38 received carboplatin alone (Arm B). Median age was 59.1 yrs (range: 25.4-75.8). Four patients (5.3%) had a known gBRCA1/2 mutation. Sixty-two (82.7%) patients received 0 prior lines (ITT population) and 13 (17.3%) 1 prior line of metastatic therapy. Sixty-seven patients (89.3%) experienced any grade ≥2 treatment-related adverse event (AE). The most frequent AE were platelet count decrease (n=40; 53.3%), anemia (n=36; 48.0%), neutrophil count decrease (n=33; 44.0%) and fatigue (n=24; 32.0%). Grade 3/4 AE were observed in 46 (61.3%) patients, and there was one grade 5 AE (COVID19 pneumonia). Any grade ≥2 immune-related AE (irAE) were observed in 25 of the 37 (67.6%) patients treated with carboplatin + nivolumab. Grade 3/4 irAE were observed in 11 (29.7%) patients. In the ITT population (32 on Arm A; 30 on Arm B), median PFS was 4.2 months with carboplatin + nivolumab, and 5.5 months with carboplatin (stratified HR 0.98, 95% CI [0.51 - 1.88]; p=0.95). ORR was 25% vs. 23.3%, respectively. At a median follow-up of 23.5 months, median OS was 17.5 months vs. 10.7 months (stratified HR 0.63, 95% CI [0.32 - 1.24]; p=0.18). In patients with PD-L1+ mTNBC (13 on Arm A; 11 on Arm B), median PFS was 8.3 months and 4.7 months, respectively (stratified HR 0.63, 95% CI [0.21 - 1.89]; p=0.41). ORR was 23.1% vs. 27.3%, respectively. Median OS was 17.5 months vs. 9.6 months (stratified HR 0.59, 95% CI [0.20 - 1.75]; p=0.34). Conclusions: Addition of nivolumab to carboplatin in patients with previously untreated mTNBC, unselected by PD-L1 status, did not significantly improve PFS. A trend toward improved PFS and OS was observed in patients with PD-L1+ mTNBC. Tissue, blood and intestinal microbiome biomarker analyses are planned; bulk tumor and single-cell sequencing, and TCR sequencing in peripheral blood are ongoing. Clinical trial information: NCT03414684. Citation Format: Ana C Garrido-Castro, Noah Graham, Kevin Bi, Jihye Park, Jingxin Fu, Tanya Keenan, Edward Thomas Richardson, Ricardo Pastorello, Paulina Lange, Victoria Attaya, Robert Wesolowski, Natalie Sinclair, Zarah Lucas, Steve Lo, Nadine Tung, Meredith Faggen, Peter A Kaufman, Caroline C Block, Fred Briccetti, Madhavi Toke, Wendy Chen, Kai Wucherpfennig, Sascha Marx, Ye Tian, Judith Agudo, Jennifer L Guerriero, Stuart Schnitt, Nancy U Lin, Eric P Winer, Elizabeth A Mittendorf, Nabihah Tayob, Eliezer Van Allen, Sara M Tolaney. A randomized phase II trial of carboplatin with or without nivolumab in metastatic triple-negative breast cancer [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr P2-14-18.
- Published
- 2022
15. Abstract P2-10-01: Estimating risk of breast cancer-specific mortality (BCSM) and non-BCSM in patients with triple-negative breast cancer
- Author
-
Jose P Leone, Noah Graham, Julieta Leone, Sara M Tolaney, Bernardo A Leone, Rachel A Freedman, Michael J Hassett, Carlos T Vallejo, Eric P Winer, Nancy U Lin, and Nabihah Tayob
- Subjects
Cancer Research ,Oncology - Abstract
Background: Triple-negative breast cancer (TNBC) is associated with high risk of distant recurrence and death. At present, our ability to estimate risk of death from causes other than breast cancer is limited. Particularly among elderly patients (pts), who have been historically underrepresented in clinical trials. In pts with TNBC, assessing both risks is important for our treatment recommendations. The aim of this study was to evaluate risk of BCSM and non-BCSM in TNBC by patient (pt) and tumor characteristics. Methods: Using data from the Surveillance, Epidemiology, and End Results (SEER) program, we identified women diagnosed with non-metastatic invasive TNBC between 2010-2016. Fine and Gray regression was used to evaluate the association of BCSM with pre-specified variables including pt age, tumor size (T), nodal status (N), and tumor grade, while considering deaths from other causes as competing events. We then estimated cumulative risk of BCSM, non-BCSM and all-cause mortality within subgroups defined by baseline clinical and pathologic variables. We conducted a subset analysis of N0 pts older than 50 years, given that we anticipated this subgroup would have the most clinically useful balance between BCSM and non-BCSM. Results: We included 37,293 pts. Age distribution was: 27.1% 80 years vs 50-69 years (Hazard ratio [HR] 1.62; 95% CI, [1.45 - 1.80]), T4 vs T1a (HR 8.51; 95% CI, [6.20 - 11.68]), N3 vs N0 (HR 6.31; 95% CI, [5.70 - 7.00]) and grade III/IV vs grade I (HR 2.10; 95% CI, [1.44 - 3.07]). The cumulative risk of BCSM in year 0-7 was 10.7% for N0, 27.9% for N1, 46.4% for N2 and 64.0% for N3. In contrast, the cumulative risk of non-BCSM over the same period ranged from 7.5% in N1 to 8.7% in N2. The table shows risks of BCSM, non-BCSM and all-cause mortality among pts with N0 disease by age at diagnosis and tumor size. Pts 50-69 years had an increasing cumulative risk of BCSM by tumor size up to 13.0% in those with T2 tumors, while the risk of non-BCSM ranged from 4.8% to 5.9%. Pts aged 70-79 years with T1a/b, N0 tumors had risks of BCSM that were approximately 60% lower than the risks of non-BCSM. In pts aged ≥80 years, the risk of non-BCSM increased and is significantly higher than BCSM in patients with T1b-T2 disease. Conclusions: The risk of BCSM in TNBC depends on traditional clinicopathologic factors and is in general, much higher than the risk of non-BCSM. However, the high risk of non-BCSM among older pts is substantial which needs to be taken into consideration when making treatment recommendations. An interactive tool to estimate risks of BCSM, non-BCSM and all-cause mortality for TNBC will be presented at the meeting. BCSMnon-BCSMAll-cause mortalityCumulative risk (%) and 95% CICumulative risk (%) and 95% CICumulative risk (%) and 95% CIyears 0-7years 0-7years 0-7Tumor size among age 50-69, N0 onlyT1a2.6 (1.0 - 4.3)5.9 (3.2 - 8.6)8.5 (5.3 - 11.6)T1b3.9 (2.8 - 5.0)4.8 (3.3 - 6.3)8.7 (6.9 - 10.5)T1c8.1 (6.9 - 9.4)4.8 (3.9 - 5.8)13.0 (11.4 - 14.5)T213.0 (11.6 - 14.4)5.5 (4.4 - 6.5)18.5 (16.8 - 20.2)Tumor size among age 70-79, N0 onlyT1a6.1 (0 - 12.7)13.9 (7.0 - 20.9)20.0 (10.2 - 28.7)T1b5.3 (3.0 - 7.7)13.3 (9.0 - 17.7)18.6 (13.7 - 23.3)T1c11.0 (8.7 - 13.4)14.3 (11.4 - 17.2)25.3 (21.7 - 28.8)T221.0 (17.4 - 24.6)17.4 (13.4 - 21.5)38.5 (33.3 - 43.2)Tumor size among age ≥80, N0 onlyT1a6.6 (0 - 19.7)27.0 (11.0 - 43.1)33.7 (11.8 - 50.1)T1b7.1 (2.1 - 12.2)33.2 (23.2 - 43.2)40.3 (28.9 - 49.9)T1c8.4 (5.2 - 11.6)32.7 (26.4 - 39.0)41.1 (34.1 - 47.3)T222.7 (18.1 - 27.3)41.6 (34.2 - 49.1)64.3 (56.0 - 71.1) Citation Format: Jose P Leone, Noah Graham, Julieta Leone, Sara M Tolaney, Bernardo A Leone, Rachel A Freedman, Michael J Hassett, Carlos T Vallejo, Eric P Winer, Nancy U Lin, Nabihah Tayob. Estimating risk of breast cancer-specific mortality (BCSM) and non-BCSM in patients with triple-negative breast cancer [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr P2-10-01.
- Published
- 2022
16. Abstract P2-07-03: Correlation of immune-related protein expression with hormone receptor (HR) status and pathologic response to neoadjuvant paclitaxel/trastuzumab/pertuzumab (THP) among patients with early-stage HER2+ breast cancer
- Author
-
Adrienne G. Waks, Ying Huang, Tyler Hether, Esther R Ogayo, Sapana Kadel, Jillian Alberti, Sara M Tolaney, Ian E Krop, Prajan Divakar, Otto Metzger, Madison L O'Donnell, Sarah Church, Eric P Winer, Jennifer L. Guerriero, and Elizabeth A Mittendorf
- Subjects
Cancer Research ,Oncology - Abstract
Background: Immunological differences between HER2+ breast tumors that are HR+ versus HR- have not been widely explored. DAPHNe was a single-arm prospective clinical trial enrolling patients with clinical anatomic stage II-III HER2+ breast cancer to neoadjuvant treatment with THP. Here, we examine differential expression of immune-related proteins between HR+/HER2+ and HR-/HER2+ tumors in pre-treatment biopsies from trial participants, and analyze correlates of pathologic response to THP, overall and by HR status. Methods: A baseline research biopsy was required in all participants prior to initiation of neoadjuvant therapy. All patients received 12 weeks of neoadjuvant THP; 5 patients also received neoadjuvant AC given incomplete clinical response to THP, and are excluded from residual cancer burden (RCB) analyses. The trial primary endpoint, feasibility of de-escalation to antibody doublet therapy only (HP) following pCR, was previously reported. Protein expression profiling of baseline research biopsies was performed on the NanoString GeoMx® platform using a 58-protein immune cell profiling panel. Regions of interest (ROI; up to 12 per slide) were identified by a pathologist based on tumor cell, immune cell, and/or T cell presence, and all ROI were segmented into tumor vs stroma. Protein data quality controls were normalized by isotypes. Linear mixed effect models were used for differential expression comparisons; p-values were estimated using Satterthwaite’s method. Results: 97 pre-treatment breast tumor specimens were analyzed: 64 patients had HR+ tumors, 32 HR-, 1 unknown; 67 patients had favorable (RCB 0/1) response to preop therapy, 30 patients had unfavorable response (RCB 2/3). Multiple significant differences were observed between protein expression in HR+ versus HR- tumors (Table 1). Multiple immune cell types, as well as checkpoint proteins PD-L1 and IDO1, were higher in HR- patients, whereas checkpoint proteins B7-H3 and TIM3 were higher in HR+ patients. In the overall population, higher HER2 and PD-L1 expression were significant predictors of favorable RCB response, while higher ER alpha and Bcl-2 expression were significant predictors of unfavorable RCB response (Table 2). Conclusions: As anticipated, the strength of HER2 expression and ER expression were the most significant predictors of favorable and unfavorable RCB response to neoadjuvant THP, respectively. This highly multiplexed protein expression analysis demonstrated significant differences between the immune microenvironment of HR+ and HR- HER2+ breast tumors, implying that distinct immunological targets should be explored in the treatment of HER2+ breast cancer according to HR status. Table 1.Differences between HR+/HER2+ and HR-/HER2+ patientsStroma compartmentTumor compartmentHigher in HR+ER alpha* (fold-change 2.25)ER alpha* (8.94)Fibronectin (1.68)Bcl-2* (1.98)B7-H3 (1.44)Fibronectin* (1.90)PR (1.69)TIM-3 (1.37)Higher in HR-CD27* (fold-change 1.36)IDO1 (1.50)IDO1 (1.77)S100B (1.44)CD45 (1.45)MART1 (1.33)CD3 (1.35)CD20 (1.34)STING (1.33)CD4 (1.30)ICOS (1.30)CD45RO (1.28)CD40 (1.28)CD127 (1.27)VISTA (1.25)PD-L1 (1.24)4-1BB (1.21)*FDR p-value < 0.05 (otherwise, p-value < 0.05). Table 2: Predictors of RCB response Overall populationHigher in RCB 0/1HER2* (fold-change 3.75)PD-L1 (1.25)Higher in RCB 2/3ER alpha* (2.44)Bcl-2 (1.46)HR+/HER2+ patientsStroma compartmentTumor compartmentHigher in RCB 0/1HER2* (fold-change 4.28)HER2* (4.21)CTLA4 (1.64)Higher in RCB 2/3Bcl-2* (2.18)*FDR p-value < 0.05 (otherwise, p-value < 0.05).HR-/HER2+ patients are not shown in Table 2 as only 2 HR- patients had unfavorable RCB response. Citation Format: Adrienne G. Waks, Ying Huang, Tyler Hether, Esther R Ogayo, Sapana Kadel, Jillian Alberti, Sara M Tolaney, Ian E Krop, Prajan Divakar, Otto Metzger, Madison L O'Donnell, Sarah Church, Eric P Winer, Jennifer L. Guerriero, Elizabeth A Mittendorf. Correlation of immune-related protein expression with hormone receptor (HR) status and pathologic response to neoadjuvant paclitaxel/trastuzumab/pertuzumab (THP) among patients with early-stage HER2+ breast cancer [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr P2-07-03.
- Published
- 2022
17. Abstract P2-13-02: Pathologic nodal staging and systemic therapy among patients with cT1-2N0 HER2+ breast cancer: A prospective single institution cohort analysis
- Author
-
Anna Weiss, Adrienne G. Waks, Alison Laws, Sara M. Tolaney, Eric P. Winer, Elizabeth A. Mittendorf, Ann H. Partridge, and Tari A. King
- Subjects
Cancer Research ,Oncology - Abstract
Background: The optimal systemic therapy strategy, neoadjuvant vs adjuvant therapy, for patients with small clinically node negative HER2+ breast cancers is uncertain. The goal of this study was to evaluate the incidence of pathologic nodal disease in cT1-2 N0 HER2+ patients, treated with upfront surgery or neoadjuvant chemotherapy (NAC), to better define selection criteria for initial treatment approach. Methods: Our prospectively maintained database was queried for cT1-2 N0 HER2+ breast cancer patients diagnosed 2/12/2015 - 10/13/2020 (after publication of the Adjuvant Paclitaxel and Trastuzumab trial). Patients without axillary surgery were excluded (n =23). HER2 positivity was defined per ASCO/CAP guidelines. Pre-NAC and/or pre-operative axillary ultrasound was not routinely performed. Patient characteristics and rates of pN+ disease were compared by treatment strategy using Chi square and Wilcoxon rank-sum tests. Nodes with isolated tumor cells were considered negative (pN0) in the upfront surgery setting but positive (pN+) after NAC. Logistic regression determined factors associated with pN+ disease. Results: A total of 579 eligible patients were identified; 368 (63.6%) were treated with upfront surgery and 211 (36.4%) with NAC. Upfront surgery patients were older (median 55 years [range 23-88] vs 49 [24-79], p Table 1.Pathologic nodal stage among upfront surgery and NAC patientsUpfront Surgery (N=368)pN+pN0P valuecT category< 0.001T1mi (N=48)6 (10.4%)42 (89.6%)T1a (N=26)3 (11.5%)23 (88.5%)T1b (N=87)7 (8.0%)80 (92.0%)T1c (N=154)38 (24.7%)116 (75.3%)T2 (N=53)19 (35.8%)34 (64.2%)NAC (N=211)ypN+ypN0P valuecT category0.719T1b (N=7)1 (14.3%)6 (85.7%)T1c (N=30)5 (16.7%)25 (83.3%)T2 (N=174)20 (11.5%)154 (88.5%) Citation Format: Anna Weiss, Adrienne G. Waks, Alison Laws, Sara M. Tolaney, Eric P. Winer, Elizabeth A. Mittendorf, Ann H. Partridge, Tari A. King. Pathologic nodal staging and systemic therapy among patients with cT1-2N0 HER2+ breast cancer: A prospective single institution cohort analysis [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr P2-13-02.
- Published
- 2022
18. Abstract GS2-10: Nimbus: A phase 2 trial of nivolumab plus ipilimumab for patients with hypermutated her2-negative metastatic breast cancer (MBC)
- Author
-
Romualdo Barroso-Sousa, Tianyu Li, Sangeetha Reddy, Leisha A. Emens, Beth Overmoyer, Paulina Lange, Molly K Dilullo, Victoria Attaya, Jeffrey Kimmel, Eric P. Winer, Elizabeth A. Mittendorf, Nabihah Tayob, and Sara M. Tolaney
- Subjects
Cancer Research ,Oncology - Abstract
Background: While high tumor mutational burden (TMB-H) has been used as a tissue-agnostic biomarker for approval of immune checkpoint inhibitors (ICI), there is a paucity of data regarding efficacy of ICI in TMB-H MBC. The aim of this study was to evaluate if patients with TMB-H HER2-negative MBC benefit from the combination of nivolumab plus ipilimumab. Methods: This is an open-label, single-arm, multicenter, phase 2 study assessing the efficacy of nivolumab 3 mg/kg intravenously (IV) every 14 days plus ipilimumab 1 mg/kg IV every 6 weeks in subjects with TMB-H HER2-negative MBC. Eligible patients were required to have measurable HER2-negative MBC, TMB ≥9 Mut/Mb assessed by a cancer-gene panel evaluating > 300 genes and performed in a CLIA-certified laboratory, and 0-3 prior lines of chemotherapy in the advanced setting. The primary objective was overall response rate (ORR) according to RECIST 1.1. Secondary objectives include safety and tolerability, progression-free survival (PFS), and overall survival (OS). The study followed a two-stage design. In the first stage, 14 patients were enrolled. The study required at least 1 objective response in order to continue to the second stage where an additional 16 patients were enrolled. At least 4 objective responses among the 30 patients would suggest the regimen is worthy of further study. If the true response rate is 25%, the chance that the regimen is declared worthy of further study is > 90%. Tumor biopsies, peripheral blood mononuclear cells, circulating tumor DNA, and stool collection were mandatory and were obtained at baseline and on treatment (end of cycle 1). Results: From February 2019 to June 2021, 31 patients were enrolled across 3 different academic institutions. Among 30 patients who initiated study treatment, the median age was 63 yo, 20 had hormone-receptor positive (HR+) breast cancer and 10 had triple-negative breast cancer (TNBC), and median number of prior lines of chemotherapy was 1.5 (0-3). Among the 10 patients with TNBC, PD-L1 status was known in 7 patients (3 positive and 4 negative). Median TMB was 10.9 Mut/Mb and 16.7% (n = 5) of patients had a TMB ≥14 mut/Mb. After a median follow-up of 9.7 (4.4 - 16.4) months, 4 (13.3%) patients achieved a confirmed objective response (all partial responses) meeting the primary endpoint of this study. The median duration of response has not been reached and 3 of these patients are still progression-free for at least 15 months. Two patients have short follow-up, and one has an unconfirmed partial response and the other has a stable disease at the time of the data cut. Median PFS and OS was respectively 1.4 (95% CI 1.3 - 9.5) months and 8.8 (95% CI 4.2 - not reached). Exploratory analysis did not show a difference in response rate according to HR status and PD-L1 status (data not shown) but tumors with TMB ≥14 mut/Mb had a response rate of 60% vs 4% in the group with TMB between ≥9 and Citation Format: Romualdo Barroso-Sousa, Tianyu Li, Sangeetha Reddy, Leisha A. Emens, Beth Overmoyer, Paulina Lange, Molly K Dilullo, Victoria Attaya, Jeffrey Kimmel, Eric P. Winer, Elizabeth A. Mittendorf, Nabihah Tayob, Sara M. Tolaney. Nimbus: A phase 2 trial of nivolumab plus ipilimumab for patients with hypermutated her2-negative metastatic breast cancer (MBC) [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr GS2-10.
- Published
- 2022
19. Abstract GS2-05: Randomized comparison of adjuvant aromatase inhibitor exemestane (E) plus ovarian function suppression (OFS) vs tamoxifen (T) plus OFS in premenopausal women with hormone receptor-positive (HR+) early breast cancer (BC): update of the combined TEXT and SOFT trials
- Author
-
Meredith M Regan, Barbara A Walley, Gini F Fleming, Prudence A Francis, Marco A Colleoni, István Láng, Henry L Gómez, Carlo A Tondini, Harold J Burstein, Matthew P Goetz, Eva M Ciruelos, Vered Stearns, Hervé R Bonnefoi, Silvana Martino, Charles E Geyer, Claudio Chini, Alessandro M Minisini, Simon Spazzapan, Thomas Ruhstaller, Eric P Winer, Barbara Ruepp, Sherene Loi, Alan S Coates, Aron Goldhirsch, Richard D Gelber, and Olivia Pagani
- Subjects
Cancer Research ,Oncology - Abstract
Background The updated combined SOFT+TEXT analysis, after 9 years median follow-up (MFU), revealed that adjuvant E+OFS vs T+OFS significantly improved disease-free survival (DFS) and distant recurrence-free interval (DRFI) but not overall survival (OS) in premenopausal women with HR+ early BC (Francis et al NEJM 2018). Given the high rate of OS in both arms and the long-term risk of relapse in HR+ BC, continued follow-up is key to assessing treatment benefit. We report a planned update analysis including OS with database lock of May 2021, after 13 years MFU.. Methods TEXT and SOFT enrolled premenopausal women with HR+ early BC from November 2003 to April 2011 (2660 in TEXT, 3047 in SOFT intention-to-treat (ITT) populations). TEXT randomized women within 12 weeks of surgery to 5 years E+OFS vs T+OFS; chemotherapy (CT) was optional and concurrent with OFS. SOFT randomized women to 5 years E+OFS vs T+OFS vs T alone, within 12 weeks of surgery if no CT planned, or within 8 months of completing (neo)adjuvant CT. Both trials were stratified by CT use. For the combined analysis of E+OFS vs T+OFS, the primary endpoint was DFS defined as invasive local, regional, distant recurrence, contralateral BC, second malignancy, death. Secondary endpoints included invasive breast cancer-free interval (BCFI), DRFI and OS.. Results: At database lock there were 953 DFS events and 473 deaths among 4690 pts assigned to T+OFS or E+OFS. In the ITT population, DFS, BCFI and DRFI outcomes for pts assigned E+OFS (n=2346) continued to be significantly improved over T+OFS (n=2344). 12-yr DFS was 80.5% vs. 75.9% (4.6% absolute improvement; HR 0.79 95% CI 0.70-0.90), 12-yr BCFI was improved by 4.1% and 12-yr DRFI by 1.8%. At 12 years OS was excellent in both groups, 90.1% in pts assigned E+OFS vs 89.1% in pts assigned T+OFS (HR 0.93; 95% CI, 0.78-1.11). There was heterogeneity of relative treatment effect according to HER2 status. When enrollment commenced, anti-HER2 adjuvant therapy was not standard; 53% of 583 pts with HER2+ tumors received HER2-targeted therapy. Below are Kaplan-Meier 12-yr estimates for patients with HER2 negative tumors by trial and chemotherapy stratum and for those with high-grade tumours, as an example of high-risk feature (Table). There is an emerging OS benefit for E+OFS vs T+OFS in pts with HER2 negative tumors who received chemotherapy in both trials.In pts with HER2-negative tumors, clinically-relevant outcome benefits were also seen in other high-risk subgroups: 12-yr DFS and OS were improved by 7.4% and 2.7%, respectively, in pts with pN1a disease, and by 10.6% and 4.5%, respectively, in those with tumors >2cm. Conclusions After 13 years MFU, adjuvant E+OFS, as compared with T+OFS, shows a sustained reduction in the risk of recurrence, more consistent in HER2 negative patients and in those with high-risk disease features, e.g., indication for adjuvant chemotherapy and G3 tumors. Oncologists may use this information to discuss potential benefits of E+OFS with individual patients. Follow-up continues for 5 additional years. Chemotherapy HER2-negativeSOFTT+OFS (n=424)E+OFS (n=411)Absolute difference12-yr DFS67.4%74.1%6.7%12-yr OS81.1%84.4%3.3%TEXTT+OFS (n=656)E+OFS (n=661)Absolute difference12-yr DFS71.0%78.4%7.4%12-yr OS83.5%86.8%3.3%No chemotherapy HER2-negativeSOFTT+OFS (n=445)E+OFS (n=447)Absolute difference12-yr DFS82.9%88.2%5.3%12-yr OS96.1%96.9%0.9%TEXTT+OFS (n=499)E+OFS (n=492)Absolute difference12-yr DFS80.2%86.7%6.5%12-yr OS95.9%96.2%0.2%G3 HER2-negativeT+OFS (n=423)E+OFS (n=405)Absolute difference12-yr DFS62.7%73.0%10.3%12-yr OS78.1%83.6%5.5% Citation Format: Meredith M Regan, Barbara A Walley, Gini F Fleming, Prudence A Francis, Marco A Colleoni, István Láng, Henry L Gómez, Carlo A Tondini, Harold J Burstein, Matthew P Goetz, Eva M Ciruelos, Vered Stearns, Hervé R Bonnefoi, Silvana Martino, Charles E Geyer, Jr, Claudio Chini, Alessandro M Minisini, Simon Spazzapan, Thomas Ruhstaller, Eric P Winer, Barbara Ruepp, Sherene Loi, Alan S Coates, Aron Goldhirsch, Richard D Gelber, Olivia Pagani. Randomized comparison of adjuvant aromatase inhibitor exemestane (E) plus ovarian function suppression (OFS) vs tamoxifen (T) plus OFS in premenopausal women with hormone receptor-positive (HR+) early breast cancer (BC): update of the combined TEXT and SOFT trials [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr GS2-05.
- Published
- 2022
20. Abstract P3-08-01: Clonal hematopoiesis of indeterminate potential (CHIP) in metastatic triple negative breast cancer
- Author
-
Katheryn Santos, Qingchun Jin, Peter G. Miller, Ashka Patel, Gregory J. Kirkner, Janet L. Files, Melissa E. Hughes, Samantha M. Stokes, Nabihah Tayob, Daniel G. Stover, Christopher J. Gibson, Eric P. Winer, Nancy U. Lin, Judy E. Garber, and Heather A. Parsons
- Subjects
Cancer Research ,Oncology - Abstract
Background. Patients (pts) with metastatic triple negative breast cancer (mTNBC) receive serial cytotoxic chemotherapy regimens, often with cumulative myelosuppressive effects, impairing treatment tolerance. Clonal hematopoiesis of indeterminate potential (CHIP) refers to the detection of somatic mutations in genes recurrently mutated in hematologic malignancies in the blood of adults with no evident hematologic abnormalities. Little is known about the natural history of CHIP after breast cancer treatment. We sought to characterize CHIP in pts undergoing treatment for mTNBC. Methods. In this retrospective cohort study we identified 149 pts with biopsy-proven mTNBC at a single tertiary care institution with at least one blood sample collected within six months of metastatic diagnosis. We performed targeted sequencing of cryopreserved peripheral blood mononuclear cell (PBMC)-derived genomic DNA and defined CHIP as the presence of at least one pathogenic somatic mutation present at variant allelic fraction (VAF) of 0.02-0.35. We assessed the relationship between CHIP status and overall survival (OS), demographics, clinicopathologic features, germline mutation status, and type and timing of therapy. Results. We identified 27 unique CHIP variants across 22/149 pts (15%) within six months of metastatic diagnosis. Frequency of mutated genes were as follows: DNMT3A (n=15), PPM1D (n=4), TP53 (n=3), TET2 (n=2), SRCAP (n=1), ZBTB33 (n=1), ZNF318 (n=1). Median follow-up in the cohort was 37.9 months (IQR: 23.9-Not reached). The median age at time of blood draw was 55 years (IQR: 8.5) for pts with CHIP vs. 51 years (IQR: 16.5) for pts without CHIP. Ten (45%) pts with CHIP and 47 (37%) pts without CHIP were current or former smokers. Two (9%) pts with CHIP and 10 (7.9%) pts without CHIP were known germline mutation carriers of BRCA1, BRCA2 or PALB2. Twenty-two (100%) pts with CHIP and 124 (98%) pts without CHIP had received systemic chemotherapy for mTNBC prior to blood draw. There were no significant differences in type of chemotherapy regimen received between patients with or without CHIP. Twenty (90.9%) pts with CHIP vs. 96 (75.6%) of pts without CHIP had received radiation therapy prior to blood draw. Pts with CHIP had similar OS to those without CHIP (median OS 7.75 [2.20-31.7] vs. 9.33 [8.02-11.73] months). No pts developed therapy-related myeloid neoplasms (t-MN) or died of complications of cardiac disease. Conclusions. Pts with mTNBC had a higher frequency of CHIP than previously reported in age-matched healthy populations, but similar CHIP prevalence to what has been seen in cohorts of pts with solid tumors. Our study assessed for the presence of CHIP at only a single time point early in the metastatic course, but serial blood sampling later in treatment might reveal additional cases of CHIP. Though this cohort of patients with life-limiting mTNBC was small, presence of CHIP in the first six months of metastatic diagnosis was not associated with worse survival. Citation Format: Katheryn Santos, Qingchun Jin, Peter G. Miller, Ashka Patel, Gregory J. Kirkner, Janet L. Files, Melissa E. Hughes, Samantha M. Stokes, Nabihah Tayob, Daniel G. Stover, Christopher J. Gibson, Eric P. Winer, Nancy U. Lin, Judy E. Garber, Heather A. Parsons. Clonal hematopoiesis of indeterminate potential (CHIP) in metastatic triple negative breast cancer [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr P3-08-01.
- Published
- 2022
21. Phase 1b Clinical Trial with Alpelisib plus Olaparib for Patients with Advanced Triple-Negative Breast Cancer
- Author
-
Felipe Batalini, Niya Xiong, Nabihah Tayob, Madeline Polak, Julia Eismann, Lewis C. Cantley, Geoffrey I. Shapiro, Viktor Adalsteinsson, Eric P. Winer, Panagiotis A. Konstantinopoulos, Alan D'Andrea, Elizabeth M. Swisher, Ursula A. Matulonis, Gerburg M. Wulf, and Erica L. Mayer
- Subjects
Cancer Research ,Triple Negative Breast Neoplasms ,Poly(ADP-ribose) Polymerase Inhibitors ,Piperazines ,Article ,Phosphatidylinositol 3-Kinases ,Thiazoles ,Oncology ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Phthalazines ,Female ,Neoplasm Recurrence, Local ,Cell-Free Nucleic Acids - Abstract
Purpose: We had previously reported on the safety and the recommended phase 2 dose (RP2D) of olaparib in combination with the PI3Kα-specific inhibitor alpelisib in patients with high-grade serous ovarian cancer as studied in a phase 1b trial (NCT01623349). Here, we report on the breast cancer cohort from that study. Patients and Methods: Eligible patients had recurrent triple-negative breast cancer (TNBC) or recurrent breast cancer of any subtype with a germline BRCA mutation and were enrolled to a dose-escalation or -expansion cohort. After definition of the RP2D, secondary end points included safety and objective response rate (ORR). Exploratory analyses were performed using circulating-free DNA (cfDNA). Results: Seventeen patients with TNBC were enrolled with a median of three prior lines of chemotherapy. The most common treatment-related grade 3–4 adverse events were hyperglycemia (18%) and rash (12%). The ORR was 18% (23% for patients treated at the RP2D) and 59% had disease control. The median duration of response was 7.4 months. Analysis of cfDNA tumor fractions (TFx) revealed that patients with TFx < 15% after completion of the first cycle had a longer progression-free survival compared with those with TFx ≥ 15% (6.0 vs. 0.9 months; P = 0.0001). Conclusions: Alpelisib in combination with olaparib is tolerable in patients with pre-treated TNBC, with evidence of activity in non-BRCA carriers. cfDNA provided important prognostic information. Results highlight potential synergistic use of a PI3K inhibitor to sensitize HR-proficient (BRCA wild-type) TNBC to PARP inhibition and suggest the potential to expand the use of PARP inhibition beyond BRCA-mutant tumors.
- Published
- 2022
22. Adjuvant Palbociclib for Early Breast Cancer: The PALLAS Trial Results (ABCSG-42/AFT-05/BIG-14-03)
- Author
-
Michael, Gnant, Amylou C, Dueck, Sophie, Frantal, Miguel, Martin, Hal J, Burstein, Richard, Greil, Peter, Fox, Antonio C, Wolff, Arlene, Chan, Eric P, Winer, Georg, Pfeiler, Kathy D, Miller, Marco, Colleoni, Jennifer M, Suga, Gabor, Rubovsky, Judith M, Bliss, Ingrid A, Mayer, Christian F, Singer, Zbigniew, Nowecki, Olwen, Hahn, Jacqui, Thomson, Norman, Wolmark, Kepa, Amillano, Hope S, Rugo, Guenther G, Steger, Blanca, Hernando Fernández de Aránguiz, Tufia C, Haddad, Antonia, Perelló, Meritxell, Bellet, Hannes, Fohler, Otto, Metzger Filho, Anita, Jallitsch-Halper, Kadine, Solomon, Céline, Schurmans, Kathy P, Theall, Dongrui R, Lu, Kathleen, Tenner, Christian, Fesl, Angela, DeMichele, and Erica L, Mayer
- Subjects
Cancer Research ,Time Factors ,Antineoplastic Agents, Hormonal ,Pyridines ,Breast Neoplasms ,Middle Aged ,Disease-Free Survival ,Neoadjuvant Therapy ,Piperazines ,Progression-Free Survival ,Oncology ,Chemotherapy, Adjuvant ,Antineoplastic Combined Chemotherapy Protocols ,Disease Progression ,Humans ,Female ,Prospective Studies ,Protein Kinase Inhibitors ,Mastectomy ,Neoplasm Staging - Abstract
PURPOSE Palbociclib is a cyclin-dependent kinase 4 and 6 inhibitor approved for advanced breast cancer. In the adjuvant setting, the potential value of adding palbociclib to endocrine therapy for hormone receptor–positive breast cancer has not been confirmed. PATIENTS AND METHODS In the prospective, randomized, phase III PALLAS trial, patients with hormone receptor–positive, human epidermal growth factor receptor 2–negative early breast cancer were randomly assigned to receive 2 years of palbociclib (125 mg orally once daily, days 1-21 of a 28-day cycle) with adjuvant endocrine therapy or adjuvant endocrine therapy alone (for at least 5 years). The primary end point of the study was invasive disease-free survival (iDFS); secondary end points were invasive breast cancer–free survival, distant recurrence-free survival, locoregional cancer-free survival, and overall survival. RESULTS Among 5,796 patients enrolled at 406 centers in 21 countries worldwide over 3 years, 5,761 were included in the intention-to-treat population. At the final protocol-defined analysis, at a median follow-up of 31 months, iDFS events occurred in 253 of 2,884 (8.8%) patients who received palbociclib plus endocrine therapy and in 263 of 2,877 (9.1%) patients who received endocrine therapy alone, with similar results between the two treatment groups (iDFS at 4 years: 84.2% v 84.5%; hazard ratio, 0.96; CI, 0.81 to 1.14; P = .65). No significant differences were observed for secondary time-to-event end points, and subgroup analyses did not show any differences by subgroup. There were no new safety signals for palbociclib in this trial. CONCLUSION At this final analysis of the PALLAS trial, the addition of adjuvant palbociclib to standard endocrine therapy did not improve outcomes over endocrine therapy alone in patients with early hormone receptor–positive breast cancer.
- Published
- 2022
23. The Phase II MutHER Study of Neratinib Alone and in Combination with Fulvestrant in HER2-Mutated, Non-amplified Metastatic Breast Cancer
- Author
-
Cynthia X. Ma, Jingqin Luo, Rachel A. Freedman, Timothy J. Pluard, Julie R. Nangia, Janice Lu, Frances Valdez-Albini, Melody Cobleigh, Jason M. Jones, Nancy U. Lin, Eric P. Winer, P. Kelly Marcom, Shana Thomas, Jill Anderson, Brittney Haas, Leslie Bucheit, Richard Bryce, Alshad S. Lalani, Lisa A. Carey, Matthew P. Goetz, Feng Gao, Gretchen Kimmick, Mark D. Pegram, Matthew J. Ellis, and Ron Bose
- Subjects
Cancer Research ,Oncology ,Receptor, ErbB-2 ,Antineoplastic Combined Chemotherapy Protocols ,Quinolines ,Humans ,Breast Neoplasms ,Female ,skin and connective tissue diseases ,Fulvestrant - Abstract
Purpose:HER2 mutations (HER2mut) induce endocrine resistance in estrogen receptor–positive (ER+) breast cancer.Patients and Methods:In this single-arm multi-cohort phase II trial, we evaluated the efficacy of neratinib plus fulvestrant in patients with ER+/HER2mut, HER2 non-amplified metastatic breast cancer (MBC) in the fulvestrant-treated (n = 24) or fulvestrant-naïve cohort (n = 11). Patients with ER-negative (ER−)/HER2mut MBC received neratinib monotherapy in an exploratory ER− cohort (n = 5).Results:The clinical benefit rate [CBR (95% confidence interval)] was 38% (18%–62%), 30% (7%–65%), and 25% (1%–81%) in the fulvestrant-treated, fulvestrant-naïve, and ER− cohorts, respectively. Adding trastuzumab at progression in 5 patients resulted in three partial responses and one stable disease ≥24 weeks. CBR appeared positively associated with lobular histology and negatively associated with HER2 L755 alterations. Acquired HER2mut were detected in 5 of 23 patients at progression.Conclusions:Neratinib and fulvestrant are active for ER+/HER2mut MBC. Our data support further evaluation of dual HER2 blockade for the treatment of HER2mut MBC.
- Published
- 2022
24. Trastuzumab Emtansine Plus Pertuzumab Versus Taxane Plus Trastuzumab Plus Pertuzumab After Anthracycline for High-Risk Human Epidermal Growth Factor Receptor 2-Positive Early Breast Cancer: The Phase III KAITLIN Study
- Author
-
Ian E. Krop, Seock-Ah Im, Carlos Barrios, Hervé Bonnefoi, Julie Gralow, Masakazu Toi, Paul A. Ellis, Luca Gianni, Sandra M. Swain, Young-Hyuck Im, Michelino De Laurentiis, Zbigniew Nowecki, Chiun-Sheng Huang, Louis Fehrenbacher, Yoshinori Ito, Jigna Shah, Thomas Boulet, Haiying Liu, Harrison Macharia, Peter Trask, Chunyan Song, Eric P. Winer, and Nadia Harbeck
- Subjects
Adult ,Cancer Research ,Antibiotics, Antineoplastic ,Time Factors ,Receptor, ErbB-2 ,Breast Neoplasms ,Middle Aged ,Trastuzumab ,Ado-Trastuzumab Emtansine ,Antibodies, Monoclonal, Humanized ,Disease-Free Survival ,Oncology ,Chemotherapy, Adjuvant ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Anthracyclines ,Female ,Aged - Abstract
PURPOSE We aimed to improve efficacy and reduce toxicity of high-risk human epidermal growth factor receptor 2 (HER2)–positive early breast cancer (EBC) treatment by replacing taxanes and trastuzumab with trastuzumab emtansine (T-DM1). METHODS The phase III KAITLIN study ( NCT01966471 ) included adults with excised HER2-positive EBC (node-positive or node-negative, hormone receptor–negative, and tumor > 2.0 cm). Postsurgery, patients were randomly assigned 1:1 to anthracycline-based chemotherapy (three-four cycles) and then 18 cycles of T-DM1 plus pertuzumab (AC-KP) or taxane (three-four cycles) plus trastuzumab plus pertuzumab (AC-THP). Adjuvant radiotherapy/endocrine therapy was permitted. Coprimary end points were invasive disease-free survival (IDFS) in the intention-to-treat node-positive and overall populations with hierarchical testing. RESULTS The median follow-up was 57.1 months (interquartile range, 52.1-60.1 months) for AC-THP (n = 918) and 57.0 months (interquartile range, 52.1-59.8 months) for AC-KP (n = 928). There was no significant IDFS difference between arms in the node-positive (n = 1,658; stratified hazard ratio [HR], 0.97; 95% CI, 0.71 to 1.32) or overall population (n = 1846; stratified HR, 0.98; 95% CI, 0.72 to 1.32). In the overall population, the three-year IDFS was 94.2% (95% CI, 92.7 to 95.8) for AC-THP and 93.1% (95% CI, 91.4 to 94.7) for AC-KP. Treatment completion rates (ie, 18 cycles) were 88.4% for AC-THP and 65.0% for AC-KP (difference driven by T-DM1 discontinuation because of laboratory abnormalities [12.5%]). Similar rates of grade ≥ 3 (55.4% v 51.8%) and serious adverse events (23.3% v 21.4%) occurred with AC-THP and AC-KP, respectively. KP decreased clinically meaningful deterioration in global health status versus THP (stratified HR, 0.71; 95% CI, 0.62 to 0.80). CONCLUSION The primary end point was not met. Both arms achieved favorable IDFS. Trastuzumab plus pertuzumab plus chemotherapy remains the standard of care for high-risk HER2-positive EBC.
- Published
- 2023
25. Adjuvant Exemestane With Ovarian Suppression in Premenopausal Breast Cancer: Long-Term Follow-Up of the Combined TEXT and SOFT Trials
- Author
-
Olivia, Pagani, Barbara A, Walley, Gini F, Fleming, Marco, Colleoni, István, Láng, Henry L, Gomez, Carlo, Tondini, Harold J, Burstein, Matthew P, Goetz, Eva M, Ciruelos, Vered, Stearns, Hervé R, Bonnefoi, Silvana, Martino, Charles E, Geyer, Claudio, Chini, Fabio, Puglisi, Simon, Spazzapan, Thomas, Ruhstaller, Eric P, Winer, Barbara, Ruepp, Sherene, Loi, Alan S, Coates, Richard D, Gelber, Aron, Goldhirsch, Meredith M, Regan, and Prudence A, Francis
- Subjects
Cancer Research ,Oncology - Abstract
Clinical trials frequently include multiple end points that mature at different times. The initial report, typically based on the primary end point, may be published when key planned co-primary or secondary analyses are not yet available. Clinical Trial Updates provide an opportunity to disseminate additional results from studies, published in JCO or elsewhere, for which the primary end point has already been reported. The combined analysis of SOFT-TEXT compared outcomes in 4,690 premenopausal women with estrogen/progesterone receptor–positive (ER/PgR+) early breast cancer randomly assigned to 5 years of exemestane + ovarian function suppression (OFS) versus tamoxifen + OFS. After a median follow-up of 9 years, exemestane + OFS significantly improved disease-free survival (DFS) and distant recurrence-free interval (DRFI), but not overall survival, compared with tamoxifen + OFS. We now report DFS, DRFI, and overall survival after a median follow-up of 13 years. In the intention-to-treat (ITT) population, the 12-year DFS (4.6% absolute improvement, hazard ratio [HR], 0.79; 95% CI, 0.70 to 0.90; P < .001) and DRFI (1.8% absolute improvement, HR, 0.83; 95% CI, 0.70 to 0.98; P = .03), but not overall survival (90.1% v 89.1%, HR, 0.93; 95% CI, 0.78 to 1.11), continued to be significantly improved for patients assigned exemestane + OFS over tamoxifen + OFS. Among patients with human epidermal growth factor receptor 2-negative tumors (86.0% of the ITT population), the absolute improvement in 12-year overall survival with exemestane + OFS was 2.0% (HR, 0.85; 95% CI, 0.70 to 1.04) and 3.3% in those who received chemotherapy (45.9% of the ITT population). Overall survival benefit was clinically significant in high-risk patients, eg, women age < 35 years (4.0%) and those with > 2 cm (4.5%) or grade 3 tumors (5.5%). These sustained reductions of the risk of recurrence with adjuvant exemestane + OFS, compared with tamoxifen + OFS, provide guidance for selecting patients for whom exemestane should be preferred over tamoxifen in the setting of OFS. [Media: see text]
- Published
- 2023
26. Adjuvant Endocrine Therapy in Premenopausal Breast Cancer: 12-Year Results From SOFT
- Author
-
Prudence A, Francis, Gini F, Fleming, István, Láng, Eva M, Ciruelos, Hervé R, Bonnefoi, Meritxell, Bellet, Antonio, Bernardo, Miguel A, Climent, Silvana, Martino, Begoña, Bermejo, Harold J, Burstein, Nancy E, Davidson, Charles E, Geyer, Barbara A, Walley, James N, Ingle, Robert E, Coleman, Bettina, Müller, Fanny, Le Du, Sibylle, Loibl, Eric P, Winer, Barbara, Ruepp, Sherene, Loi, Marco, Colleoni, Alan S, Coates, Richard D, Gelber, Aron, Goldhirsch, and Meredith M, Regan
- Subjects
Cancer Research ,Oncology - Abstract
Clinical trials frequently include multiple end points that mature at different times. The initial report, typically based on the primary end point, may be published when key planned co-primary or secondary analyses are not yet available. Clinical Trial Updates provide an opportunity to disseminate additional results from studies, published in JCO or elsewhere, for which the primary end point has already been reported. The Suppression of Ovarian Function Trial (SOFT; ClinicalTrials.gov identifier: NCT00066690 ) randomly assigned premenopausal women with hormone receptor–positive breast cancer to 5 years of adjuvant tamoxifen, tamoxifen plus ovarian function suppression (OFS), or exemestane plus OFS. The primary analysis compared disease-free survival (DFS) between tamoxifen plus OFS versus tamoxifen alone; exemestane plus OFS versus tamoxifen was a secondary objective. After 8 years, SOFT reported a significant reduction in recurrence and improved overall survival (OS) with adjuvant tamoxifen plus OFS versus tamoxifen alone. Here, we report outcomes after median follow-up of 12 years. DFS remained significantly improved with tamoxifen plus OFS versus tamoxifen (hazard ratio, 0.82; 95% CI, 0.69 to 0.98) with a 12-year DFS of 71.9% with tamoxifen, 76.1% with tamoxifen plus OFS, and 79.0% with exemestane plus OFS. OS was improved with tamoxifen plus OFS versus tamoxifen (hazard ratio, 0.78; 95% CI, 0.60 to 1.01) and was 86.8% with tamoxifen, 89.0% with tamoxifen plus OFS, and 89.4% with exemestane plus OFS at 12 years. Among those who received prior chemotherapy for human epidermal growth factor receptor-2–negative tumors, OS was 78.8% with tamoxifen, 81.1% with tamoxifen plus OFS, and 84.4% with exemestane plus OFS. In conclusion, after 12 years, there remains a benefit from including OFS in adjuvant endocrine therapy, with an absolute improvement in OS more apparent with higher baseline risk of recurrence. [Media: see text]
- Published
- 2022
27. Customizing local and systemic therapies for women with early breast cancer: the St. Gallen International Consensus Guidelines for treatment of early breast cancer 2021
- Author
-
H.J. Burstein, G. Curigliano, B. Thürlimann, W.P. Weber, P. Poortmans, M.M. Regan, H.J. Senn, E.P. Winer, M. Gnant, Stephan Aebi, Fabrice André, Carlos Barrios, Jonas Bergh, Herve Bonnefoi, Denisse Bretel Morales, Sara Brucker, Harold Burstein, David Cameron, Fatima Cardoso, Lisa Carey, Boon Chua, Eva Ciruelos, Marco Colleoni, Giuseppe Curigliano, Suzette Delaloge, Carsten Denkert, Peter Dubsky, Bent Ejlertsen, Florian Fitzal, Prudence Francis, Viviana Galimberti, Hebatallah Gamal El Din Mohamed Mahmoud, Judy Garber, Michael Gnant, William Gradishar, Bahadir Gulluoglu, Nadia Harbeck, Chiun-Sheng Huang, Jens Huober, Andre Ilbawi, Zefei Jiang, Steven Johnston, Eun Sook Lee, Sibylle Loibl, Monica Morrow, Ann Partridge, Martine Piccart, Philip Poortmans, Aleix Prat, Meredith Regan, Isabella Rubio, Hope Rugo, Emiel Rutgers, Felix Sedlmayer, Vladimir Semiglazov, Hans-Joerg Senn, Zhiming Shao, Tanja Spanic, Petra Tesarova, Beat Thürlimann, Sergei Tjulandin, Masakazu Toi, Maureen Trudeau, Nicholas Turner, Inez Vaz Luis, Giuseppe Viale, Toru Watanabe, Walter P. Weber, Eric P. Winer, Binghe Xu, and Panelists of the St Gallen Consensus Conference
- Subjects
medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,medicine.medical_treatment ,media_common.quotation_subject ,Hematology ,medicine.disease ,Radiation therapy ,Presentation ,Breast cancer ,Oncology ,Multidisciplinary approach ,Family medicine ,Pandemic ,medicine ,Human medicine ,business ,Neoadjuvant therapy ,media_common ,Early breast cancer ,Genetic testing - Abstract
The 17th St Gallen International Breast Cancer Consensus Conference in 2021 was held virtually, owing to the global COVID-19 pandemic. More than 3300 participants took part in this important bi-annual critical review of the 'state of the art' in the multidisciplinary care of early-stage breast cancer. Seventy-four expert panelists (see Appendix 1) from all continents discussed and commented on the previously elaborated consensus questions, as well as many key questions on early breast cancer diagnosis and treatment asked by the audience. The theme of this year's conference was 'Customizing local and systemic therapies.' A well-organized program of pre-recorded symposia, live panel discussions and real-time panel voting results drew a worldwide audience of thousands, reflecting the farreaching impact of breast cancer on every continent. The interactive technology platform allowed, for the first time, audience members to ask direct questions to panelists, and to weigh in with their own vote on several key panel questions. A hallmark of this meeting was to focus on customized recommendations for treatment of early-stage breast cancer. There is increasing recognition that the care of a breast cancer patient depends on highly individualized clinical features, including the stage at presentation, the biological subset of breast cancer, the genetic factors that may underlie breast cancer risk, the genomic signatures that inform treatment recommendations, the extent of response before surgery in patients who receive neoadjuvant therapy, and patient preferences. This customized approach to treatment requires integration of clinical care between patients and radiology, pathology, genetics, and surgical, medical and radiation oncology providers. It also requires a dynamic response from clinicians as they encounter accumulating clinical information at the time of diagnosis and then serially with each step in the treatment plan and follow-up, reflecting patient experiences and treatment response.
- Published
- 2021
28. Prospective Study Testing a Simplified Paclitaxel Premedication Regimen in Patients with Early Breast Cancer
- Author
-
Michael Constantine, Romualdo Barroso-Sousa, Tianyu Li, Eric P. Winer, Meredith Faggen, Antonio Di Meglio, Nabihah Tayob, Harold J. Burstein, Caroline Block, Natalie Faye Sinclair, Rebecca Rees, Nan Lin, Ann H. Partridge, Ines Vaz-Luis, Sara M. Tolaney, Jiani Hu, Michael J. Hassett, Jose Pablo Leone, Lindsey Milisits, Frederick Briccetti, Lorenzo Trippa, and Adrienne G. Waks
- Subjects
Cancer Research ,Paclitaxel ,Dose-dense chemotherapy ,medicine.drug_class ,Premedication ,Breast Neoplasms ,chemistry.chemical_compound ,Dose‐dense chemotherapy ,Breast Cancer ,Hypersensitivity ,Humans ,Medicine ,Prospective Studies ,Prospective cohort study ,Dexamethasone ,business.industry ,Diphenhydramine ,Regimen ,Oncology ,chemistry ,Anesthesia ,Corticosteroid ,Female ,business ,medicine.drug - Abstract
Background In early trials, hypersensitivity reactions (HSRs) to paclitaxel were common, thus prompting the administration of antihistamines and corticosteroids before every paclitaxel dose. We tested the safety of omitting corticosteroids after cycle 2 during the paclitaxel portion of the dose‐dense (DD) doxorubicin‐cyclophosphamide (AC)–paclitaxel regimen. Patients, Materials, and Methods In this prospective, single‐arm study, patients who completed four cycles of DD‐AC for stage I–III breast cancer received paclitaxel 175 mg/m2 every 2 weeks for four cycles. Patients received a standard premedication protocol containing dexamethasone, diphenhydramine, and a histamine H2 blocker prior to the first two paclitaxel cycles. Dexamethasone was omitted in cycles three and four if there were no HSRs in previous cycles. We estimated the rate of grade 3–4 HSRs. Results Among 127 patients enrolled, 125 received more than one dose of protocol therapy and are included in the analysis. Fourteen (11.2%; 90% confidence interval, 6.9%–20.0%) patients had any‐grade HSRs, for a total of 22 (4.5%; 3.1%–6.4%) HSRs over 486 paclitaxel cycles. Any‐grade HSRs occurred in 1.6% (0.3%–5.0%), 6.5% (3.3%–11.3%), 7.4% (3.9%–12.5%), and 2.6% (0.7%–6.6%) of patients after paclitaxel cycles 1, 2, 3, and 4, respectively. Dexamethasone use was decreased by 92.8% in cycles 3 and 4. Only one patient experienced grade 3 HSR in cycles 3 or 4, for a rate of grade 3/4 HSR 0.4% (0.02%–2.0%) (1/237 paclitaxel infusions). That patient had grade 2 HSR during cycle 2, and the subsequent grade 3 event occurred despite usual dexamethasone premedication. A sensitivity analysis restricted to patients not known to have received dexamethasone in cycles 3 and 4 found that any‐grade HSRs occurred in 2.7% (3/111; 0.7%–6.8%) and 0.9% (1/109; 0.05%–4.3%) of patients in cycle 3 and 4, respectively. Conclusion Corticosteroid premedication can be safely omitted in cycles 3 and 4 of dose‐dense paclitaxel if HSRs are not observed during cycles 1 and 2. Implications for Practice Because of the potential for hypersensitivity reactions (HSRs) to paclitaxel, corticosteroids are routinely prescribed prior to each dose, on an indefinite basis. This prospective study, including 125 patients treated with 486 paclitaxel cycles, demonstrates that corticosteroids can be safely omitted in future cycles if HSRs did not occur during cycles 1 and 2 of paclitaxel and that this strategy reduces the use of corticosteroids in cycles 3 and 4 by 92.8% relative to current standard of care., To avoid hypersensitivity reactions, corticosteroids are routinely prescribed before each dose of paclitaxel. This article reports the results of a study that focused on whether corticosteroids could be safely omitted in later cycles of treatment if reactions did not occur during earlier cycles.
- Published
- 2021
29. Molecular correlates of response to eribulin and pembrolizumab in hormone receptor-positive metastatic breast cancer
- Author
-
Romualdo Barroso-Sousa, Tanya Keenan, Elizabeth A. Mittendorf, Anita Giobbie-Hurder, Ann H. Partridge, Sara M. Tolaney, Gerburg M. Wulf, Laura Spring, Rinath Jeselsohn, Jake Conway, Brendan Reardon, Tianyu Li, Tess O’Meara, Ian E. Krop, Ines Vaz-Luis, Jihye Park, Erin Shannon, Jiani Hu, Victoria Attaya, Meng Xiao He, Eric P. Winer, Leilani Anderson, Nabihah Tayob, Judith Agudo, Jennifer L. Guerriero, Eliezer M. Van Allen, Nan Lin, Mariano Severgnini, and David Liu
- Subjects
Male ,Oncology ,General Physics and Astronomy ,Phases of clinical research ,Pembrolizumab ,B7-H1 Antigen ,chemistry.chemical_compound ,Breast cancer ,Antineoplastic Combined Chemotherapy Protocols ,Neoplasm Metastasis ,Immune Checkpoint Inhibitors ,Antigen Presentation ,Multidisciplinary ,Genomics ,Ketones ,Middle Aged ,Metastatic breast cancer ,Survival Rate ,Treatment Outcome ,Receptors, Estrogen ,Cytokines ,Biomarker (medicine) ,Female ,Receptors, Progesterone ,Signal Transduction ,Eribulin ,Adult ,medicine.medical_specialty ,Combination therapy ,Science ,Breast Neoplasms ,Antibodies, Monoclonal, Humanized ,Article ,General Biochemistry, Genetics and Molecular Biology ,Genetic Heterogeneity ,Lymphocytes, Tumor-Infiltrating ,Internal medicine ,medicine ,Humans ,Furans ,Aged ,Genome, Human ,business.industry ,Gene Expression Profiling ,Cancer ,Estrogens ,General Chemistry ,medicine.disease ,chemistry ,Drug Resistance, Neoplasm ,Mutation ,business - Abstract
Immune checkpoint inhibitors (ICIs) have minimal therapeutic effect in hormone receptor-positive (HR+ ) breast cancer. We present final overall survival (OS) results (n = 88) from a randomized phase 2 trial of eribulin ± pembrolizumab for patients with metastatic HR+ breast cancer, computationally dissect genomic and/or transcriptomic data from pre-treatment tumors (n = 52) for molecular associations with efficacy, and identify cytokine changes differentiating response and ICI-related toxicity (n = 58). Despite no improvement in OS with combination therapy (hazard ratio 0.95, 95% CI 0.59–1.55, p = 0.84), immune infiltration and antigen presentation distinguished responding tumors, while tumor heterogeneity and estrogen signaling independently associated with resistance. Moreover, patients with ICI-related toxicity had lower levels of immunoregulatory cytokines. Broadly, we establish a framework for ICI response in HR+ breast cancer that warrants diagnostic and therapeutic validation. ClinicalTrials.gov Registration: NCT03051659., A randomized phase 2 clinical trial has recently shown no benefit of the combination eribulin and pembrolizumab over pembrolizumab alone in HR + metastatic breast cancer patients (NCT03051659). Here, the authors are reporting the final OS data and biomarker analyses on a subset of samples to analyze molecular correlates
- Published
- 2021
30. Treatment discontinuation, patient-reported toxicities and quality-of-life by age following trastuzumab emtansine or paclitaxel/trastuzumab (ATEMPT)
- Author
-
Tal Sella, Yue Zheng, Nabihah Tayob, Kathryn J. Ruddy, Rachel A. Freedman, Chau Dang, Denise Yardley, Steven J. Isakoff, Vicente Valero, Michelle DeMeo, Harold J. Burstein, Eric P. Winer, Antonio C. Wolff, Ian Krop, Ann H. Partridge, and Sara M. Tolaney
- Subjects
Oncology ,Pharmacology (medical) ,Radiology, Nuclear Medicine and imaging - Abstract
In the ATEMPT trial, adjuvant trastuzumab emtansine (T-DM1) compared to paclitaxel plus trastuzumab (TH) for stage I HER2-positive breast cancer improved patient-reported outcomes (PROs), while maintaining excellent disease outcomes. We report treatment discontinuation and use multivariable models to compare, patient-reported toxicity and quality-of-life (QOL) by age (≤50, >50) and treatment arm at 18 months post-enrollment among 366 eligible participants randomized in a 3:1 ratio to T-DM1 or TH. T-DM1 discontinuation was higher among women >50 vs. ≤50 (23% vs. 9%, p = 0.003, Fisher’s Exact test) with 4%, 8%, and 17% of older patients discontinuing treatment by 3, 6, and 9 months, respectively. Superior QOL with T-DM1 vs. TH was observed among women ≤50 with estimated mean difference of 6.48 (95% confidence interval (CI) 0.51–12.46) and driven by better social/family well-being and breast cancer-specific sub-scores. Among women >50, T-DM1 was associated with superior physical well-being and less activity impairment, with no differences in global QOL. Older women had decreased neuropathy with T-DM1 vs. TH. De-escalated treatment regimens for HER2 positive breast cancer may have age-varying impact on treatment tolerance, toxicities and subsequent QOL, which should be considered when selecting therapy options.Clinical Trial Registration: ClinicalTrials.gov, NCT01853748
- Published
- 2022
31. Updated Standardized Definitions for Efficacy End Points (STEEP) in Adjuvant Breast Cancer Clinical Trials: STEEP Version 2.0
- Author
-
Jane Perlmutter, Victoria S. Blinder, Laleh Amiri-Kordestani, Jared C. Foster, Eileen Rakovitch, Eric P. Winer, Angelo DiLeo, Julia White, Anthony D. Elias, Ana F. Best, Lynn Pearson Butler, Sara M. Tolaney, Lawrence Baizer, Patricia A. Spears, Elena Schwartz, David Cameron, Elizabeth Garrett-Mayer, Neelima Denduluri, Elizabeth S. Frank, Larissa A. Korde, E. Shelley Hwang, Nadine Tung, and Judith M Bliss
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Endpoint Determination ,business.industry ,medicine.medical_treatment ,MEDLINE ,Breast Neoplasms ,medicine.disease ,Clinical trial ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Research Design ,030220 oncology & carcinogenesis ,Internal medicine ,Special Articles ,Humans ,Medicine ,Female ,030212 general & internal medicine ,business ,Adjuvant - Abstract
PURPOSEThe Standardized Definitions for Efficacy End Points (STEEP) criteria, established in 2007, provide standardized definitions of adjuvant breast cancer clinical trial end points. Given the evolution of breast cancer clinical trials and improvements in outcomes, a panel of experts reviewed the STEEP criteria to determine whether modifications are needed.METHODSWe conducted systematic searches of ClinicalTrials.gov for adjuvant systemic and local-regional therapy trials for breast cancer to investigate if the primary end points reported met STEEP criteria. On the basis of common STEEP deviations, we performed a series of simulations to evaluate the effect of excluding non–breast cancer deaths and new nonbreast primary cancers from the invasive disease–free survival end point.RESULTSAmong 11 phase III breast cancer trials with primary efficacy end points, three had primary end points that followed STEEP criteria, four used STEEP definitions but not the corresponding end point names, and four used end points that were not included in the original STEEP manuscript. Simulation modeling demonstrated that inclusion of second nonbreast primary cancer can increase the probability of incorrect inferences, can decrease power to detect clinically relevant efficacy effects, and may mask differences in recurrence rates, especially when recurrence rates are low.CONCLUSIONWe recommend an additional end point, invasive breast cancer–free survival, which includes all invasive disease–free survival events except second nonbreast primary cancers. This end point should be considered for trials in which the toxicities of agents are well-known and where the risk of second primary cancer is small. Additionally, we provide end point recommendations for local therapy trials, low-risk populations, noninferiority trials, and trials incorporating patient-reported outcomes.
- Published
- 2021
32. Phase II trial of veliparib and temozolomide in metastatic breast cancer patients with and without BRCA1/2 mutations
- Author
-
Jing Xu, Nadine Tung, Beth Overmoyer, Rebecca Gelman, Karleen Habin, Judy Garber, Eric P. Winer, Tanya Keenan, Leif W. Ellisen, Steven J. Isakoff, Bruce A. Chabner, Beow Y. Yeap, and Paul E. Goss
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Temozolomide ,endocrine system diseases ,Veliparib ,business.industry ,BRCA mutation ,medicine.disease ,Metastatic breast cancer ,chemistry.chemical_compound ,Breast cancer ,chemistry ,Tolerability ,Internal medicine ,Cohort ,medicine ,Clinical endpoint ,skin and connective tissue diseases ,business ,medicine.drug - Abstract
We evaluated the efficacy and safety of poly-(adenosine diphosphate-ribose) polymerase (PARP) 1 and 2 inhibitor veliparib and temozolomide in metastatic breast cancer patients with and without germline BRCA1/2 mutations. In this single-arm phase II trial, patients with metastatic breast cancer received veliparib 30 to 40 mg twice daily on days 1 to 7 with concurrent temozolomide 150 mg/m2 on days 1 to 5 of a 28-day cycle. The primary cohort was unselected for BRCA mutation status, and an expansion cohort enrolled only BRCA1/2 carriers. The primary endpoint was objective response rate (ORR) in each cohort. Secondary endpoints included progression-free survival (PFS), clinical benefit rate (CBR), and evaluation of safety and tolerability. In the primary cohort of 41 unselected patients, which included 9 BRCA mutation carriers, the ORR was 10% and clinical benefit rate at 4 months (CBR) was 27%. In the expansion cohort of 21 BRCA1/2 carriers, the ORR was 14% and CBR was 43%. Among all 30 BRCA1/2 carriers, the ORR was 23% versus 0% among non-carriers. In the subset of BRCA1/2 carriers, the ORR was 32% among platinum-naive patients versus 9% among platinum-exposed patients. The median PFS was 3.3 months among BRCA1/2 carriers compared to 1.8 months among non-carriers (HR: 0.48, p = 0.006). A longer median PFS of 6.2 months was observed among BRCA1/2 carriers who had no prior platinum therapy. The most common grade 3 and 4 toxicities were thrombocytopenia (32%) and neutropenia (21%) that generally improved with dose modifications. Veliparib and temozolomide demonstrated clinical activity in platinum-naive BRCA-associated metastatic breast cancer with manageable toxicity at doses of veliparib well below the single-agent active dose. Although the study did not meet its primary endpoint in unselected nor BRCA-associated breast cancer, this regimen was further evaluated in the BROCADE 2 study. NCT01009788 (ClinicalTrials.gov), November 9, 2009
- Published
- 2021
33. Small molecule inhibition of deubiquitinating enzyme JOSD1 as a novel targeted therapy for leukemias with mutant JAK2
- Author
-
Wai Cheung Chan, Lucia Cabal-Hierro, James D. Griffin, Sara J. Buhrlage, Jing Yang, Jarrod A. Marto, Eric P. Winer, Robert S. Magin, Ellen Weisberg, Bin Hu, Xiaoxi Liu, Shengzhe Zhang, Chengcheng Meng, Richard Stone, Laura M. Doherty, Martin Sattler, Ilaria Lamberto, and Nathan J. Schauer
- Subjects
Cancer Research ,Janus kinase 2 ,biology ,Kinase ,Chemistry ,Mutant ,food and beverages ,Myeloid leukemia ,Hematology ,Protein degradation ,Deubiquitinating enzyme ,Oncology ,hemic and lymphatic diseases ,biology.protein ,Cancer research ,Kinase activity ,Chemical genetics ,hormones, hormone substitutes, and hormone antagonists - Abstract
Mutations in the Janus Kinase 2 (JAK2) gene resulting in constitutive kinase activation represent the most common genetic event in myeloproliferative neoplasms (MPN), a group of diseases involving overproduction of one or more kinds of blood cells, including red cells, white cells, and platelets. JAK2 kinase inhibitors, such as ruxolitinib, provide clinical benefit, but inhibition of wild-type (wt) JAK2 limits their clinical utility due to toxicity to normal cells, and small molecule inhibition of mutated JAK2 kinase activity can lead to drug resistance. Here, we present a strategy to target mutated JAK2 for degradation, using the cell's intracellular degradation machinery, while sparing non-mutated JAK2. We employed a chemical genetics screen, followed by extensive selectivity profiling and genetic studies, to identify the deubiquitinase (DUB), JOSD1, as a novel regulator of mutant JAK2. JOSD1 interacts with and stabilizes JAK2-V617F, and inactivation of the DUB leads to JAK2-V617F protein degradation by increasing its ubiquitination levels, thereby shortening its protein half-life. Moreover, targeting of JOSD1 leads to the death of JAK2-V617F-positive primary acute myeloid leukemia (AML) cells. These studies provide a novel therapeutic approach to achieving selective targeting of mutated JAK2 signaling in MPN.
- Published
- 2021
34. PD-L1 Immunohistochemistry Assay Comparison in Atezolizumab Plus nab-Paclitaxel–Treated Advanced Triple-Negative Breast Cancer
- Author
-
Carlos H. Barrios, Mark M. Kockx, Véronique Diéras, Eric P. Winer, Stephen Y. Chui, Luciana Molinero, Sylvia Adams, Hope S. Rugo, Hiroji Iwata, Dieter Peeters, Andreas Schneeweiss, Sherene Loi, Giuseppe Viale, Jennifer C Lin, Anh Nguyen Duc, Peter Schmid, and Leisha A. Emens
- Subjects
Cancer Research ,medicine.medical_specialty ,Lung Neoplasms ,Concordance ,Population ,Oncology and Carcinogenesis ,Triple Negative Breast Neoplasms ,Gastroenterology ,B7-H1 Antigen ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Atezolizumab ,Internal medicine ,Breast Cancer ,medicine ,Biomarkers, Tumor ,Humans ,Oncology & Carcinogenesis ,education ,Triple-negative breast cancer ,030304 developmental biology ,Cancer ,0303 health sciences ,education.field_of_study ,business.industry ,Hazard ratio ,Editorials ,Evaluation of treatments and therapeutic interventions ,Articles ,medicine.disease ,Immunohistochemistry ,Confidence interval ,Good Health and Well Being ,Oncology ,030220 oncology & carcinogenesis ,6.1 Pharmaceuticals ,business ,AcademicSubjects/MED00010 - Abstract
Background In the phase III IMpassion130 study, atezolizumab plus nab-paclitaxel (A+nP) showed clinical benefit in advanced or metastatic triple-negative breast cancer patients who were programmed death-ligand 1 (PD-L1)+ (tumor-infiltrating immune cells [IC] ≥1%) using the SP142 immunohistochemistry assay. Here we evaluate 2 other PD-L1 assays for analytical concordance with SP142 and patient-associated clinical outcomes. Methods Samples from 614 patients (68.1% of intention-to-treat population) were centrally evaluated by immunohistochemistry for PD-L1 status on IC (VENTANA SP142, SP263, Dako 22C3) or as a combined positive score (CPS; 22C3). Results Using SP142, SP263, and 22C3 assays, PD-L1 IC ≥1% prevalence was 46.4% (95% confidence interval [CI] = 42.5% to 50.4%), 74.9% (95% CI = 71.5% to 78.3%), and 73.1% (95% CI = 69.6% to 76.6%), respectively; 80.9% were 22C3 CPS ≥1. At IC ≥1% (+), the analytical concordance between SP142 and SP263 and 22C3 was 69.2% and 68.7%, respectively. Almost all SP142+ cases were captured by other assays (double positive), but several SP263+ (29.6%) or 22C3+ (29.0%) cases were SP142– (single positive). A+nP clinical activity vs placebo+nP in SP263+ and 22C3+ patients (progression-free survival [PFS] hazard ratios [HRs] = 0.64 to 0.68; overall survival [OS] HRs = 0.75 to 0.79) was driven by double-positive cases (PFS HRs = 0.60 to 0.61; OS HRs = 0.71 to 0.75) rather than single-positive cases (PFS HRs = 0.68 to 0.81; OS HRs = 0.87 to 0.95). Concordance for harmonized cutoffs for SP263 (IC ≥4%) and 22C3 (CPS ≥10) to SP142 (IC ≥1%) was subpar (approximately 75%). Conclusions 22C3 and SP263 assays identified more patients as PD-L1+ (IC ≥1%) than SP142. No inter-assay analytical equivalency was observed. Consistent improved A+nP efficacy was captured by the SP142 PD-L1 IC ≥1% subgroup nested within 22C3 and SP263 PD-L1+ (IC ≥1%) populations.
- Published
- 2021
35. Multidimensional Molecular Profiling of Metastatic Triple-Negative Breast Cancer and Immune Checkpoint Inhibitor Benefit
- Author
-
Romualdo Barroso-Sousa, Juliet Forman, Katharine Collier, Zachary T. Weber, Tejas R. Jammihal, Katrina Z. Kao, Edward T. Richardson, Tanya Keenan, Ofir Cohen, Michael P. Manos, Ryan C. Brennick, Patrick A. Ott, F. Stephen Hodi, Deborah A. Dillon, Victoria Attaya, Tess O'Meara, Nancy U. Lin, Eliezer M. Van Allen, Scott Rodig, Eric P. Winer, Elizabeth A. Mittendorf, Catherine J. Wu, Nikhil Wagle, Daniel G. Stover, Sachet A. Shukla, and Sara M. Tolaney
- Subjects
Cancer Research ,Oncology ,Mutation ,Biomarkers, Tumor ,Humans ,Triple Negative Breast Neoplasms ,ORIGINAL REPORTS ,Immune Checkpoint Inhibitors ,Progression-Free Survival - Abstract
PURPOSE In metastatic triple-negative breast cancer (mTNBC), consistent biomarkers of immune checkpoint inhibitor (ICI) therapy benefit remain elusive. We evaluated the immune, genomic, and transcriptomic landscape of mTNBC in patients treated with ICIs. METHODS We identified 29 patients with mTNBC treated with pembrolizumab or atezolizumab, either alone (n = 9) or in combination with chemotherapy (n = 14) or targeted therapy (n = 6), who had tumor tissue and/or blood available before ICI therapy for whole-exome sequencing. RNA sequencing and CIBERSORTx-inferred immune population analyses were performed (n = 20). Immune cell populations and programmed death-ligand 1 expression were assessed using multiplexed immunofluorescence (n = 18). Clonal trajectories were evaluated via serial tumor/circulating tumor DNA whole-exome sequencing (n = 4). Association of biomarkers with progression-free survival and overall survival (OS) was assessed. RESULTS Progression-free survival and OS were longer in patients with high programmed death-ligand 1 expression and tumor mutational burden. Patients with longer survival also had a higher relative inferred fraction of CD8+ T cells, activated CD4+ memory T cells, M1 macrophages, and follicular helper T cells and enrichment of inflammatory gene expression pathways. A mutational signature of defective repair of DNA damage by homologous recombination was enriched in patients with both shorter OS and primary resistance. Exploratory analysis of clonal evolution among four patients treated with programmed cell death protein 1 blockade and a tyrosine kinase inhibitor suggested that clonal stability post-treatment was associated with short time to progression. CONCLUSION This study identified potential biomarkers of response to ICIs among patients with mTNBC: high tumor mutational burden; presence of CD8+, CD4 memory T cells, follicular helper T cells, and M1 macrophages; and inflammatory gene expression pathways. Pretreatment deficiencies in the homologous recombination DNA damage repair pathway and the absence of or minimal clonal evolution post-treatment may be associated with worse outcomes.
- Published
- 2022
36. Efficacy of neoadjuvant chemotherapy in male breast cancer compared with female breast cancer
- Author
-
José Pablo Leone, Michael J. Hassett, Julieta Leone, Sara M. Tolaney, Carlos T. Vallejo, Bernardo A. Leone, Eric P. Winer, and Nancy U. Lin
- Subjects
Male ,Cancer Research ,Oncology ,Chemotherapy, Adjuvant ,Receptor, ErbB-2 ,Antineoplastic Combined Chemotherapy Protocols ,Humans ,Breast Neoplasms ,Female ,Prognosis ,Neoadjuvant Therapy ,Breast Neoplasms, Male - Abstract
Neoadjuvant chemotherapy (NAC) is standard for many females with breast cancer (FBC). The efficacy of NAC in male breast cancer (MaBC) is unclear. The aim of this study was to compare proportions of pathologic complete response (pCR) between MaBC and FBC by tumor subtype (TS).MaBC and FBC treated with NAC between 2010 and 2016, with known TS, were evaluated from the National Cancer Database. Proportions of pCR (ypT0/Tis ypN0) were compared between sexes within TS by Fisher test. Multivariable logistic regression assessed the independent association of sex with pCR. Overall survival (OS) was estimated by Kaplan-Meier.A total of 385 MaBC and 68,065 FBC were included. Median time from initiation of NAC to surgery was 143 days in MaBC and 148 days in FBC. Proportions of pCR in MaBC and FBC by TS were: hormone receptor-positive/human epidermal growth factor receptor 2-negative (HR+/HER2-): 4.9% vs 9.7%, p = .01; HR+/HER2+: 16.1% vs 33.6%, p .001; HR-/HER2+: 44.0% vs 53.2%, p = .42; and HR-/HER2-: 21.4% vs 32.1%, p = .18, respectively. FBC had twice the odds of pCR than MaBC (adjusted odds ratio, 2.0; 95% CI, 1.5-2.8; p .001). Five-year OS for MaBC with pCR vs not was 90% vs 64.7%; p = .02. Five-year OS for FBC with pCR vs not was 91.9% vs 75.3%; p .01.Proportions and odds of pCR to NAC were numerically lower in MaBC compared with FBC for each TS and statistically significant for HR+/HER2- and HR+/HER2+. The independent association of sex with pCR was confirmed in multivariable analysis. pCR is prognostic in both MaBC and FBC.
- Published
- 2022
37. Genomic features of rapid versus late relapse in triple negative breast cancer
- Author
-
Meghan Wyse, Yi-Zhou Jiang, Leming Shi, Zhi-Ming Shao, David Tallman, Jerome F. Bey, Sagar Sardesai, Maryam B. Lustberg, Mathew Cherian, Elizabeth J. Adams, Steven T. Sizemore, Robert Wesolowski, Daniel G. Stover, Jeffrey VanDeusen, Claire F. Verschraegen, Jasneet Singh, Gina M. Sizemore, Nan Lin, Eric P. Winer, William Nock, Zachary Weber, Samilia Obeng-Gyasi, Ding Ma, Sarah Asad, Kristin L. Dean, Nicole Williams, Bhuvaneswari Ramaswamy, Peng Wang, Yiqing Zhang, Sinclair Stockard, and Wei Huang
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,Time Factors ,Datasets as Topic ,Triple Negative Breast Neoplasms ,Disease ,Logistic regression ,Transcriptome ,0302 clinical medicine ,Surgical oncology ,Triple-negative breast cancer ,Fisher's exact test ,Mastectomy ,RC254-282 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Middle Aged ,Prognosis ,Primary tumor ,Neoadjuvant Therapy ,Gene Expression Regulation, Neoplastic ,Chemotherapy, Adjuvant ,030220 oncology & carcinogenesis ,symbols ,Female ,Adult ,medicine.medical_specialty ,DNA Copy Number Variations ,Risk Assessment ,Disease-Free Survival ,03 medical and health sciences ,symbols.namesake ,Breast cancer ,Internal medicine ,Breast Cancer ,Machine learning ,Genetics ,medicine ,Biomarkers, Tumor ,Humans ,Models, Genetic ,business.industry ,Research ,Gene Expression Profiling ,medicine.disease ,030104 developmental biology ,Logistic Models ,Mutation ,Neoplasm Recurrence, Local ,business ,Follow-Up Studies - Abstract
Background Triple-negative breast cancer (TNBC) is a heterogeneous disease and we have previously shown that rapid relapse of TNBC is associated with distinct sociodemographic features. We hypothesized that rapid versus late relapse in TNBC is also defined by distinct clinical and genomic features of primary tumors. Methods Using three publicly-available datasets, we identified 453 patients diagnosed with primary TNBC with adequate follow-up to be characterized as ‘rapid relapse’ (rrTNBC; distant relapse or death ≤2 years of diagnosis), ‘late relapse’ (lrTNBC; > 2 years) or ‘no relapse’ (nrTNBC: > 5 years no relapse/death). We explored basic clinical and primary tumor multi-omic data, including whole transcriptome (n = 453), and whole genome copy number and mutation data for 171 cancer-related genes (n = 317). Association of rapid relapse with clinical and genomic features were assessed using Pearson chi-squared tests, t-tests, ANOVA, and Fisher exact tests. We evaluated logistic regression models of clinical features with subtype versus two models that integrated significant genomic features. Results Relative to nrTNBC, both rrTNBC and lrTNBC had significantly lower immune signatures and immune signatures were highly correlated to anti-tumor CD8 T-cell, M1 macrophage, and gamma-delta T-cell CIBERSORT inferred immune subsets. Intriguingly, lrTNBCs were enriched for luminal signatures. There was no difference in tumor mutation burden or percent genome altered across groups. Logistic regression mModels that incorporate genomic features significantly outperformed standard clinical/subtype models in training (n = 63 patients), testing (n = 63) and independent validation (n = 34) cohorts, although performance of all models were overall modest. Conclusions We identify clinical and genomic features associated with rapid relapse TNBC for further study of this aggressive TNBC subset.
- Published
- 2021
38. Tumor subtypes and survival in male breast cancer
- Author
-
Bernardo Amadeo Leone, Nan Lin, Sara M. Tolaney, Carlos Teodoro Vallejo, Eric P. Winer, Julieta Leone, Jose Pablo Leone, and Rachel A. Freedman
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,Univariate analysis ,medicine.medical_specialty ,education.field_of_study ,Multivariate analysis ,Proportional hazards model ,business.industry ,Hazard ratio ,Population ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Breast cancer ,030220 oncology & carcinogenesis ,Male breast cancer ,Internal medicine ,medicine ,Stage (cooking) ,skin and connective tissue diseases ,business ,education - Abstract
Male breast cancer is an uncommon disease, and population-based information regarding prognostic factors is limited. Most cases are hormone receptor (HR) positive; however, the association of tumor subtype with overall survival (OS) and breast cancer-specific survival (BCSS) is unclear. Using SEER data, we identified men with invasive breast cancer between 2010 and 2017 with known HR and HER2 status. We examined tumor subtypes by patient characteristics and performed multivariate Cox proportional hazards analyses to determine the associations of each variable with OS and BCSS. We included 2389 men with a median follow-up of 43 months (IQR 19–68). Median age was 66 years. Tumor subtype distribution was 84.1% HR+/HER2−, 12.7% HR+/HER2+ , 0.8% HR−/HER2+, and 2.3% triple-negative (TN). In univariate analysis, OS at 5 years was 76.5% for HR+/HER2−, 65.1% for HR+/HER2+ , 84.2% for HR-/HER2+, and 48.1% for TN (p
- Published
- 2021
39. Twenty-year risks of breast cancer-specific mortality for stage III breast cancer in the surveillance, epidemiology, and end results registry
- Author
-
Carlos Teodoro Vallejo, Sara M. Tolaney, Nan Lin, Bernardo Amadeo Leone, Eric P. Winer, Julieta Leone, Nabihah Tayob, Jose Pablo Leone, Michael J. Hassett, and Rachel A. Freedman
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Disease ,medicine.disease ,Primary tumor ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Breast cancer ,030220 oncology & carcinogenesis ,Internal medicine ,Epidemiology ,Surveillance, Epidemiology, and End Results ,medicine ,Stage IIIC ,Stage (cooking) ,business ,Cause of death - Abstract
We aimed to report the 20-year risk of breast cancer-specific mortality (BCSM), report the risk of BCSM conditional on having survived 5 years, and identify factors associated with late deaths in stage III breast cancer. Using Surveillance, Epidemiology, and End Results data, we included women with stage III breast cancer diagnosed from 1990 to 2005. We excluded women with unknown hormone receptor (HR) status, women who did not undergo resection of the primary tumor or axillary nodes, or unknown cause of death. We estimated risks of BCSM using cumulative incidence function and used Fine and Gray regression to identify factors associated with late deaths. Final sample was 36,500 patients with 14 years of median follow-up. For each stage subgroup, the risk of BCSM at 20 years was significantly higher for HR-negative vs HR-positive tumors (IIIA: 49.8% vs 43.2%, P
- Published
- 2021
40. Abstract PS6-13: Population-based tool to estimate residual risks of breast cancer specific mortality (BCSM) and non-BCSM
- Author
-
Nan Lin, Sara M. Tolaney, Bernardo Amadeo Leone, Carlos Teodoro Vallejo, Julieta Leone, Rachel A. Freedman, Jose Pablo Leone, Michael J. Hassett, Eric P. Winer, and Nabihah Tayob
- Subjects
Residual risk ,Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Internal medicine ,medicine ,Population based ,Breast cancer specific ,business - Abstract
Background: The risk of breast cancer death persists for at least 20 years from diagnosis. Most reports describing this risk have been based on selected patients (pts) enrolled into clinical trials. These studies report absolute risks at fixed timepoints (i.e. 10 or 20 years) and do not consider the dynamic changes in risks over time. The aim of this study was to develop a tool to calculate residual risks of BCSM and non-BCSM based on individual pt and tumor characteristics, at any given time after breast cancer diagnosis. Methods: Using data from the Surveillance, Epidemiology, and End Results (SEER) program, we identified women diagnosed with non-metastatic invasive breast cancer between 1990-2005, with one primary cancer in their lifetime, and known hormone receptor (HR) status. We estimated the effect of baseline clinical and pathologic variables known to be prognostic, including pt age, HR status, tumor size (T), nodal status (N), and tumor grade, on residual cumulative risks of BCSM and non-BCSM over time. The tool generates the residual death risk (RDR), which is a nonparametric estimate of the cumulative risk of BCSM and non-BCSM by year 20 after any given time from initial diagnosis, among patients defined by baseline clinical and pathologic variables using the method of Gray (1988) implemented in the cmprsk package in R. Results: We included 235,015 pts (median follow-up = 14 years). Among all breast cancer deaths, the proportion occurring after 5 years was 60% for HR+ vs 24% for HR- (p BCSMnon-BCSMAll-cause mortality% Event-FreeCumulative risk (%)Cumulative risk (%)Cumulative risk (%)at 5 yat 10 yy 5-20y 0-20y 0-20y 0-20Nodal status by HR status (any T)HR+N097.293.98.610.633.243.8N192.183.920.025.225.550.8N281.566.236.145.621.266.8N367.749.150.163.116.179.2HR-N089.285.47.317.022.739.7N174.268.312.234.317.051.3N257.049.320.053.514.568.0N340.733.526.268.79.378.0Tumor size among N0 onlyHR+T1a99.097.64.25.030.435.4T1b98.997.25.15.934.440.2T1c97.694.38.710.532.943.3HR-T1a97.394.74.97.323.030.3T1b95.191.96.210.527.137.7T1c91.787.97.514.823.037.8 Citation Format: Jose P Leone, Sara M Tolaney, Bernardo A Leone, Rachel A Freedman, Michael J Hassett, Julieta Leone, Carlos T Vallejo, Eric P Winer, Nancy U Lin, Nabihah Tayob. Population-based tool to estimate residual risks of breast cancer specific mortality (BCSM) and non-BCSM [abstract]. In: Proceedings of the 2020 San Antonio Breast Cancer Virtual Symposium; 2020 Dec 8-11; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2021;81(4 Suppl):Abstract nr PS6-13.
- Published
- 2021
41. Abstract PS4-25: Comprehensive genomic analysis reveals molecular correlates of response to immune checkpoint inhibitors (ICI) in metastatic triple-negative breast cancer (mTNBC)
- Author
-
Zachary Weber, Ryan C. Brennick, Catherine J. Wu, F. Steve Hodi, Michael Manos, Romualdo Barroso-Sousa, Katherine A Collier, Deborah A. Dillon, Patrick A. Ott, Katrina Z. Kao, Sara M. Tolaney, Nikhil Wagle, Tanya Keenan, Scott J. Rodig, Elizabeth A. Mittendorf, Daniel G. Stover, Eliezer E. Van Allen, Nan Lin, Ofir Cohen, Sachet A. Shukla, Eric P. Winer, Juliet Forman, and Edward T. Richardson
- Subjects
Cancer Research ,Oncology ,business.industry ,Immune checkpoint inhibitors ,Cancer research ,Medicine ,business ,Triple-negative breast cancer - Abstract
Background: Genomic mechanisms associated with response to ICI in mTNBC are largely unknown. The aim of this work is to assess the genomic and immune profiles of mTNBC samples collected from patients (pts) treated with ICI. Methods: We identified 31 women with mTNBC treated with ICI (pembrolizumab, n=6, NCT02447003; atezolizumab, n=4, NCT01375842; nivolumab + cabozantinib, n = 6, NCT03316586; pembrolizumab + eribulin, n=8, NCT02513472; atezolizumab + nab-paclitaxel, n=7, NCT01633970) who had tumor tissue or blood available for sequencing obtained before and after ICI. Clinical benefit (CB), here defined as any objective response or stable disease (SD) for > 24 weeks, was observed in 20 pts (65%). An extraordinary responder was defined as having CB ≥ 2 yrs; 5 pts were considered extraordinary responders (range 26-60months). Whole exome sequencing (WES) was done on each tumor and on germline DNA from blood (23 pts had successful WES performed on samples collected before ICI; 5 of these had WES on samples taken after disease progression). RNA sequencing (RNAseq) was successfully performed in 18 of the tumors with WES performed on samples before ICI; and 3 of these had RNAseq on samples taken after disease progression. 18 pts had tumors assessed by multiplex immunofluorescence (mIF) panels encompassing CD4, CD8, PD-1, PD-L1, and cytokeratin on samples collected before ICI. WES, deep targeted panel and low coverage whole genome sequencing were performed on serially collected plasma samples from 22 pts to evaluate tumor fraction and specific mutations. The association between biomarkers and clinical benefit to ICI was assessed. Results: 21 of 31 pts (67%) had received ≥1 prior lines of systemic therapy in the metastatic setting before starting ICI. Among the most frequently mutated genes at baseline are: TP53 (57%); PIK3CA (18%); DNAH5, MYH8 (both 13%); KMT2C, AKT1, LAMA2 (all 9%). Pts with CB had a higher tumor mutational burden (TMB) than pts with no CB (p=0.018). Differential expression analysis of RNAseq data revealed an upregulation of several immune-related genes in pts with CB, indicating increased immune infiltration in that group. Gene set enrichment analysis of this expression data using hallmark and canonical pathway gene sets from MSigDB (nominal p-val < 0.05) showed that, compared to samples from pts without CB, extraordinary responders had elevated transcriptional signatures of several cancer-related pathways associated with cell survival, proliferation and metabolism, as well as genes associated with increased immune infiltration and upregulation of inflammatory response programs. The mIF showed that the tumor microenvironment (TME) of pts with CB were enriched in Cytokeratin-negative/PD-L1-positive cells compared to those without CB (p=0.014). Expression of CD4, CD8 and PD-1 was not significantly different between pts with and without CB. Genomic analysis of circulating tumor DNA, and tumor evolutionary analysis for pts with both pre- and post-ICI samples (acquired resistance) will be presented. Conclusions: Clinical benefit to ICI in mTNBC was associated with upregulation of immune-related pathways, enrichment of non-tumoral PD-L1-positive cells in TME, and high TMB. Citation Format: Romualdo Barroso-Sousa, Juliet Forman, Zachary T. Weber, Katherine Collier, Katrina Z. Kao, Edward T. Richardson, III, Tanya Keenan, Ofir Cohen, Michael P. Manos, Ryan C. Brennick, Patrick Ott, F. Steve Hodi, Deborah A. Dillon, Nancy U. Lin, Eliezer E. Van Allen, Scott Rodig, Eric P. Winer, Elizabeth A. Mittendorf, Catherine J. Wu, Daniel Stover, Nikhil Wagle, Sachet Shukla, Sara Tolaney. Comprehensive genomic analysis reveals molecular correlates of response to immune checkpoint inhibitors (ICI) in metastatic triple-negative breast cancer (mTNBC) [abstract]. In: Proceedings of the 2020 San Antonio Breast Cancer Virtual Symposium; 2020 Dec 8-11; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2021;81(4 Suppl):Abstract nr PS4-25.
- Published
- 2021
42. Abstract PD14-07: Association between biomarkers and response to pembrolizumab in patients with metastatic triple-negative breast cancer (mTNBC): Exploratory analysis from KEYNOTE-086
- Author
-
Sherene Loi, Peter Schmid, Javier Cortes, David W. Cescon, Eric P. Winer, Deborah L. Toppmeyer, Hope S. Rugo, Michelino De Laurentiis, Rita Nanda, Hiroji Iwata, Ahmad Awada, Antoinette R. Tan, Roberto Salgado, Vassiliki Karantza, Petar Jelinic, Anran Wang, Lingkang Huang, Razvan Cristescu, Lakshman Annamalai, Jennifer Yearley, and Sylvia Adams
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Cancer ,Pembrolizumab ,medicine.disease ,Exact test ,Breast cancer ,Internal medicine ,Cohort ,medicine ,Biomarker (medicine) ,business ,Exome ,Triple-negative breast cancer - Abstract
Background: In the phase 2 KEYNOTE-086 study (NCT02447003), pembrolizumab monotherapy had durable antitumor activity in a subset of patients with previously treated mTNBC (cohort A; n = 170) and in patients with previously untreated PD-L1-positive mTNBC (cohort B; n = 84). In this exploratory analysis of KEYNOTE-086, we evaluated the association between several biomarkers and response to pembrolizumab. Methods: Cohort A enrolled patients regardless of PD-L1 expression who had documented disease progression following ≥1 systemic therapy for metastatic disease. Cohort B enrolled patients with PD-L1-positive (combined positive score [CPS] ≥1) tumors who had not received prior systemic therapy for metastatic disease. Immunohistochemistry was used to measure PD-L1 CPS and CD8 density; H&E staining for percentage of stromal tumor infiltrating lymphocytes (sTILs); RNA-sequencing for 18-gene T-cell-inflamed gene expression profile (GEP), angiogenesis, and glycolysis signatures; and whole exome sequencing (paired tumor and germline) for TMB (TMB-H defined as ≥175 mut/exome), HRD-LOH (DNA damage), Signature 3, and APOBEC. Biomarkers were analyzed as continuous variables in cohorts A and B combined and individually. Area under the receiver operating characteristic curve was estimated between each biomarker and overall response rate (ORR). Wald test P-values were calculated using logistic regression adjusted for cohort and Eastern Cooperative Oncology Group performance status. Germline and somatic BRCA1/2 mutations were pooled; one-sided P-value was calculated using Fisher’s exact test. Spearman’s correlation was used for correlations. Results: Biomarker data were available in the following number of patients: 253 (99.6%; PD-L1), 204 (80.3%; CD8), 187 (73.6%; GEP), 171 (67.3%; TMB/HRD), 228 (89.8%; sTILs), 163 (64.2%; Signature 3/APOBEC), and 132 (52.0%; angiogenesis/glycolysis). When data from cohorts A and B were combined, PD-L1 CPS (median 2; IQR 0-10), CD8 (median 159; IQR 62-319), GEP (median -0.34; IQR -0.57 to -0.11), TMB (median 82 mut/exome; IQR 50-139), and sTILs (median 5; IQR 2-20) were significantly associated with ORR (Table). There were moderate correlations between PD-L1 and GEP (r = 0.532), PD-L1 and sTILs (r = 0.451), and GEP and sTILs (r = 0.490). No correlation was observed between TMB and PD-L1 (r = 0.038), GEP (r = -0.035), and sTILs (r = -0.031). When cohorts were combined, TMB was significantly associated with ORR, PFS, and OS after adjustment for PD-L1, GEP, CD8, or sTILs. HRD-LOH score, Signature 3, and APOBEC were not significantly associated with ORR (Table); P for BRCA1/2 was 0.2385. The angiogenesis signature was associated with lack of response while the glycolysis signature was associated with response to pembrolizumab (Table). Conclusions: In this exploratory biomarker analysis from KEYNOTE-086, higher levels of PD-L1, GEP, TMB, CD8 IHC, sTILs, and the glycolysis signature were associated with increased response to pembrolizumab monotherapy. These findings may help identify patients with mTNBC who are most likely to respond to pembrolizumab. Table. Association of Biomarkers as Continuous Variables With Pembrolizumab Objective ResponseBiomarkerCombined Cohorts AUCCombined Cohorts P*Combined Cohorts Multitest corrected PCohort A AUCCohort B AUCPD-L10.6740.040-0.5440.654GEP0.7480.003-0.8370.561TMB0.6270.007-0.5480.710CD8 IHC0.760.000020.000120.850.68sTILs0.6710.012-0.6320.641HRD0.3940.874-0.5220.316Signature 30.6830.072-0.6970.736APOBEC0.5280.537-0.6740.623Angiogenesis0.6770.0090.0450.6610.731Glycolysis0.6120.0090.0360.8590.459AUC, area under the curve.*One-sided P-values are shown for all biomarkers except for Signature 3 and APOBEC, for which 2-sided P-values are shown. Citation Format: Sherene Loi, Peter Schmid, Javier Cortes, David W. Cescon, Eric P. Winer, Deborah L. Toppmeyer, Hope S. Rugo, Michelino De Laurentiis, Rita Nanda, Hiroji Iwata, Ahmad Awada, Antoinette R. Tan, Roberto Salgado, Vassiliki Karantza, Petar Jelinic, Anran Wang, Lingkang Huang, Razvan Cristescu, Lakshman Annamalai, Jennifer Yearley, Jennifer Yearley, Sylvia Adams. Association between biomarkers and response to pembrolizumab in patients with metastatic triple-negative breast cancer (mTNBC): Exploratory analysis from KEYNOTE-086 [abstract]. In: Proceedings of the 2020 San Antonio Breast Cancer Virtual Symposium; 2020 Dec 8-11; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2021;81(4 Suppl):Abstract nr PD14-07.
- Published
- 2021
43. Genomic Characterization of de novo Metastatic Breast Cancer
- Author
-
Ethan Cerami, Daniel Xia, Maxwell R. Lloyd, William T. Barry, Ian E. Krop, Nan Lin, Priti Kumari, Barrett J. Rollins, Yvonne Y. Li, Simona Di Lascio, Eric P. Winer, Andrew D. Cherniack, Romualdo Barroso-Sousa, Deborah A. Dillon, Ayesha Mohammed-Abreu, Nikhil Wagle, Colin Mackichan, Janet Files, Liam F. Spurr, Laura E. MacConaill, Bruce E. Johnson, Hao Guo, Max Krevalin, Brittany L. Bychkovsky, Esha Jain, Ana C. Garrido-Castro, Melissa E. Hughes, and Neal I. Lindeman
- Subjects
0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Intrinsic resistance ,medicine.disease ,Metastatic breast cancer ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Breast cancer ,Text mining ,SETD2 ,030220 oncology & carcinogenesis ,Internal medicine ,medicine ,In patient ,Stage (cooking) ,business ,Gene - Abstract
Purpose: In contrast to recurrence after initial diagnosis of stage I–III breast cancer [recurrent metastatic breast cancer (rMBC)], de novo metastatic breast cancer (dnMBC) represents a unique setting to elucidate metastatic drivers in the absence of treatment selection. We present the genomic landscape of dnMBC and association with overall survival (OS). Experimental Design: Targeted DNA sequencing (OncoPanel) was prospectively performed on either primary or metastatic tumors from 926 patients (212 dnMBC and 714 rMBC). Single-nucleotide variants, copy-number variations, and tumor mutational burden (TMB) in treatment-naïve dnMBC primary tumors were compared with primary tumors in patients who ultimately developed rMBC, and correlated with OS across all dnMBC. Results: When comparing primary tumors by subtype, MYB amplification was enriched in triple-negative dnMBC versus rMBC (21.1% vs. 0%, P = 0.0005, q = 0.111). Mutations in KMTD2, SETD2, and PIK3CA were more prevalent, and TP53 and BRCA1 less prevalent, in primary HR+/HER2− tumors of dnMBC versus rMBC, though not significant after multiple comparison adjustment. Alterations associated with shorter OS in dnMBC included TP53 (wild-type: 79.7 months; altered: 44.2 months; P = 0.008, q = 0.107), MYC (79.7 vs. 23.3 months; P = 0.0003, q = 0.011), and cell-cycle (122.7 vs. 54.9 months; P = 0.034, q = 0.245) pathway genes. High TMB correlated with better OS in triple-negative dnMBC (P = 0.041). Conclusions: Genomic differences between treatment-naïve dnMBC and primary tumors of patients who developed rMBC may provide insight into mechanisms underlying metastatic potential and differential therapeutic sensitivity in dnMBC. Alterations associated with poor OS in dnMBC highlight the need for novel approaches to overcome potential intrinsic resistance to current treatments.
- Published
- 2021
44. Atezolizumab and nab-Paclitaxel in Advanced Triple-Negative Breast Cancer: Biomarker Evaluation of the IMpassion130 Study
- Author
-
Anh Nguyen-Duc, Marina Nechaeva, Peter Schmid, Sherene Loi, Hiroji Iwata, Carlos H. Barrios, Andreas Schneeweiss, Véronique Diéras, Luciana Molinero, Hope S. Rugo, Sylvia Adams, Leisha A. Emens, Amreen Husain, Stephen Y. Chui, and Eric P. Winer
- Subjects
Oncology ,Cancer Research ,medicine.medical_treatment ,Triple Negative Breast Neoplasms ,B7-H1 Antigen ,chemistry.chemical_compound ,0302 clinical medicine ,Monoclonal ,Antineoplastic Combined Chemotherapy Protocols ,Tumor Microenvironment ,Lymphocytes ,Humanized ,Triple-negative breast cancer ,Cancer ,0303 health sciences ,Articles ,6.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,AcademicSubjects/MED00010 ,medicine.medical_specialty ,Paclitaxel ,Clinical Trials and Supportive Activities ,Oncology and Carcinogenesis ,Antibodies, Monoclonal, Humanized ,Antibodies ,Olaparib ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,Immune system ,Breast cancer ,Clinical Research ,Atezolizumab ,Albumins ,Internal medicine ,Breast Cancer ,medicine ,Humans ,Tumor-Infiltrating ,Oncology & Carcinogenesis ,Progression-free survival ,030304 developmental biology ,Chemotherapy ,Tumor-infiltrating lymphocytes ,business.industry ,Evaluation of treatments and therapeutic interventions ,medicine.disease ,Good Health and Well Being ,chemistry ,business ,Biomarkers - Abstract
Background Understanding the impact of the tumor immune microenvironment and BRCA1/2-related DNA repair deficiencies on the clinical activity of immune checkpoint inhibitors may help optimize both patient and treatment selection in metastatic triple-negative breast cancer. In this substudy from the phase 3 IMpassion130 trial, immune biomarkers and BRCA1/2 alterations were evaluated for association with clinical benefit with atezolizumab and nab-paclitaxel (A+nP) vs placebo and nP in unresectable (P+nP) locally advanced or metastatic triple-negative breast cancer. Methods Patients were randomly assigned 1:1 to nab-paclitaxel 100 mg/m2 (days 1, 8, and 15 of a 28-day cycle) and atezolizumab 840 mg every 2 weeks or placebo until progression or toxicity. Progression-free survival and overall survival were evaluated based on programmed death-ligand 1 (PD-L1) expression on immune cells (IC) and tumor cells, intratumoral CD8, stromal tumor-infiltrating lymphocytes, and BRCA1/2 mutations. Results PD-L1 IC+ in either primary or metastatic tumor tissue was linked to progression-free survival and overall survival benefit with A+nP. PD-L1 IC+ low (26.9%; 243 of 902 patients) and high (13.9%; 125 of 902 patients) populations had improved outcomes that were comparable. Intratumoral CD8 and stromal tumor-infiltrating lymphocytes positivity (sTIL+) were associated with PD-L1 IC+ status; improved outcomes were observed with A+nP vs P+nP only in CD8+ and sTIL+ patients who were also PD-L1 IC+. BRCA1/2 mutations (occurring in 14.5% [89 of 612 patients]) were not associated with PD-L1 IC status, and PD-L1 IC+ patients benefited from A+nP regardless of BRCA1/2 mutation status. Conclusions Although A+nP was more efficacious in patients with richer tumor immune microenvironment, clinical benefit was only observed in patients whose tumors were PD-L1 IC+.
- Published
- 2021
45. Palbociclib with adjuvant endocrine therapy in early breast cancer (PALLAS): interim analysis of a multicentre, open-label, randomised, phase 3 study
- Author
-
Julie Lemieux, Michael Gnant, Amylou C. Dueck, Georg Pfeiler, Meritxell Bellet-Ezquerra, Antonio C. Wolff, Magdalena Schwarz, Marcus Vetter, Angela DeMichele, Kathy D. Miller, Kathy Puyana Theall, Miguel Gil-Gil, Hannes Fohler, Patrick G. Morris, Maria Koehler, Stacy L. Moulder, Matthew Bidwell Goetz, Manuel Ruiz-Borrego, Arlene Chan, Aleix Prat, G. Rubovszky, Silvia Antolin Novoa, Otto Metzger Filho, Miguel Martín, Debora Fumagalli, Erica L. Mayer, D. Lu, Fernando Henao, Christian Fesl, Eric P. Winer, Florian Fitzal, Alistair Ring, Tiffany A. Traina, Carter Dufrane, Harold J. Burstein, Sibylle Loibl, Nicholas Zdenkowski, Daniel Egle, Hope S. Rugo, Cynthia Huang Bartlett, Daniel G. Anderson, Eleftherios P. Mamounas, Alan P. Lyss, and Zbigniew Nowecki
- Subjects
Adult ,Oncology ,medicine.medical_specialty ,Adolescent ,Pyridines ,Receptor, ErbB-2 ,Population ,Breast Neoplasms ,Palbociclib ,Disease-Free Survival ,Piperazines ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Clinical endpoint ,Humans ,030212 general & internal medicine ,education ,Adverse effect ,Aged ,Proportional Hazards Models ,education.field_of_study ,Aromatase Inhibitors ,business.industry ,Comment ,Middle Aged ,medicine.disease ,Interim analysis ,Metastatic breast cancer ,Tamoxifen ,Regimen ,Receptors, Estrogen ,Child, Preschool ,030220 oncology & carcinogenesis ,Female ,business - Abstract
Summary Background Palbociclib added to endocrine therapy improves progression-free survival in hormone-receptor-positive, HER2-negative, metastatic breast cancer. The PALLAS trial aimed to investigate whether the addition of 2 years of palbociclib to adjuvant endocrine therapy improves invasive disease-free survival over endocrine therapy alone in patients with hormone-receptor-positive, HER2-negative, early-stage breast cancer. Methods PALLAS is an ongoing multicentre, open-label, randomised, phase 3 study that enrolled patients at 406 cancer centres in 21 countries worldwide with stage II–III histologically confirmed hormone-receptor-positive, HER2-negative breast cancer, within 12 months of initial diagnosis. Eligible patients were aged 18 years or older with an Eastern Cooperative Oncology Group performance score of 0 or 1. Patients were randomly assigned (1:1) in permuted blocks of random size (4 or 6), stratified by anatomic stage, previous chemotherapy, age, and geographical region, by use of central telephone-based and web-based interactive response technology, to receive either 2 years of palbociclib (125 mg orally once daily on days 1–21 of a 28-day cycle) with ongoing standard provider or patient-choice adjuvant endocrine therapy (tamoxifen or aromatase inhibitor, with or without concurrent luteinising hormone-releasing hormone agonist), or endocrine therapy alone, without masking. The primary endpoint of the study was invasive disease-free survival in the intention-to-treat population. Safety was assessed in all randomly assigned patients who started palbociclib or endocrine therapy. This report presents results from the second pre-planned interim analysis triggered on Jan 9, 2020, when 67% of the total number of expected invasive disease-free survival events had been observed. The trial is registered with ClinicalTrials.gov (NCT02513394) and EudraCT (2014-005181-30). Findings Between Sept 1, 2015, and Nov 30, 2018, 5760 patients were randomly assigned to receive palbociclib plus endocrine therapy (n=2883) or endocrine therapy alone (n=2877). At the time of the planned second interim analysis, at a median follow-up of 23·7 months (IQR 16·9–29·2), 170 of 2883 patients assigned to palbociclib plus endocrine therapy and 181 of 2877 assigned to endocrine therapy alone had invasive disease-free survival events. 3-year invasive disease-free survival was 88·2% (95% CI 85·2–90·6) for palbociclib plus endocrine therapy and 88·5% (85·8–90·7) for endocrine therapy alone (hazard ratio 0·93 [95% CI 0·76–1·15]; log-rank p=0·51). As the test statistic comparing invasive disease-free survival between groups crossed the prespecified futility boundary, the independent data monitoring committee recommended discontinuation of palbociclib in patients still receiving palbociclib and endocrine therapy. The most common grade 3–4 adverse events were neutropenia (1742 [61·3%] of 2840 patients on palbociclib and endocrine therapy vs 11 [0·3%] of 2903 on endocrine therapy alone), leucopenia (857 [30·2%] vs three [0·1%]), and fatigue (60 [2·1%] vs ten [0·3%]). Serious adverse events occurred in 351 (12·4%) of 2840 patients on palbociclib plus endocrine therapy versus 220 (7·6%) of 2903 patients on endocrine therapy alone. There were no treatment-related deaths. Interpretation At the planned second interim analysis, addition of 2 years of adjuvant palbociclib to adjuvant endocrine therapy did not improve invasive disease-free survival compared with adjuvant endocrine therapy alone. On the basis of these findings, this regimen cannot be recommended in the adjuvant setting. Long-term follow-up of the PALLAS population and correlative studies are ongoing. Funding Pfizer.
- Published
- 2021
46. Survival, Pathologic Response, and Genomics in CALGB 40601 (Alliance), a Neoadjuvant Phase III Trial of Paclitaxel-Trastuzumab With or Without Lapatinib in HER2-Positive Breast Cancer
- Author
-
Lucas J. Huebner, Mei Yin C. Polley, Eric P. Winer, Chau T. Dang, Ann H. Partridge, Lisa A. Carey, Donald A. Berry, Clifford A. Hudis, Charles M. Perou, Jonathan Shepherd, Katherine A. Hoadley, Ian E. Krop, David W. Hillman, N. Lynn Henry, Joel S. Parker, Aranzazu Fernandez-Martinez, Lyndsay Harris, Olwen Hahn, and Sara M. Tolaney
- Subjects
Adult ,0301 basic medicine ,Oncology ,Cancer Research ,medicine.medical_specialty ,Paclitaxel/trastuzumab ,Paclitaxel ,Receptor, ErbB-2 ,medicine.medical_treatment ,Breast Neoplasms ,Lapatinib ,Young Adult ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,HER2 Positive Breast Cancer ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Humans ,Pathologic Response ,skin and connective tissue diseases ,Human Epidermal Growth Factor Receptor 2 ,Complete response ,Aged ,Neoplasm Staging ,Aged, 80 and over ,Chemotherapy ,business.industry ,ORIGINAL REPORTS ,Middle Aged ,Trastuzumab ,Survival Analysis ,Neoadjuvant Therapy ,030104 developmental biology ,030220 oncology & carcinogenesis ,Female ,Transcriptome ,business ,medicine.drug - Abstract
PURPOSE CALGB 40601 assessed whether dual versus single human epidermal growth factor receptor 2 (HER2) –targeting drugs added to neoadjuvant chemotherapy increased pathologic complete response (pCR). Here, we report relapse-free survival (RFS), overall survival (OS), and gene expression signatures that predict pCR and survival. PATIENTS AND METHODS Three hundred five women with untreated stage II and III HER2-positive breast cancer were randomly assigned to receive weekly paclitaxel combined with trastuzumab plus lapatinib (THL), trastuzumab (TH), or lapatinib (TL). The primary end point was pCR, and secondary end points included RFS, OS, and gene expression analyses. mRNA sequencing was performed on 264 pretreatment samples. RESULTS One hundred eighteen patients were randomly allocated to THL, 120 to TH, and 67 to TL. At more than 7 years of follow-up, THL had significantly better RFS and OS than did TH (RFS hazard ratio, 0.32; 95% CI, 0.14 to 0.71; P = .005; OS hazard ratio, 0.34; 95% CI, 0.12 to 0.94; P = .037), with no difference between TH and TL. Of 688 previously described gene expression signatures, significant associations were found in 215 with pCR, 45 with RFS, and only 22 with both pCR and RFS (3.2%). Specifically, eight immune signatures were significantly correlated with a higher pCR rate and better RFS. Among patients with residual disease, the immunoglobulin G signature was an independent, good prognostic factor, whereas the HER2-enriched signature, which was associated with a higher pCR rate, showed a significantly shorter RFS. CONCLUSION In CALGB 40601, dual HER2-targeting resulted in significant RFS and OS benefits. Integration of intrinsic subtype and immune signatures allowed for the prediction of pCR and RFS, both overall and within the residual disease group. These approaches may provide means for rational escalation and de-escalation treatment strategies in HER2-positive breast cancer.
- Published
- 2020
47. A Randomized Placebo Controlled Phase II Trial Evaluating Exemestane with or without Enzalutamide in Patients with Hormone Receptor–Positive Breast Cancer
- Author
-
Frankie A. Holmes, Laura Biganzoli, Catherine M. Kelly, Zhou Zhu, Maureen E. Trudeau, Patrick G. Morris, Claudio Zamagni, Marco Colleoni, Lorenzo Sica, Denise A. Yardley, Thomas O’Brien, Ayca Gucalp, Laura García-Estévez, Ahmad Awada, Lowell L. Hart, Denka Markova, Eric P. Winer, Lee S. Schwartzberg, Joyce Steinberg, Jamal Tarazi, Vandana G. Abramson, Tiffany A. Traina, Ian E. Krop, and Stephen Chan
- Subjects
Adult ,Oncology ,Cancer Research ,medicine.medical_specialty ,Receptor, ErbB-2 ,Population ,Breast Neoplasms ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Breast cancer ,Double-Blind Method ,Exemestane ,Internal medicine ,Antineoplastic Combined Chemotherapy Protocols ,Nitriles ,Phenylthiohydantoin ,medicine ,Clinical endpoint ,Humans ,Enzalutamide ,030212 general & internal medicine ,education ,Aged ,Aged, 80 and over ,education.field_of_study ,business.industry ,Hazard ratio ,Middle Aged ,Prognosis ,medicine.disease ,Metastatic breast cancer ,Androstadienes ,Survival Rate ,Receptors, Estrogen ,chemistry ,030220 oncology & carcinogenesis ,Benzamides ,Cohort ,Female ,Receptors, Progesterone ,business ,Follow-Up Studies - Abstract
Purpose: To determine whether the androgen receptor (AR) inhibitor, enzalutamide, improves effectiveness of endocrine therapy (ET) in hormone receptor–positive (HR+) breast cancer. Patients and Methods: In this phase II trial, patients with HR+/HER2 normal advanced/metastatic breast cancer were randomized 1:1 to exemestane 25 mg with placebo or exemestane 50 mg with enzalutamide 160 mg daily (NCT02007512). Two parallel cohorts enrolled patients with 0 (cohort 1) or 1 (cohort 2) prior ET for advanced disease. Progression-free survival (PFS) was the primary endpoint in the intent-to-treat (ITT) population of each cohort. Biomarkers were evaluated in an exploratory analysis. Results: Overall, 247 patients were randomized (cohort 1, n = 127 and cohort 2, n = 120). PFS was not improved in either cohort of the ITT population [HR, 0.82 (95% confidence interval (CI), 0.54–1.26); P = 0.3631 for cohort 1 and HR, 1.02 (95% CI, 0.66–1.59); P = 0.9212 for cohort 2]. In cohort 1, high levels of AR mRNA were associated with greater benefit of enzalutamide (Pinteraction = 0.0048). This effect was particularly apparent in patients with both high levels of AR mRNA and low levels of ESR1 mRNA [HR, 0.24 (95% CI, 0.10–0.60); P = 0.0011]. The most common any grade adverse events in the enzalutamide arms were nausea (39%) in cohort 1 and fatigue (37%) in cohort 2. Conclusions: Enzalutamide with exemestane was well tolerated. While PFS was not improved by the addition of enzalutamide to exemestane in an unselected population, ET-naïve patients with high AR mRNA levels, particularly in combination with low ESR1 mRNA levels, may benefit from enzalutamide with exemestane.
- Published
- 2020
48. Road Map to Safe and Well-Designed De-escalation Trials of Systemic Adjuvant Therapy for Solid Tumors
- Author
-
Florentine Hilbers, Patricia A. Spears, Elizabeth S. Frank, Eva Schumacher, Martine Piccart, Meredith M. Regan, Carmela Caballero, Judith M Bliss, Larry Norton, Nancy E. Davidson, Rachida Naït Kaoudjt, Eric P. Winer, Patrick Renault, and Richard D. Gelber
- Subjects
Cancer Research ,medicine.medical_specialty ,Accrual ,business.industry ,MEDLINE ,Cancer ,Antineoplastic Agents ,medicine.disease ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Oncology ,Chemotherapy, Adjuvant ,Neoplasms ,030220 oncology & carcinogenesis ,medicine ,Adjuvant therapy ,Humans ,030212 general & internal medicine ,Road map ,Intensive care medicine ,Adverse effect ,business ,De-escalation ,Randomized Controlled Trials as Topic - Abstract
An important challenge in the field of cancer is finding the balance between delivering effective treatments and avoiding adverse effects and financial toxicity caused by innovative, yet expensive, drugs. To address this, several treatment de-escalation trials have been conducted, but only a few of these have provided clear answers. A few trials had poor accrual or had design flaws that led to conflicting results. Members of the Breast International Group (BIG) and North American Breast Cancer Group (NABCG) believe the way forward is to understand the lessons from these trials and listen more carefully to what truly matters to our patients. We reviewed several adjuvant trials of different cancer types and developed a road map for improving the design and implementation of future de-escalation trials. The road map incorporates patients’ insights obtained through focused group discussions across the BIG-NABCG networks. Considerations for the development of de-escalation trials for systemic adjuvant treatment, including noninferiority trial design, choice of end points, and prioritization of a patient’s perspectives, are presented in this consensus article.
- Published
- 2020
49. A prospective trial of treatment de-escalation following neoadjuvant paclitaxel/trastuzumab/pertuzumab in HER2-positive breast cancer
- Author
-
Adrienne G. Waks, Neelam V. Desai, Tianyu Li, Philip D. Poorvu, Ann H. Partridge, Natalie Sinclair, Laura M. Spring, Meredith Faggen, Michael Constantine, Otto Metzger, Jillian Alberti, Julia Deane, Shoshana M. Rosenberg, Elizabeth Frank, Sara M. Tolaney, Ian E. Krop, Nadine M. Tung, Nabihah Tayob, Tari A. King, Elizabeth A. Mittendorf, and Eric P. Winer
- Subjects
Oncology ,Pharmacology (medical) ,Radiology, Nuclear Medicine and imaging - Abstract
De-escalating adjuvant therapy following pathologic complete response (pCR) to an abbreviated neoadjuvant regimen in human epidermal growth factor receptor 2-positive (HER2+) breast cancer is the focus of international research efforts. However, the feasibility of this approach and its appeal to patients and providers had not been formally investigated. We aimed to assess adherence to de-escalated adjuvant antibody doublet therapy (trastuzumab and pertuzumab [HP], without chemotherapy) among patients with pCR following neoadjuvant paclitaxel/HP (THP). In this single-arm prospective trial, patients with treatment-naïve stage II-III HER2+ breast cancer received neoadjuvant weekly paclitaxel ×12 and HP every 3 weeks ×4. The primary endpoint was receipt of adjuvant non-HER2-directed cytotoxic chemotherapy. Ninety-eight patients received ≥1 dose of THP on study. Patients had median age of 50 years, 86% had stage II tumors, and 34% were hormone receptor-negative. Five patients had incomplete clinical response following THP and received doxorubicin and cyclophosphamide before surgery; they were classified as non-pCR and censored from further analyses. The overall pCR rate was 56.7%. Among patients with pCR, the adherence rate to de-escalated antibody-only therapy (HP) was 98.2% (95% CI 90.3–100.0%), and the primary feasibility endpoint was reached. The majority of patients felt positive or neutral about their adjuvant treatment plans. With brief follow-up (median 19.1 months), there were no breast cancer recurrences. De-escalation of adjuvant chemotherapy among patients who experience pCR in early-stage HER2+ breast cancer is a practicable approach for both patients and physicians. Planned and ongoing prospective trials will determine the long-term efficacy of this approach.Trial registration clinicaltrials.gov, NCT03716180, https://clinicaltrials.gov/ct2/show/NCT03716180.
- Published
- 2022
50. Estimating long-term mortality in women with hormone receptor-positive breast cancer: The 'ESTIMATE' tool
- Author
-
José P. Leone, Noah Graham, Sara M. Tolaney, Bernardo A. Leone, Rachel A. Freedman, Michael J. Hassett, Julieta Leone, Carlos T. Vallejo, Eric P. Winer, Nancy U. Lin, and Nabihah Tayob
- Subjects
Adult ,Cancer Research ,Oncology ,Humans ,Breast Neoplasms ,Female ,Breast ,Middle Aged ,Prognosis ,Neoplasm Staging ,SEER Program - Abstract
The risk of breast cancer-specific mortality (BCSM) persists for at least 20 years from diagnosis. Estimating the risk of BCSM over this extended period along with competing risks of death would aid clinical decision-making. We aimed to develop an interactive tool called 'ESTIMATE', to explore the Surveillance, Epidemiology, and End Results (SEER) registry to quantify residual risks of BCSM, non-BCSM and all-cause mortality in non-metastatic, hormone receptor (HR)-positive breast cancer patient subgroups at any given time after diagnosis, up to 20 years.Using SEER data, we included 264,237 women with invasive, non-metastatic, HR-positive breast cancer diagnosed from 1990 to 2006. We developed a tool that provides a nonparametric estimate of the residual cumulative risk of BCSM and non-BCSM by year 20 after any specified time from initial diagnosis, among patients defined by baseline clinical and pathologic variables, using Gray's subdistribution method.ESTIMATE allows the user to input patient and tumour characteristics and the preferred timeframe. For example, patients in the age group of 40-49 diagnosed with T1cN1, grade II breast cancer who survived 7 years, have a 14% (95% confidence interval [CI]: 11.9%-16.1%) residual cumulative risk of BCSM in the next 13 years, and a 6.4% (95% CI: 4.7%-8.1%) residual cumulative risk of non-BCSM over the same period.ESTIMATE provides population-based risks of BCSM, non-BCSM and all-cause mortality through 20 years after diagnosis of HR-positive breast cancer, based on patient and tumour characteristics. ESTIMATE can inform discussions about prognosis, a balance between competing risks and aid clinical decision-making.
- Published
- 2022
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.