680 results on '"L Pusztai"'
Search Results
102. Expression of sigma 1 receptor in human breast cancer.
- Author
-
B. Wang, R. Rouzier, C. T. Albarracin, A. Sahin, P. Wagner, Y. Yang, T. L. Smith, F. Meric-Bernstam, C. Marcelo. Aldaz, G. N. Hortobagyi, and L. Pusztai
- Published
- 2005
- Full Text
- View/download PDF
103. Hormone receptor status and pathologic response of HER2-positive breast cancer treated with neoadjuvant chemotherapy and trastuzumab.
- Author
-
F. Peintinger, A. U. Buzdar, H. M. Kuerer, J. A. Mejia, C. Hatzis, A. M. Gonzalez-Angulo, L. Pusztai, F. J. Esteva, S. S. Dawood, M. C. Green, G. N. Hortobagyi, and W. F. Symmans
- Subjects
- *
BREAST cancer treatment , *HORMONE receptors , *ADJUVANT treatment of cancer , *CANCER chemotherapy , *TRASTUZUMAB , *COHORT analysis - Abstract
Background: The aim of this study was to compare the extent of pathologic response in patients with HER2-positive (HER2+) breast cancer treated with standard neoadjuvant chemotherapy, with or without trastuzumab (H), according to hormone receptor (HR) status. Patients and methods: We included 199 patients with HER2+ breast cancer from three successive cohorts of neo-adjuvant chemotherapy on the basis of paclitaxel (Taxol) (P) administered weekly (w) or three weekly (3-w), followed by 5-fluorouracil (F), doxorubicin (A) or epirubicin (E), and cyclophosphamide (C). Residual cancer burden (RCB) was determined from pathologic review of the primary tumor and lymph nodes and was classified as pathologic complete response (pCR) or minimal (RCB-I), moderate (RCB-II), or extensive (RCB-III) residual disease. Results: In HR-positive (HR+) cancers, a higher rate of pathologic response (pCR/RCB-I) was observed with concurrent H + 3-wP/FEC (73%) than with 3-wP/FEC (34%, P = 0.002) or wP/FAC (47%; P = 0.02) chemotherapy alone. In HR-negative (HR−) cancers, there were no significant differences in the rate of pathologic response (pCR/RCB-I) from 3-wP/FAC (50%), wP/FAC (68%), or concurrent H + 3-wP/FEC (72%). Conclusions: Patients with HR+/HER2+ breast cancer obtained significant benefit from addition of trastuzumab to P/FEC chemotherapy; pathologic response rate was similar to that seen in HR−/HER2+ breast cancers. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
104. Imatinib mesylate (Gleevec(R)) in advanced breast cancer-expressing C-Kit or PDGFR-{beta}: clinical activity and biological correlations.
- Author
-
M. Cristofanilli, P. Morandi, S. Krishnamurthy, J. M. Reuben, B.-N. Lee, D. Francis, D. J. Booser, M. C. Green, B. K. Arun, L. Pusztai, A. Lopez, R. Islam, V. Valero, and G. N. Hortobagyi
- Subjects
- *
BREAST cancer treatment , *ANTINEOPLASTIC agents , *METHANESULFONATES , *IMATINIB , *PROTEIN-tyrosine kinases , *CANCER cells , *IMMUNOREGULATION , *MEDICAL research - Abstract
Background: Novel molecular therapies for metastatic breast cancer (MBC) are necessary to improve the dismal prognosis of this condition. Imatinib mesylate (Gleevec®) inhibits several protein tyrosine kinases, including platelet-derived growth factor receptor (PDGFR) and c-kit, which are preferentially expressed in tumor cells. We tested the activity of imatinib mesylate in MBC with overexpression of PDGFR or c-kit. Additionally, we sought to determine the biological correlates and immunomodulatory effects. Patients and methods: Thirteen patients were treated with Imatinib administered orally at 400 mg p.o. b.i.d. (800 mg/day), until disease progression. All patients demonstrated PDGFR-β overexpression and none showed c-kit expression. Results: No objective responses were observed among the 13 patients treated in an intention-to-treat analysis. All patients experienced disease progression, with a median time to progression of 1.2 months. Twelve patients have died, and the median overall survival was 7.7 months. No patient had a serious adverse event. Imatinib therapy had no effect on the plasma levels of the angiogenesis-related cytokines, vascular endothelial growth factor, PDGF, b-fibroblast growth factor, and E-selectin. Immune studies showed imatinib inhibits interferon-γ production by TCR-activated CD4+ T cells. Conclusion: Imatinib as a single agent has no clinical activity in PDGFR-overexpressing MBC and has potential immunosuppressive effects. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
105. Expression patterns and predictive value of phosphorylated AKT in early-stage breast cancer.
- Author
-
F. Andre, R. Nahta, R. Conforti, T. Boulet, M. Aziz, L. X. H. Yuan, F. Meslin, M. Spielmann, G. Tomasic, L. Pusztai, G. N. Hortobagyi, S. Michiels, S. Delaloge, and F. J. Esteva
- Subjects
- *
BREAST cancer , *PHOSPHORYLATION , *IMMUNOHISTOCHEMISTRY , *TUMORS , *ADJUVANT treatment of cancer , *CANCER chemotherapy - Abstract
Background: AKT phosphorylation is a critical step in the activation of growth factor receptors and can mediate tumor resistance to anthracyclines. We evaluated the expression patterns and predictive value of phosphorylated AKT (pAKT) in breast cancer tissues. Patients and methods: pAKT expression was assessed by immunohistochemistry in 823 tumors from patients with early breast cancer enrolled in two randomized trials. The distribution of pAKT expression was correlated with HER2 and epidermal growth factor receptor (EGFR) expression. The predictive value of pAKT for the efficacy of adjuvant chemotherapy was determined by test for interaction. Results: pAKT, EGFR, and HER2 were expressed in 119 of 781 (15%), 118 of 758 (16%), and 99 of 775 (13%) assessable tumors. Staining was positive for pAKT in 28 of 99 (28%) and 90 of 676 (13%) HER2+ and HER2− tumors (P P = 0.49). A positive staining for pAKT did not correlate with prognosis (P = 0.94), and did not predict the resistance to anthracyclines (test for interaction, P = 0.70). Conclusions: AKT phosphorylation is associated with HER2 expression but not EGFR expression in patients with early breast cancer. pAKT is not predictive for the efficacy of anthracycline-based adjuvant chemotherapy. [ABSTRACT FROM AUTHOR]
- Published
- 2008
- Full Text
- View/download PDF
106. Inclusion of taxanes, particularly weekly paclitaxel, in preoperative chemotherapy improves pathologic complete response rate in estrogen receptor-positive breast cancers.
- Author
-
C Mazouni, S-W Kau, D Frye, F Andre, HM Kuerer, TA Buchholz, WF Symmans, K Anderson, KR Hess, AM Gonzalez-Angulo, GN Hortobagyi, AU Buzdar, and L Pusztai
- Subjects
- *
DRUG therapy , *BREAST cancer , *PACLITAXEL , *FLUOROURACIL , *DOXORUBICIN , *ADJUVANT treatment of cancer , *TUMORS - Abstract
Background: We examined if inclusion of a taxane and more prolonged preoperative chemotherapy improves pathologic complete response (pCR) rate in estrogen receptor (ER)-positive breast cancer compared with three to four courses of 5-fluorouracil, doxorubicin, cyclophosphamide (FAC). Patients and methods: Pooled analysis of results from seven consecutive neo-adjuvant chemotherapy trials including 1079 patients was carried out. These studies were conducted at MD Anderson Cancer Center from 1974 to 2001. Four hundred and twenty-six (39.5%) patients received taxane-based neo-adjuvant therapy. pCR rates and survival times were analyzed as a function of chemotherapy regimen and ER status. Multivariate logistic and Cox regression analysis were carried out to identify variables associated with pCR and survival. Results: Patients with ER-negative cancer had higher overall pCR rate than patients with ER-positive tumors (20.1% versus 4.9%, P < 0.001). In ER-negative patients, the pCR rates were 29% and 15% with and without a taxane (P < 0.001). In ER-positive patients, the pCR rates were 8.8% and 2.0% with and without a taxane (P < 0.001). In multivariate analysis, clinical tumor size (P < 0.001), ER-negative status (P < 0.001) and inclusion of a taxane (P = 0.01) were independently associated with pCR. For patients with pCR, survival was similar regardless of ER status or the type of regimen that induced pCR. Conclusion: pCR rates increased for patients with both ER-positive and ER-negative tumors as regimens started to include a taxane and became longer. This indicates that a subset of patients with ER-positive breast cancer benefits from more aggressive chemotherapy, similarly to patients with ER-negative tumors. [ABSTRACT FROM AUTHOR]
- Published
- 2007
- Full Text
- View/download PDF
107. Pathologic complete response (pCR) rates for patients with HR+/HER2- high-risk, early-stage breast cancer (EBC) by clinical and molecular features in the phase II I-SPY2 clinical trial.
- Author
-
Huppert LA, Wolf D, Yau C, Brown-Swigart L, Hirst GL, Isaacs C, Pusztai L, Pohlmann PR, DeMichele A, Shatsky R, Yee D, Thomas A, Nanda R, Perlmutter J, Heditsian D, Hylton N, Symmans F, Van't Veer LJ, Esserman L, and Rugo HS
- Subjects
- Humans, Female, Middle Aged, Adult, Aged, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Disease-Free Survival, Pathologic Complete Response, Breast Neoplasms pathology, Breast Neoplasms genetics, Breast Neoplasms metabolism, Breast Neoplasms drug therapy, Breast Neoplasms therapy, Receptor, ErbB-2 metabolism, Receptor, ErbB-2 genetics, Receptors, Estrogen metabolism, Receptors, Progesterone metabolism, Neoadjuvant Therapy methods, Neoplasm Staging, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism
- Abstract
Background: Hormone receptor-positive (HR+), human epidermal growth factor receptor 2 (HER2)-negative early-stage breast cancer (EBC) is a heterogenous disease. Identification of better clinical and molecular biomarkers is essential to guide optimal therapy for each patient., Patients and Methods: We analyzed rates of pathologic complete response (pCR) and distant recurrence-free survival (DRFS) for patients with HR+/HER2-negative EBC in eight neoadjuvant arms in the I-SPY2 trial by clinical/molecular features: age, stage, histology, percentage estrogen receptor (ER) positivity, ER/progesterone receptor status, MammaPrint (MP)-High1 (0 to -0.57) versus MP-High2 (<-0.57), BluePrint (BP)-Luminal-type versus BP-Basal-type, and ImPrint immune signature. We quantified the clinical/molecular heterogeneity, assessed overlap among these biomarkers, and evaluated associations with pCR and DRFS., Results: Three hundred and seventy-nine patients with HR+/HER2-negative EBC were included in this analysis, with an observed pCR rate of 17% across treatment arms. pCR rates were higher in patients with stage II versus III disease (21% versus 9%, P = 0.0013), ductal versus lobular histology (19% versus 11%, P = 0.049), lower %ER positivity (≤66% versus >66%) (35% versus 9%, P = 3.4E-09), MP-High2 versus MP-High1 disease (31% versus 11%, P = 1.1E-05), BP-Basal-type versus BP-Luminal-type disease (34% versus 10%, P = 1.62E-07), and ImPrint-positive versus -negative disease (38% versus 10%, P = 1.64E-09). Patients with lower %ER were more likely to have MP-High2 and BP-Basal-type disease. At a median follow-up of 4.8 years, patients who achieved pCR had excellent outcomes irrespective of clinical/molecular features. Among patients who did not achieve pCR, DRFS events were more frequent in patients with MP-High2 and BP-Basal-type disease than those with MP-High1 and BP-Luminal-type disease., Conclusions: Among patients with high molecular-risk HR+/HER2-negative EBC, the MP-High2, BP-Basal-type, and ImPrint-positive signatures identified a partially overlapping subset of patients who were more likely to achieve pCR in response to neoadjuvant chemotherapy ± targeted agents or immunotherapy compared to patients with MP-High1, BP-Luminal-type, and ImPrint-negative disease. I-SPY2.2 is incorporating the use of these biomarkers to molecularly define specific patient populations and optimize treatment selection., (Copyright © 2024 European Society for Medical Oncology. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
108. Neoadjuvant nivolumab and chemotherapy in early estrogen receptor-positive breast cancer: a randomized phase 3 trial.
- Author
-
Loi S, Salgado R, Curigliano G, Romero Díaz RI, Delaloge S, Rojas García CI, Kok M, Saura C, Harbeck N, Mittendorf EA, Yardley DA, Suárez Zaizar A, Caminos FR, Ungureanu A, Reinoso-Toledo JG, Guarneri V, Egle D, Ades F, Pacius M, Chhibber A, Chandra R, Nathani R, Spires T, Wu JQ, Pusztai L, and McArthur H
- Abstract
Patients with estrogen receptor-positive (ER+), human epidermal growth factor receptor 2-negative (HER2-) primary breast cancer (BC) have low pathological complete response (pCR) rates with neoadjuvant chemotherapy. A subset of ER+/HER2- BC contains dense lymphocytic infiltration. We hypothesized that addition of an anti-programmed death 1 agent may increase pCR rates in this BC subtype. We conducted a randomized, multicenter, double-blind phase 3 trial to investigate the benefit of adding nivolumab to neoadjuvant chemotherapy in patients with newly diagnosed, high-risk, grade 3 or 2 (ER 1 to ≤10%) ER+/HER2- primary BC. In total, 510 patients were randomized to receive anthracycline and taxane-based chemotherapy with either intravenous nivolumab or placebo. The primary endpoint of pCR was significantly higher in the nivolumab arm compared with placebo (24.5% versus 13.8%; P = 0.0021), with greater benefit observed in patients with programmed death ligand 1-positive tumors (VENTANA SP142 ≥1%: 44.3% versus 20.2% respectively). There were no new safety signals identified. Of the five deaths that occurred in the nivolumab arm, two were related to study drug toxicity; no deaths occurred in the placebo arm. Adding nivolumab to neoadjuvant chemotherapy significantly increased pCR rates in high-risk, early-stage ER+/HER2- BC, particularly among patients with higher stromal tumor-infiltrating lymphocyte levels or programmed death ligand 1 expression, suggesting a new treatment paradigm that emphasizes the role of immunotherapy and T cell immunosurveillance in luminal disease. Clinical trials.gov identifier: NCT04109066., Competing Interests: Competing interests: S.L. reports institutional research funding from AstraZeneca/Daiichi Sankyo, Bristol Myers Squibb, Novartis, Puma Biotechnology, Roche-Genentech and Seattle Genetics; consulting fees from Amaroq Therapeutics, Astra Zeneca/Daiichi Sankyo, Bristol Myers Squibb, Domain Therapeutics Gilead, Lilly, Mersana Therapeutics, MSD, Roche-Genentech and Novartis; honoraria from Amaroq Therapeutics, AstraZeneca/Daiichi Sankyo, BioNTech, Bristol Myers Squibb, Domain Therapeutics, Gilead Sciences, Lilly, Mersana Therapeutics, MSD, Novartis and Roche-Genentech; meeting/ travel support from Bristol Myers Squibb and Lilly. R.S. reports consulting fees from Owkin; honoraria from AstraZeneca, Daiichi Sankyo and Exact Sciences; meeting/travel support from AstraZeneca and Daichii Sankyo; leadership position at The International Immuno-Oncology Biomarker Working Group; research support from Bristol Myers Squibb and providing unpaid advice to Case 45. G.C. reports participation on advisory boards for AstraZeneca, Bristol Myers Squibb, Daichii Sankyo, Celcuity, Ellipsis, Exact Science, Gilead, Lilly, Menarini, Merck, Novartis, Pfizer, Roche, Sanofi and Veracyte. S.D. reports institutional research funding from Amgen, AstraZeneca, Bristol Myers Squibb, European Commission, the French government, Lilly, Novartis, Orion, Pierre Fabre, Pfizer, Roche-Genentech, Sanofi and Taiho; meeting/travel support from Novartis, Roche and Seagen; participation on advisory boards for AstraZeneca, Besins, Decibio, Elsan, Gilead and Sanofi; participation on independent data monitoring committee activity for Breast International Group and Pfizer. C.I.R.G. reports honoraria from AstraZeneca, Bristol Myers Squibb, Knight, MSD, Pfizer, Roche and Tecnofarma; participation on data safety monitoring/advisory boards for Bristol Myers Squibb, Pfizer and Sanofi. M.K. reports institutional research funding from AstraZeneca, Bristol Myers Squibb and Roche; honoraria from Bristol Myers Squibb and Gilead; participation on advisory boards for AstraZeneca, BioNTech, Bristol Myers Squibb, MSD and Roche. C.S. reports research funding from AstraZeneca, Boehringer Ingelheim, Bristol Myers Squibb, Byondis BV, Daiichi Sankyo, Eisai, Genentech, Gilead, Glaxo, Menarini, Merus, MSD, Novartis, Pfizer, Philips Healthcare, Piere Fabre, Puma Biotechnology, Roche, Sanofi Aventis, Seagen, Synthon and Zymeworks; consulting fees from AstraZeneca, Daiichi Sankyo, Eisai, Gilead, Lilly, MediTech, Novartis, Pfizer, Philips Healthcare, Pharmalex, Piere Fabre, Puma Biotechnology, Roche, Seagen, Synthon and Zymeworks; honoraria from AstraZeneca, Daiichi Sankyo, Exeter Pharmaceuticals, Lilly, Pfizer, Pierre Fabre, Puma Biotechnology and Seagen; payment for expert testimony from AX’s Consulting SARL, Boehringer Ingelheim, Bristol Myers Squibb, Genentech, Innoup, MSD España, Novartis and Sanofi; meeting/travel support from AstraZeneca, Daiichi Sankyo, Eisai Europe, Gilead, Lilly, Novartis, Pfizer, Pierre Fabre, Puma Biotechnology, Roche and Seagen; participation on data safety monitoring/advisory boards for AstraZeneca, Daiichi Sankyo, Eisai Europe, Gilead, Lilly, Menarini, MSD, Novartis, Pfizer, Philips, Pierre Fabre, Roche and Seagen. N.H. reports consulting fees from Daiichi Sankyo, Gilead, Novartis, Pfizer, Roche, Seagen and Viatris; honoraria from AstraZeneca, Daiichi Sankyo, Gilead, Lilly, MSD, Novartis, Pierre Fabre, Pfizer, Roche, Seagen, Viatris and Zuelligpharma; participation on data safety monitoring/advisory boards for Gilead, Roche and Seagen; leadership position at the West German Study Group. E.A.M. reports research funding from Genentech, Gilead and Roche; consulting fees from AstraZeneca, BioNTech, Moderna and MSD; honoraria from MSD; meeting/travel support from MSD; leadership positions at the American Society of Clinical Oncology Board of Directors 2019–2023 and Scientific Advisor for Susan G. Komen. D.A.Y. reports institutional research funding from AbbVie, Ambrx, AstraZeneca, Dana Farber Cancer Institute, Lilly, Roche-Genentech, G1 Therapeutics, Gilead, Incyte, Innocrin Pharmaceuticals, Merck, Novartis, Polyphor, Stemline Therapeutics, US Oncology and UT Southwestern; and consulting fees paid to institution from AstraZeneca, Daiichi Sankyo, Gilead, Integra Connect, Novartis and Stemline Therapeutics. V.G. reports honoraria from AstraZeneca, Daiichi Sankyo, Exact Sciences, Gilead, GlaxoSmithKline, Lilly, Menarini Stemline, Novartis, Roche, and Zentiva; payment for expert testimony from Lilly; meeting/travel support from AstraZeneca and Gilead; institutional patent for Her2DX; and participation on data safety monitoring/advisory boards for AstraZeneca, Daiichi Sankyo, Exact Sciences, Gilead, Lilly, MSD, Novartis, Olema Oncology, Pierre Fabre, Pfizer-Seagen and Roche. D.E. reports consulting fees from AstraZeneca, Daiichi Sankyo, Gilead, Lilly, Menarini, MSD, Novartis, Pfizer, Roche, Seagen and Sirius Medical; honoraria from AstraZeneca, Daiichi Sankyo, Gilead, Lilly, Menarini, MSD, Novartis, Pfizer, Roche, Seagen and Sirius Medical; meeting/travel support from Daiichi Sankyo and Pfizer; and receipt of medical writing support from Sirius Medical. F.A. reports being an employee of Bristol Myers Squibb. M.P. reports being an employee and shareholder of Bristol Myers Squibb. A.C. reports being an employee of Bristol Myers Squibb and shareholder of Merck. R.C. reports being an employee of Bristol Myers Squibb. R.N. reports being an employee and shareholder of Bristol Myers Squibb. T.S. reports being an employee and shareholder of Bristol Myers Squibb. J.Q.W. reports being an employee and shareholder of Bristol Myers Squibb. L.P. reports institutional research funding from AstraZeneca, Bristol Myers Squibb, GlaxoSmithKline, Merck, Pfizer and Seagen; consulting fees from AstraZeneca, Bristol Myers Squibb, Daiichi, Exact Sciences, Genentech-Roche, GlaxoSmithKline, Merck, Natera, Novartis, Personalis, Pfizer and Stemline-Menarini; and honoraria for advisory board participation from AstraZeneca, Bristol Myers Squibb, Daiichi, Exact Sciences, Genentech-Roche, GlaxoSmithKline, Merck, Natera, Novartis, Personalis, Pfizer and Stemline-Menarini. H.M. reports consulting fees from Amgen, AstraZeneca, Bristol Myers Squibb, Calithera, Celgene, Crown Bioscience Daiichi Sankyo, Lilly, Genentech-Roche, Gilead, Immunomedics, Merck, OBI Pharma, Peregrine, Pfizer, Puma, Spectrum Pharmaceuticals, Syndax Pharmaceuticals, Seattle Genetics and TapImmune; and research funding from Bristol Myers Squibb, BTG, LLC/AstraZeneca, MedImmune and Merck. R.I.R.D., A.S.Z., F.R.C., A.U. and J.G.R.-T. declare no competing interests., (© 2025. Crown.)
- Published
- 2025
- Full Text
- View/download PDF
109. Hormone Receptor-Positive HER2-Negative/MammaPrint High-2 Breast Cancers Closely Resemble Triple-Negative Breast Cancers.
- Author
-
Rios-Hoyo A, Xiong K, Dai J, Yau C, Marczyk M, Garcia-Milian R, Wolf DM, Huppert LA, Nanda R, Hirst GL, Cobain EF, van 't Veer LJ, Esserman LJ, and Pusztai L
- Subjects
- Humans, Female, Prognosis, Middle Aged, Gene Expression Regulation, Neoplastic, Receptors, Progesterone metabolism, Receptors, Progesterone genetics, Gene Expression Profiling, Adult, Aged, Neoadjuvant Therapy, Breast Neoplasms genetics, Breast Neoplasms pathology, Breast Neoplasms mortality, Breast Neoplasms metabolism, Receptor, ErbB-2 metabolism, Receptor, ErbB-2 genetics, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms pathology, Triple Negative Breast Neoplasms mortality, Triple Negative Breast Neoplasms metabolism, Receptors, Estrogen metabolism, Receptors, Estrogen genetics, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism
- Abstract
Purpose: The MammaPrint (MP) prognostic assay categorizes breast cancers into high- and low-risk subgroups, and the high-risk group can be further subdivided into high-1 (MP-H1), and very high-risk high-2 (MP-H2). The aim of this analysis was to assess clinical and molecular differences between the hormone receptor-positive (HR+)/HER2-negative MP-H1, -H2, and triple-negative (TN) MP-H1 and -H2 cancers., Experimental Design: Pretreatment gene expression data from 742 HER2-negative breast cancers enrolled in the I-SPY2 neoadjuvant trial were used. Prognostic risk categories were assigned using the MP assay. Transcriptional similarities across the four receptor and prognostic groups were assessed using principal component analyses and by identifying differentially expressed genes. We also examined pathologic complete response rates and event-free survivals by risk group., Results: Principal component analysis showed that HR+/MP-H2 tumors clustered with TN/MP-H2 cancers. Only 125 genes showed differential expression between the HR+/MP-H2 and TN/MP-H2 cancers, whereas 1,465 genes were differentially expressed between HR+/MP-H2 and -H1. Gene set analysis revealed similarly high expression of cell cycle, DNA repair, and immune infiltration-related pathways in HR+/MP-H2 and TN/MP-H2 cancers. HR+/MP-H2 cancers also showed low estrogen receptor-related gene expression. Pathologic complete response rates were similarly high in TN/MP-H2 and HR+/MP-H2 cancers (42% vs. 30.5%; P = 0.11), and MP-H2 cancers with residual cancer had similarly poor event-free survival regardless of estrogen receptor status., Conclusions: In conclusion, HR+/MP-H2 cancers closely resemble TN breast cancers in transcriptional and clinical features and benefit from similar treatment strategies., (©2024 American Association for Cancer Research.)
- Published
- 2025
- Full Text
- View/download PDF
110. GABA A receptor π forms channels that stimulate ERK through a G-protein-dependent pathway.
- Author
-
Wang Y, Zhang Y, Li W, Salovska B, Zhang J, Li T, Li H, Liu Y, Kaczmarek LK, Pusztai L, and Klein DE
- Subjects
- Humans, HEK293 Cells, MAP Kinase Signaling System, Signal Transduction, Receptors, GABA-A metabolism, Receptors, GABA-A genetics, Receptors, GABA-A chemistry, Cryoelectron Microscopy, gamma-Aminobutyric Acid metabolism, Extracellular Signal-Regulated MAP Kinases metabolism
- Abstract
The rare γ-aminobutyric acid type-A receptor (GABA
A R) subunit π (GABRP) is highly expressed in certain cancers, where it stimulates growth through extracellular-regulated kinase (ERK) signaling by an uncharacterized pathway. To elucidate GABRP's signaling mechanism, we determined cryoelectron microscopy (cryo-EM) structures of GABRP embedded in native nanodiscs, both in the presence and absence of GABA. Structurally, GABRP homopentamers closely resemble heteropentameric GABAA R anion channels, transitioning from a closed "resting" state to an open "active" state upon GABA binding. However, functional assays reveal that GABRP responds more like a type-B metabotropic receptor. At physiological concentrations of GABA, chloride flux is not detected. Rather, GABRP activates a G-protein-coupled pathway leading to ERK signaling. Ionotropic activity is only triggered at supraphysiological GABA concentrations, effectively decoupling it from GABRP's signaling functions. These findings provide a structural and functional blueprint for GABRP, opening new avenues for targeted inhibition of GABA growth signals in GABRP-positive cancers., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 Elsevier Inc. All rights reserved.)- Published
- 2025
- Full Text
- View/download PDF
111. Clinical Implications of Breast Cancer Intrinsic Subtypes.
- Author
-
Rios-Hoyo A, Shan NL, Karn PL, and Pusztai L
- Subjects
- Humans, Female, Chemotherapy, Adjuvant, Gene Expression Regulation, Neoplastic, Prognosis, Neoplasm Recurrence, Local genetics, Breast Neoplasms genetics, Breast Neoplasms classification, Breast Neoplasms pathology, Breast Neoplasms therapy, Breast Neoplasms metabolism, Receptors, Estrogen metabolism, Receptors, Estrogen genetics, Biomarkers, Tumor genetics, Biomarkers, Tumor metabolism
- Abstract
Estrogen receptor-positive (ER+) and estrogen receptor-negative (ER-) breast cancers have different genomic architecture and show large-scale gene expression differences consistent with different cellular origins, which is reflected in the luminal (i.e., ER+) versus basal-like (i.e., ER-) molecular class nomenclature. These two major molecular subtypes have distinct epidemiological risk factors and different clinical behaviors. Luminal cancers can be subdivided further based on proliferative activity and ER signaling. Those with a high expression of proliferation-related genes and a low expression of ER-associated genes, called luminal B, have a high risk of early recurrence (i.e., within 5 years), derive significant benefit from adjuvant chemotherapy, and may benefit from adding immunotherapy to chemotherapy. This subset of luminal cancers is identified as the genomic high-risk ER+ cancers by the MammaPrint, Oncotype DX Recurrence Score, EndoPredict, Prosigna, and several other molecular prognostic assays. Luminal A cancers are characterized by low proliferation and high ER-related gene expression. They tend to have excellent prognosis with adjuvant endocrine therapy. Adjuvant chemotherapy may not improve their outcome further. These cancers correspond to the genomic low-risk categories. However, these cancers remain at risk for distant recurrence for extended periods of time, and over 50% of distant recurrences occur after 5 years. Basal-like cancers are uniformly highly proliferative and tend to recur within 3-5 years of diagnosis. In the absence of therapy, basal-like breast cancers have the worst survival, but these also include many highly chemotherapy-sensitive cancers. Basal-like cancers are often treated with preoperative chemotherapy combined with an immune checkpoint inhibitor which results in 60-65% pathologic complete response rates that herald excellent long-term survival. Patients with residual cancer after neoadjuvant therapy can receive additional postoperative chemotherapy that improves their survival. Currently, there is no clinically actionable molecular subclassification for basal-like cancers, although cancers with high androgen receptor (AR)-related gene expression and those with high levels of immune infiltration have better prognosis, but currently their treatment is not different from basal-like cancers in general. A clinically important, minor subset of breast cancers are characterized by frequent HER2 gene amplification and high expression of a few dozen genes, many residing on the HER2 amplicon. This is an important subset because of the highly effective HER2 targeted therapies which are synergistic with endocrine therapy and chemotherapy. The clinical behavior of HER2-enriched cancers is dominated by the underlying ER subtype. ER+/HER2-enriched cancers tend to have more indolent course and lesser chemotherapy sensitivity than their ER counterparts., (© 2025. The Author(s), under exclusive license to Springer Nature Switzerland AG.)
- Published
- 2025
- Full Text
- View/download PDF
112. Rare germline variants in cancer-relevant genes are associated with breast cancer risk in young women with high-risk family history.
- Author
-
Rozenblit M, Qing T, Ye Y, Zhao H, Hofstatter E, Singh V, Reisenbichler E, Murray M, and Pusztai L
- Subjects
- Humans, Female, Adult, Case-Control Studies, Polymorphism, Single Nucleotide, Middle Aged, Biomarkers, Tumor genetics, Young Adult, Risk Factors, Breast Neoplasms genetics, Breast Neoplasms epidemiology, Genetic Predisposition to Disease, Germ-Line Mutation, Exome Sequencing
- Abstract
Introduction: It is incompletely understood why some women develop breast cancer 15-20 years earlier than the majority of women. We hypothesize that women with early-onset breast cancer and high-risk family history without germline pathogenic variants in cancer-predisposing genes (CPGs, n = 83) have a higher load of germline high functional impact variants (gHFI) in cancer hallmark genes (n = 1508) compared to healthy controls., Methods: Germline whole exome sequencing data were analyzed from 5818 breast cancer cases from UKBiobank and from 94 young women with breast cancer without CPG pathogenic variants from the Yale Cancer Prevention Clinic, and compared to 149 controls from Yale GENERATIONS project and 56,917 controls from UKBiobank. Rare gHFI variants were compared between cases and controls using the burden test and the optimal unified SNP-set Kernel Association Test (SKAT-O). We assessed germline versus somatic origins of pathway level alterations in the TCGA and Yale data., Results: In UKBiobank, higher gHFI variant load was seen in CPGs in cancer cases regardless of family history (p < 8.78 × 10
-8 ), and in hallmark genes in cases with family history vs controls (p = 0.0093). Similarly, numerically higher rare gHFI burden was seen in hallmark genes in the Yale cohort vs controls, but this difference was not statistically significant. Pathway level alterations were dominated by somatic events. These results suggest that rare germline variants in cancer biology-related genes partly mediate the contribution of family history to cancer risk in individuals without germline alterations in high penetrance CPGs., Competing Interests: Declarations. Conflict of interests: LP consults for Pfizer, Bristol-Myers Squibb, AstraZeneca, Merck, Roche, Novartis, Natera. EH works for GSK. Ethics approval and consent: All participants from Yale signed informed consent for germline DNA analysis and this study was approved by the Yale Institutional Review Board. UKBioBank data were de-identified and met criteria for IRB exemption. Consent for publication: Not applicable., (© 2024. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)- Published
- 2025
- Full Text
- View/download PDF
113. Molecular characteristics of advanced colorectal cancer and multi-hit PIK3CA mutations.
- Author
-
Yasin F, Sokol E, Vasan N, Pavlick DC, Huang RSP, Pelletier M, Levy MA, Pusztai L, Lacy J, Zhang JY, Ross JS, and Cecchini M
- Subjects
- Humans, Male, Female, Middle Aged, Aged, Adult, Aged, 80 and over, Microsatellite Instability, Class I Phosphatidylinositol 3-Kinases genetics, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, Mutation
- Abstract
Introduction: Approximately 20% of patients living with colorectal cancer (CRC) have activating mutations in their tumors in the PIK3CA oncogene. Two or more activating mutations (multi-hit) for the PIK3CA allele increase PI3K⍺ signaling compared to single-point mutations, resulting in exceptional response to PI3K⍺ inhibition. We aimed to identify the prevalence of PIK3CA multi-hit mutations in metastatic CRC to identify patients who may benefit from PI3K inhibitors., Methods: The Foundation Medicine database (Boston, MA, USA) was analyzed for patients with CRC who underwent genomic profiling on tumor DNA isolated during routine clinical care from 2013 to 2021. Molecular and clinical variables were abstracted for patients with PIK3CA mutations., Results: We identified 49 051 patients with CRC who underwent Foundation Medicine testing. 710/41154 (1.7%) patients had multi-hit PIK3CA mutations, of which 53% were male (n = 448) with a median age of 60. Microsatellite status was available for 697 patients with multi-hit PIK3CA and 17.6% (123/697) were microsatellite instability-high. Clinically relevant mutations in KRAS and BRAFV600E were seen in 459/710 (64.7%) and 65/710 (9.1%), respectively. The 4 most common PIK3CA variants were H1047R (9.8%), E545K (9.2%), E542K (9.0%), and R88Q (7.1%). The most common variant pair was E542K-E545K (4.7%)., Conclusions: Multi-hit mutations in PIK3CA are seen in 1.7% of advanced CRC, a meaningful prevalence given the high burden of CRC worldwide, and may represent a subset of patients that have enhanced sensitivity to PI3K inhibition. Future investigation regarding the clinical utility of PI3K inhibitors is warranted in multi-hit PIK3CA CRC., (© The Author(s) 2024. Published by Oxford University Press.)
- Published
- 2024
- Full Text
- View/download PDF
114. Race and Clinical Outcomes in Hormone Receptor-Positive, HER2-Negative, Node-Positive Breast Cancer in the Randomized RxPONDER Trial.
- Author
-
Abdou Y, Barlow WE, Gralow JR, Meric-Bernstam F, Albain KS, Hayes DF, Lin NU, Perez EA, Goldstein LJ, Chia SKL, Dhesy-Thind S, Rastogi P, Alba E, Delaloge S, Schott AF, Shak S, Sharma P, Lew DL, Miao J, Unger JM, Tripathy D, Hortobagyi GN, Pusztai L, and Kalinsky K
- Abstract
Background: The phase III RxPONDER trial has impacted treatment for node-positive(1-3), hormone receptor-positive, HER2-negative breast cancer with 21-gene recurrence score (RS) ≤ 25. We investigated how these findings apply to different racial and ethnic groups within the trial., Methods: The trial randomized women to endocrine therapy (ET) or to chemotherapy plus ET. The primary clinical outcome was invasive disease-free survival (IDFS) with distant relapse-free survival (DRFS) as a secondary outcome. Multivariable Cox models were used to evaluate the association between race/ethnicity and survival outcomes, adjusting for clinicopathologic characteristics, RS, and treatment., Results: A total of 4,048 women with self-reported race/ethnicity were included: Hispanic (15.1%), non-Hispanic Black (NHB)(6.1%), Native American/Pacific Islander (0.8%), Asian (8.0%), and non-Hispanic White (NHW)(70%). No differences in RS distribution, tumor size, or number of positive nodes were observed by race/ethnicity. Relative to NHWs, IDFS was worse for NHBs (5-year IDFS 91.6% vs 87.1%, HR = 1.37; 95% CI 1.03-1.81) and better for Asians (91.6% vs 93.9%, HR = 0.64; 95% CI 0.46-0.91). Relative to NHW, DRFS was worse for NHBs (5-year DRFS 95.8% vs 91.0%, HR = 1.65; 95% CI 1.17-2.32) and better for Asians (95.8% vs 96.7%, HR = 0.59; 95% CI 0.37-0.95). Adjusting for clinical characteristics, particularly body mass index, diminished the effect of race on outcomes. Chemotherapy treatment efficacy did not differ by race/ethnicity., Conclusions: NHB women had worse clinical outcomes compared to NHWs in the RxPONDER trial despite similar RS and comparable treatment. Our study emphasizes the persistent racial disparities in breast cancer outcomes while highlighting complex interactions among contributing factors., Trial Registration: ClinicalTrials.gov: NCT01272037., (© The Author(s) 2024. Published by Oxford University Press. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2024
- Full Text
- View/download PDF
115. Development and Validation of the RSClinN+ Tool to Predict Prognosis and Chemotherapy Benefit for Hormone Receptor-Positive, Node-Positive Breast Cancer.
- Author
-
Pusztai L, Hoag JR, Albain KS, Barlow WE, Stemmer SM, Meisner A, Hortobagyi GN, Shak S, Rae JM, Baehner R, Sharma P, and Kalinsky KM
- Abstract
Purpose: Clinicopathological factors and the 21-gene Oncotype DX Breast Recurrence Score (RS) test both influence prognosis. Our goal was to develop a new tool, RSClinN+, to individualize recurrence risk and chemotherapy benefit predictions by menopausal status for patients with HR+/human epidermal growth factor receptor 2-negative, lymph node-positive breast cancer by integrating the RS result with clinicopathological factors (grade, tumor size, age)., Methods: We used patient-level data from 5,283 patients treated with chemoendocrine therapy (CET) versus endocrine therapy alone (ET) in the S1007 (N = 4,916) and S8814 (N = 367) trials to develop the tool. Cox proportional hazards regression models stratified by trial were used to estimate 5-year invasive disease-free survival for pre- and postmenopausal woman, respectively. The integrated RSClinN+ model was compared with RS alone and clinicopathological models using likelihood ratio tests. Absolute CET benefit was estimated as the difference between ET and CET risk estimates. Validation of RSClinN+ was performed in 592 patients with node-positive disease in the Clalit Health Services registry., Results: RSClinN+ provides better prognostic information than RS model alone (premenopausal P = .034; postmenopausal P < .001) or clinicopathological model alone (premenopausal P = .002; postmenopausal, P < .001). In postmenopausal women, RS showed interaction with CET benefit ( P = .016), with RSClinN+ absolute CET benefit ranging from <0.1% to 21.5% over RS ranges 0-50. In premenopausal patients with RS ≤25, there was no significant interaction between RS and CET benefit. In external validation, RSClinN+ risk estimates were prognostic (hazard ratio, 1.75 [95% CI, 1.38 to 2.20]) and concordant with observed risk (Lin's concordance, 0.92)., Conclusion: RSClinN+ provides improved estimates of prognosis and absolute CET benefit for individual patients compared with RS or with clinical data alone and could be used in patient counseling.
- Published
- 2024
- Full Text
- View/download PDF
116. Phosphoenolpyruvate carboxykinase-2 (PCK2) is a therapeutic target in triple-negative breast cancer.
- Author
-
Gunasekharan V, Lin HK, Marczyk M, Rios-Hoyo A, Campos GE, Shan NL, Ahmed M, Umlauf S, Gareiss P, Raaisa R, Williams R, Cardone R, Siebel S, Kibbey R, Surovtseva YV, and Pusztai L
- Subjects
- Humans, Cell Line, Tumor, Female, Enzyme Inhibitors pharmacology, Phosphoenolpyruvate Carboxykinase (ATP) metabolism, Phosphoenolpyruvate Carboxykinase (ATP) genetics, Phosphoenolpyruvate Carboxykinase (ATP) antagonists & inhibitors, Antineoplastic Agents pharmacology, Antineoplastic Agents therapeutic use, Cell Survival drug effects, Small Molecule Libraries pharmacology, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms pathology, Triple Negative Breast Neoplasms metabolism, Phosphoenolpyruvate Carboxykinase (GTP) metabolism, Phosphoenolpyruvate Carboxykinase (GTP) genetics, Phosphoenolpyruvate Carboxykinase (GTP) antagonists & inhibitors, Cell Proliferation drug effects
- Abstract
Purpose: Metabolic rewiring in malignant transformation is often accompanied by altered expression of metabolic isozymes. Phosphoenolpyruvate carboxykinase-2 (PCK2) catalyzes the rate-limiting step of gluconeogenesis and is the dominant isoform in many cancers including triple-negative breast cancer (TNBC). Our goal was to identify small molecule inhibitors of PCK2 enzyme activity., Methods: We assessed the impact of PCK2 down regulation with shRNA on TNBC cell growth in vitro and used AtomNet® deep convolutional neural network software to identify potential small molecule inhibitors of PCK2-based structure. We iteratively tested candidate compounds in an in vitro PCK-2 enzyme assay. The impact of the top hit on metabolic flux and cell viability was also assessed., Results: PCK2 downregulation decreased growth of BT-549 and MDA-MB-231 cells and reduced metabolic flux through pyruvate carboxylase. The first AtomNet® in silico structural screen of 7 million compounds yielded 86 structures that were tested in PCK2 enzyme assay in vitro. The top hit (IC
50 = 2.4 µM) was used to refine a second round of in silico screen that yielded 82 candidates to be tested in vitro, which resulted in 45 molecules with inhibition > 20%. In the second in vitro screen we also included 3-(3,4-dihydroxyphenyl)-2-hydroxypropanoate, previously suggested to be PCK2 inhibitor based on structure, which emerged as the top hit. The specificity of this compound was tested in PCK1 and PCK2 enzymatic assays and showed IC50 of 500 nM and 3.5-27 nM for PCK1 and PCK2, respectively., Conclusion: 3-(3,4-dihydroxyphenyl)-2-hydroxypropanoate is a high affinity PCK2 enzyme inhibitor that also has significant growth inhibitory activity in breast cell lines in vitro and represents a potential therapeutic lead compound., (© 2024. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)- Published
- 2024
- Full Text
- View/download PDF
117. Datopotamab-deruxtecan in early-stage breast cancer: the sequential multiple assignment randomized I-SPY2.2 phase 2 trial.
- Author
-
Khoury K, Meisel JL, Yau C, Rugo HS, Nanda R, Davidian M, Tsiatis B, Chien AJ, Wallace AM, Arora M, Rozenblit M, Hershman DL, Zimmer A, Clark AS, Beckwith H, Elias AD, Stringer-Reasor E, Boughey JC, Nangia C, Vaklavas C, Omene C, Albain KS, Kalinsky KM, Isaacs C, Tseng J, Roussos Torres ET, Thomas B, Thomas A, Sanford A, Balassanian R, Ewing C, Yeung K, Sauder C, Sanft T, Pusztai L, Trivedi MS, Outhaythip A, Li W, Onishi N, Asare AL, Beineke P, Norwood P, Brown-Swigart L, Hirst GL, Matthews JB, Moore B, Fraser Symmans W, Price E, Beedle C, Perlmutter J, Pohlmann P, Shatsky RA, DeMichele A, Yee D, van 't Veer LJ, Hylton NM, and Esserman LJ
- Subjects
- Humans, Female, Middle Aged, Adult, Neoplasm Staging, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Aged, Antibodies, Monoclonal, Humanized therapeutic use, Treatment Outcome, Doxorubicin therapeutic use, Doxorubicin administration & dosage, Receptor, ErbB-2 genetics, Receptor, ErbB-2 metabolism, Cyclophosphamide therapeutic use, Immunoconjugates therapeutic use, Trastuzumab, Camptothecin analogs & derivatives, Breast Neoplasms drug therapy, Breast Neoplasms pathology, Breast Neoplasms genetics, Neoadjuvant Therapy
- Abstract
Among the goals of patient-centric care are the advancement of effective personalized treatment, while minimizing toxicity. The phase 2 I-SPY2.2 trial uses a neoadjuvant sequential therapy approach in breast cancer to further these goals, testing promising new agents while optimizing individual outcomes. Here we tested datopotamab-deruxtecan (Dato-DXd) in the I-SPY2.2 trial for patients with high-risk stage 2/3 breast cancer. I-SPY2.2 uses a sequential multiple assignment randomization trial design that includes three sequential blocks of biologically targeted neoadjuvant treatment: the experimental agent(s) (block A), a taxane-based regimen tailored to the tumor subtype (block B) and doxorubicin-cyclophosphamide (block C). Patients are randomized into arms consisting of different investigational block A treatments. Algorithms based on magnetic resonance imaging and core biopsy guide treatment redirection after each block, including the option of early surgical resection in patients predicted to have a high likelihood of pathological complete response, the primary endpoint. There are two primary efficacy analyses: after block A and across all blocks for the six prespecified breast cancer subtypes (defined by clinical hormone receptor/human epidermal growth factor receptor 2 (HER2) status and/or the response-predictive subtypes). We report results of 103 patients treated with Dato-DXd. While Dato-DXd did not meet the prespecified threshold for success (graduation) after block A in any subtype, the treatment strategy across all blocks graduated in the hormone receptor-negative HER2
- Immune- DNA repair deficiency- subtype with an estimated pathological complete response rate of 41%. No new toxicities were observed, with stomatitis and ocular events occurring at low grades. Dato-DXd was particularly active in the hormone receptor-negative/HER2- Immune- DNA repair deficiency- signature, warranting further investigation, and was safe in other subtypes in patients who followed the treatment strategy. ClinicalTrials.gov registration: NCT01042379 ., Competing Interests: Competing interests: J.L.M. reports institutional research funding from AstraZeneca, Seagen, Sermonix and Olema and advisory and consulting roles with Pfizer, Seagen, Sermonix, Novartis, Stemline, AstraZeneca, Olema, GlaxoSmithKline and GE Healthcare. C.Y. reports institutional research grant from NCI/NIH; salary support and travel reimbursement from QLHC; a United States patent titled ‘Breast cancer response prediction subtypes’ (no. 18/174,491); and University of California Inventor Share. H.S.R. reports institutional research support from AstraZeneca, Daiichi Sankyo, Inc., F. Hoffmann–La Roche AG/Genentech, Inc., Gilead Sciences, Inc., Lilly; Merck and Co., Novartis Pharmaceuticals Corporation, Pfizer, Stemline Therapeutics, OBI Pharma, Ambrx, Greenwich Pharma; and advisory and consulting roles with Chugai, Puma, Sanofi, Napo and Mylan. R.N. reports research funding from Arvinas, AstraZeneca, BMS, Corcept Therapeutics, Genentech/Roche, Gilead, GSK, Merck, Novartis, OBI Pharma, OncoSec, Pfizer, Relay, Seattle Genetics, Sun Pharma and Taiho and advisory roles with AstraZeneca, BeyondSpring, Daiichi Sankyo, Exact Sciences, Fujifilm, GE, Gilead, Guardant Health, Infinity, iTeos, Merck, Moderna, Novartis, OBI, OncoSec, Pfizer, Sanofi, Seagen and Stemline. M.D. reports research grants from NIH/NCI and NIH/NIA, and contracts from PCORI. A.J.C. reports institutional research funding from Merck, Amgen, Puma, Seagen, Pfizer and Olema and advisory roles with AstraZeneca and Genentech. A.Z. reports institutional research funding from Merck, honoraria for Medscape and participation on Pfizer Advisory Board. A.S.C. reports institutional research funding from Novartis and Lilly. A.D.E. reports support from Scorpion, Infinity and Deciphera. E.S.-R. reports grants from V Foundation, NIH, Susan G. Komen; institutional research funding from GSK, Seagen, Pfizer, Lilly; consulting and honoraria from Novartis, Merck, Seagen, AstraZeneca, Lilly; Cancer Awareness Network Board member and support from ASCO and NCCN. J.C.B. reports institutional research funding from Eli Lilly and SymBioSis, participation on the Data Safety Monitoring Committee of Cairn Surgical and honoraria from PER, PeerView, OncLive, EndoMag and UpToDate. C.V. reports institutional research funding from Pfizer, Seagen, H3 Biomedicine/Eisai, AstraZeneca, CytomX; research funding to previous institution from Genentech, Roche, Pfizer, Incyte, Pharmacyclics, Novartis, TRACON Pharmaceuticals, Innocrin Pharmaceuticals, Zymeworks and H3 Biomedicine; advisory and consulting roles with Guidepoint, Novartis, Seagen, Daiichi Sankyo, AstraZeneca and Cardinal Health; unpaid consulting with Genentech; and participation in non-CME activity with Gilead, AstraZeneca. C.O. reports consulting fees from AstraZeneca, Guardant Health and Jazz Pharmaceuticals. K.S.A. reports institutional research funding from AstraZeneca, Daiichi Sankyo, Seattle Genetics and QLHC; Independent Data and Safety Monitoring committee at Seattle Genetics. K.M.K. reports advisory and consultant roles for Eli Lilly, Pfizer, Novartis, AstraZeneca, Daiichi Sankyo, Puma, 4D Pharma, OncoSec, Immunomedics, Merck, Seagen, Mersana, Menarini Silicon Biosystems, Myovant, Takeda, Biotheranostics, Regor, Gilead, Prelude Therapeutics, RayzeBio, eFFECTOR Therapeutics and Cullinan Oncology; and reports institutional research funding from Genentech/Roche, Novartis, Eli Lilly, AstraZeneca, Daiichi Sankyo and Ascentage. C.I. reports institutional research funding from Tesaro/GSK, Seattle Genetics, Pfizer, AstraZeneca, BMS, Genentech, Novartis and Regeneron; consultancy roles with AstraZeneca, Genentech, Gilead, ION, Merck, Medscape, MJH Holdings, Novartis, Pfizer, Puma and Seagen; and royalties from Wolters Kluwer (UptoDate) and McGraw Hill (Goodman and Gillman). J.T. serves as institutional principal investigator for clinical trial with Intuitive Surgical; editor lead for ABS, CGSO, SCORE, Breast Education Committee Track Leader, ASCO SESAP 19 and Breast Co-Chair, ACS. A.T. owns stock at Johnson and Johnsons, Gilead, Bristol Myers Squibb, reports participation on Pfizer Advisory Board: AstraZeneca and reports institutional research funding from Merck and Sanofi and royalties from UptoDate. R.B. reports a consultancy role at Genentech and stock ownership at Cerus Corp. K.Y. received research support unrelated to this work and paid to the institution from Pfizer, Gilead, Seagen, Dantari Pharmaceuticals, Treadwell Therapeutics, and Relay Therapeutics; support from American Cancer Society IRG grant no. IRG-19-230-48-IRG, UC San Diego Moores Cancer Center, Specialized Cancer Center support grant NIH/NCI P30CA023100, Curebound Discovery Award (2023, 2024). T.S. reports honoraria from Hologic. L.P. reports institutional research funding from Susan Komen Foundation, Breast Cancer Research Foundation, NCI, Pfizer, AstraZeneca, Menarini/Stemline, Bristol Myers Squibb, Merck and Co.; consulting fees from AstraZeneca, Merck, Novartis, Genentech, Natera, Personalis, Exact Sciences and Stemline/Menarini; patent titled ‘Method of measuring residual cancer and predicting patient survival’ (no. 7711494); and Data and Safety Monitoring Board member of the DYNASTY Breast02, OPTIMA and PARTNER trials. M.S.T. reports institutional research funding from Lilly, Gilead Sciences, Phoenix Molecular Designs, AstraZeneca, Regeneron, Merck and Novartis. A.L.A., P.B. and P.N. are employees of QLHC. G.L.H. reports institutional research grant from NIH (1R01CA255442). W.F.S. reports shares of IONIS Pharmaceuticals and Eiger Biopharmaceuticals, received consulting fees from AstraZeneca, is a cofounder with equity in Delphi Diagnostics and issued patents for (1) a method to calculate residual cancer burden and (2) genomic signature to measure sensitivity to endocrine therapy. J.P. reports honoraria from Methods in Clinical Research—faculty SCION workshop; support from ASCO and advocate scholarship; AACR—SSP program; VIVLI, U Wisc SPORE—EAB, QuantumLEAD—COVID DSMB, PCORI—reviewer and I-SPY advocate lead. P.P. reports institutional research funding from Genentech/Roche, Fabre-Kramer, Advanced Cancer Therapeutics, Caris Centers of Excellence, Pfizer, Pieris Pharmaceuticals, Cascadian Therapeutics, Bolt, Byondis, Seagen, Orum Therapeutics and Carisma Therapeutics; consulting fees from Personalized Cancer Therapy, OncoPlex Diagnostics, Immunonet BioSciences, Pfizer, HERON, Puma Biotechnology, Sirtex, CARIS Life sciences, Juniper, Bolt Biotherapeutics and AbbVie; honoraria from DAVA Oncology, OncLive/MJH Life Sciences, Frontiers—publisher, SABCS and ASCO; Speakers’ Bureau: Genentech/Roche (past); United States patent no. 8486413, United States patent no. 8501417, United States patent no. 9023362, United States patent no. 9745377; uncompensated roles with Pfizer, Seagen and Jazz. R.A.S. reports institutional research funding from OBI Pharma, QLHC, AstraZeneca and Gilead, serves on AstraZeneca and Stemline Advisory Boards and Gilead Speaker’s Bureau and reports consultancy role with QLHC. A.D. reports institutional research funding from Novartis, Pfizer, Genentech and NeoGenomics; Program Chair, Scientific Advisory Committee, ASCO. D.Y. reports research funding from NIH/NCI P30 CA 077598, P01 CA234228-01 and R01CA251600, consulting fees from Martell Diagnostics, and honoraria and travel for speaking at the ‘International Breast Cancer Conference.’ L.J.v.V. is a founding advisor and shareholder of Exai Bio and is a part-time employee and owns stock in Agendia. N.M.H. reports institutional research funding from NIH. L.J.E. reports funding from Merck and Co., participation on an advisory board for Blue Cross Blue Shield and personal fees from UpToDate and is an unpaid board member of QLHC. The other authors declare no competing interests., (© 2024. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2024
- Full Text
- View/download PDF
118. Identification of HER2-positive breast cancer molecular subtypes with potential clinical implications in the ALTTO clinical trial.
- Author
-
Rediti M, Venet D, Joaquin Garcia A, Maetens M, Vincent D, Majjaj S, El-Abed S, Di Cosimo S, Ueno T, Izquierdo M, Piccart M, Pusztai L, Loi S, Salgado R, Viale G, Rothé F, and Sotiriou C
- Subjects
- Female, Humans, Middle Aged, Antineoplastic Agents, Immunological therapeutic use, Biomarkers, Tumor metabolism, Biomarkers, Tumor genetics, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Neoplasm Recurrence, Local genetics, Prognosis, Tumor Microenvironment genetics, Randomized Controlled Trials as Topic, Clinical Trials, Phase III as Topic, Breast Neoplasms genetics, Breast Neoplasms drug therapy, Breast Neoplasms pathology, Breast Neoplasms metabolism, Receptor, ErbB-2 metabolism, Receptor, ErbB-2 genetics, Trastuzumab therapeutic use
- Abstract
In HER2-positive breast cancer, clinical outcome and sensitivity to HER2-targeted therapies are influenced by both tumor and microenvironment features. However, we are currently unable to depict the molecular heterogeneity of this disease with sufficient granularity. Here, by performing gene expression profiling in HER2-positive breast cancers from patients receiving adjuvant trastuzumab in the ALTTO clinical trial (NCT00490139), we identify and characterize five molecular subtypes associated with the risk of distant recurrence: immune-enriched, proliferative/metabolic-enriched, mesenchymal/stroma-enriched, luminal, and ERBB2-dependent. Additionally, we validate the biological profiles of the subtypes and explore their prognostic/predictive value in external cohorts, namely the NeoALTTO trial (NCT00553358), SCAN-B (NCT02306096), I-SPY2 (NCT01042379), METABRIC and TCGA. Immune-enriched tumors present better survival outcomes, in contrast to mesenchymal/stroma-enriched and proliferative/metabolic-enriched tumors, while luminal and ERBB2-dependent tumors are characterized by low and high rates of pathological complete response, respectively. Of note, these molecular subtypes provide the rationale for treatment approaches leveraging the heterogeneous biology of HER2-positive breast cancer., Competing Interests: Competing interests: The authors declare the following competing interests. S.EA.: grants via the affiliation from Novartis for the submitted work and from Roche/Genentech, Astra Zeneca, Pfizer, and BCRF outside the submitted work. S.DC.: consultation fees from Pierre-Fabre, IQVIA, and Medica Scientia Innovation Research (MEDSIR); institutional grant from Fondazione Associazione Italiana Ricerca contro il Cancro (AIRC); and Cancer Can.Heal European EU4 Health Programme 101080009-European Commission. T.U.: honoraria from Astra Zeneca, Novartis Pharma K.K., Eisai Co., Ltd., Chugai Pharmaceutical Co. Ltd; research grant from Eli Lilly Japan K.K. M.I.: employee of Novartis, owner of Novartis shares. M.P.: invited speaker for AstraZeneca, Lilly, MSD, Novartis, Pfizer, Roche-Genentech; consultant for Roche-Genentech; advisory board for Frame Therapeutics, Gilead, Menarini, NBE Therapeutics, Odonate, Roche-Genentech, SeaGen, Seattle Genetics; member of boards of directors, scientific board for Oncolytics; research grants to her Institution from AstraZeneca, Lilly, Gilead; funding to her Institution from Menarini, MSD, Novartis, Pfizer, Radius, Roche-Genentech, Servier, Synthon. L.P.: consulting fees and honoraria for advisory board participation from Pfizer, Astra Zeneca, Merck, Novartis, Bristol-Myers Squibb, Stemline-Menarini, GlaxoSmithKline, Genentech/Roche, Personalis, Daiichi, Natera, Exact Sciences and institutional research funding from Seagen, GlaxoSmithKline, AstraZeneca, Merck, Pfizer and Bristol Myers Squibb. S.L.: research funding to institution from Novartis, Bristol Myers Squibb, MSD, Puma Biotechnology, Eli Lilly, Nektar Therapeutics, Astra Zeneca/Daiichi Sankyo and Seattle Genetics; consultant (not compensated) to Seattle Genetics, Novartis, Bristol Myers Squibb, MSD, AstraZeneca/Daiichi Sankyo, Eli Lilly, Pfizer, Gilead Therapeutics, Nektar Therapeutics, PUMA Biotechnologies, and Roche-Genentech; consultant (paid to institution) to Novartis, GlaxoSmithKline, Roche-Genentech, Astra Zeneca/Daiichi Sankyo, Pfizer, Gilead Therapeutics, Seattle Genetics, MSD, Tallac Therapeutics, Eli Lilly and Bristol Myers Squibb. R.S.: non-financial support from Merck, Case 45, and Bristol Myers Squibb (BMS); research support from Merck, Puma Biotechnology, and Roche; personal fees from Roche, BMS, Astra Zeneca, Daiichi Sankyo and Exact Sciences for advisory boards. G.V.: grants/research supports from Roche/Genentech, Ventana Medical Systems, Dako/Agilent Technologies; honoraria or consultation fees from Ventana, Dako/Agilent, Roche, MSD Oncology, AstraZeneca, Daiichi Sankyo, Pfizer, Gilead. C.S.: advisory board (receipt of honoraria or consultations fees) for Astellas, Cepheid, Vertex, Seattle genetics, Puma, Amgen, Exact Sciences, INC, Merck & Co; invited speaker for Eisai, Prime Oncology, Teva, Foundation Medicine, Exact Sciences; advisory equity from Signatur Biosciences; other support (travel, accommodation expenses) from Roche, Genentech, Pfizer. The remaining authors declare no non-financial or financial competing interests., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
119. Overall Survival with Pembrolizumab in Early-Stage Triple-Negative Breast Cancer.
- Author
-
Schmid P, Cortes J, Dent R, McArthur H, Pusztai L, Kümmel S, Denkert C, Park YH, Hui R, Harbeck N, Takahashi M, Im SA, Untch M, Fasching PA, Mouret-Reynier MA, Foukakis T, Ferreira M, Cardoso F, Zhou X, Karantza V, Tryfonidis K, Aktan G, and O'Shaughnessy J
- Subjects
- Aged, Female, Humans, Middle Aged, Antineoplastic Agents, Immunological administration & dosage, Antineoplastic Agents, Immunological adverse effects, Carboplatin administration & dosage, Carboplatin adverse effects, Chemotherapy, Adjuvant adverse effects, Chemotherapy, Adjuvant methods, Cyclophosphamide administration & dosage, Cyclophosphamide adverse effects, Epirubicin administration & dosage, Epirubicin adverse effects, Kaplan-Meier Estimate, Paclitaxel administration & dosage, Paclitaxel adverse effects, Survival Analysis, Treatment Outcome, Double-Blind Method, Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized adverse effects, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Antineoplastic Combined Chemotherapy Protocols adverse effects, Neoadjuvant Therapy adverse effects, Neoadjuvant Therapy methods, Neoplasm Staging, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms mortality, Triple Negative Breast Neoplasms pathology, Triple Negative Breast Neoplasms surgery
- Abstract
Background: In patients with early-stage triple-negative breast cancer, the phase 3 KEYNOTE-522 trial showed significant improvements in pathological complete response and event-free survival with the addition of pembrolizumab to platinum-containing chemotherapy. Here we report the final results for overall survival., Methods: We randomly assigned, in a 2:1 ratio, patients with previously untreated stage II or III triple-negative breast cancer to receive neoadjuvant therapy with four cycles of pembrolizumab (at a dose of 200 mg) or placebo every 3 weeks plus paclitaxel and carboplatin, followed by four cycles of pembrolizumab or placebo plus doxorubicin-cyclophosphamide or epirubicin-cyclophosphamide. After definitive surgery, patients received adjuvant pembrolizumab (pembrolizumab-chemotherapy group) or placebo (placebo-chemotherapy group) every 3 weeks for up to nine cycles. The primary end points were pathological complete response and event-free survival. Overall survival was a secondary end point., Results: Of the 1174 patients who underwent randomization, 784 were assigned to the pembrolizumab-chemotherapy group and 390 to the placebo-chemotherapy group. At the data-cutoff date (March 22, 2024), the median follow-up was 75.1 months (range, 65.9 to 84.0). The estimated overall survival at 60 months was 86.6% (95% confidence interval [CI], 84.0 to 88.8) in the pembrolizumab-chemotherapy group, as compared with 81.7% (95% CI, 77.5 to 85.2) in the placebo-chemotherapy group (P = 0.002). Adverse events were consistent with the established safety profiles of pembrolizumab and chemotherapy., Conclusions: Neoadjuvant pembrolizumab plus chemotherapy followed by adjuvant pembrolizumab resulted in a significant improvement, as compared with neoadjuvant chemotherapy alone, in overall survival among patients with early-stage triple-negative breast cancer. (Funded by Merck Sharp and Dohme, a subsidiary of Merck [Rahway, NJ]; KEYNOTE-522 ClinicalTrials.gov number, NCT03036488.)., (Copyright © 2024 Massachusetts Medical Society.)
- Published
- 2024
- Full Text
- View/download PDF
120. Multi-platform biomarkers of response to an immune checkpoint inhibitor in the neoadjuvant I-SPY 2 trial for early-stage breast cancer.
- Author
-
Campbell MJ, Wolf DM, Yau C, Brown-Swigart L, Wulfkuhle J, Gallagher IR, Zhu Z, Bolen J, Vandenberg S, Hoyt C, Mori H, Borowsky A, Sit L, Perlmutter J, Asare SM, Nanda R, Liu MC, Yee D, DeMichele AM, Hylton NM, Pusztai L, Berry DA, Hirst GL, Petricoin EF, Veer LV, and Esserman L
- Subjects
- Humans, Female, B7-H1 Antigen metabolism, B7-H1 Antigen antagonists & inhibitors, Neoplasm Staging, Middle Aged, Programmed Cell Death 1 Receptor antagonists & inhibitors, Programmed Cell Death 1 Receptor metabolism, Treatment Outcome, Neoadjuvant Therapy methods, Breast Neoplasms drug therapy, Breast Neoplasms immunology, Breast Neoplasms pathology, Breast Neoplasms metabolism, Immune Checkpoint Inhibitors therapeutic use, Immune Checkpoint Inhibitors pharmacology, Biomarkers, Tumor metabolism, Tumor Microenvironment immunology, Tumor Microenvironment drug effects
- Abstract
Only a subset of patients with breast cancer responds to immune checkpoint blockade (ICB). To better understand the underlying mechanisms, we analyze pretreatment biopsies from patients in the I-SPY 2 trial who receive neoadjuvant ICB using multiple platforms to profile the tumor microenvironment. A variety of immune cell populations and markers of immune/cytokine signaling associate with pathologic complete response (pCR). Interestingly, these differ by breast cancer receptor subtype. Measures of the spatial distributions of immune cells within the tumor microenvironment, in particular colocalization or close spatial proximity of PD-1
+ T cells with PD-L1+ cells (immune and tumor cells), are significantly associated with response in the overall cohort as well as the in the triple negative (TN) and HR+ HER2- subtypes. Our findings indicate that biomarkers associated with immune cell signaling, immune cell densities, and spatial metrics are predictive of neoadjuvant ICB efficacy in breast cancer., Competing Interests: Declaration of interests J.W. reports honoraria from DAVA Oncology; consults for Baylor College of Medicine; has ownership in Theralink; and is co-inventor of the RPPA technology, and phospho-HER2 and -EGFR response predictors with filed patents. C.H. is an employee of Akoya Biosciences. R.N. reports grants from Quantum Leap, AstraZeneca, Celgene, Corcept Therapeutics, Genentech/Roche, Immunomedics, Merck, OBI Pharma, Odonate Therapeutics, Pfizer, and Seattle Genetics outside the submitted work; and personal fees from Aduro, AstraZeneca, Athenex, Celgene, Daiichi Sankyo, G1 Therapeutics, Genentech, MacroGenics, Merck, Novartis, Pfizer, Puma, and Syndax. M.C.L. reports support from Eisai, Genentech, GRAIL, Menarini Silicon Biosystems, Merck, Novartis, Seattle Genetics, and Tesaro. D.Y. reports unrelated support from Boehringer Ingelheim. A.M.D. reports honoraria or consulting for Pfizer and Context Therapeutics and reports support from Novartis, Pfizer, Genentech, Calithera, and Menarini. L.P. reports consulting fees and honoraria for advisory board participation from Pfizer, AstraZeneca, Merck, Novartis, Bristol-Myers Squibb, GlaxoSmithKline, Genentech/Roche, Personalis, Daiichi, Natera, and Exact Sciences and institutional research funding from Seagen, GlaxoSmithKline, AstraZeneca, Merck, Pfizer, and Bristol-Myers Squibb. D.A.B. is co-owner of Berry Consultants LLC, a company that designs adaptive clinical trials (including I-SPY2). E.F.P. reports leadership, stock/ownership, consulting/advisory, and travel funds from Perthera and Ceres Nanosciences; stock and consulting/advisory for Theralink Technologies, Inc; support from Ceres Nanosciences, GlaxoSmithKline, AbbVie, Symphogen, Genentech, SpringWorks Therapeutics, and Deciphera Therapeutics; and patents/royalties from NIH and filed patents for p-HER2 and p-EGFR response predictors. L.v.V. is a co-inventor of the MammaPrint signature and a part-time employee and stockholder of Agendia NV. L.E. is an unpaid member of the board of directors of Quantum Leap Healthcare Collaborative (QLHC), has received grant support from QLHC for the I-SPY2 trial, and is on the Blue Cross/Blue Shield Medical Advisory Panel., (Copyright © 2024 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
121. Molecular adaptation to neoadjuvant immunotherapy in triple-negative breast cancer.
- Author
-
Denkert C, Schneeweiss A, Rey J, Karn T, Hattesohl A, Weber KE, Rachakonda S, Braun M, Huober J, Jank P, Sinn HP, Zahm DM, Felder B, Hanusch C, Teply-Szymanski J, Marmé F, Fehm T, Thomalla J, Sinn BV, Stiewe T, Marczyk M, Blohmer JU, van Mackelenbergh M, Schem C, Staib P, Link T, Müller V, Stickeler E, Stover DG, Solbach C, Metzger-Filho O, Jackisch C, Geyer CE Jr, Fasching PA, Pusztai L, Nekljudova V, Untch M, and Loibl S
- Subjects
- Humans, Female, Gene Expression Regulation, Neoplastic drug effects, Antibodies, Monoclonal therapeutic use, Prognosis, Triple Negative Breast Neoplasms immunology, Triple Negative Breast Neoplasms therapy, Triple Negative Breast Neoplasms pathology, Triple Negative Breast Neoplasms drug therapy, Neoadjuvant Therapy methods, Immunotherapy methods, Tumor Microenvironment immunology, Tumor Microenvironment drug effects
- Abstract
Therapy-induced molecular adaptation of triple-negative breast cancer is crucial for immunotherapy response and resistance. We analyze tumor biopsies from three different time points in the randomized neoadjuvant GeparNuevo trial (NCT02685059), evaluating the combination of durvalumab with chemotherapy, for longitudinal alterations of gene expression. Durvalumab induces an activation of immune and stromal gene expression as well as a reduction of proliferation-related gene expression. Immune genes are positive prognostic factors irrespective of treatment, while proliferation genes are positive prognostic factors only in the durvalumab arm. We identify stromal-related gene expression as a contributor to immunotherapy resistance and poor therapy response. The results provide evidence from clinical trial cohorts suggesting a role for stromal reorganization in therapy resistance to immunotherapy and in the generation of an immune-suppressive microenvironment, which might be relevant for future therapy approaches targeting the tumor stroma parallel to immunotherapy, such as combinations of immunotherapy with anti-angiogenic therapy., Competing Interests: Declaration of interests C.D. reports grants from European Commission H2020, grants from German Cancer Aid Translational Oncology, grants from German Breast Group, and grants from BMBF to the institution during the conduct of the study; personal fees from Novartis, personal fees from Roche, personal fees from MSD Oncology, personal fees from Daiichi Sankyo, personal fees from AstraZeneca and Molecular Health, grants from Myriad, personal fees from Merck, and other funding from Sividon Diagnostics outside the submitted work; in addition, C.D. has a patent VMScope digital pathology software with royalties paid, a patent WO2020109570A1—cancer immunotherapy pending, and a patent WO2015114146A1 and WO2010076322A1—therapy response issued. J.R. declares to be a GBG Forschungs GmbH employee. GBG Forschungs GmbH received funding for research grants from AbbVie, Amgen, AstraZeneca, BMS, Daiichi Sankyo, Gilead, Molecular Health, Novartis, Pfizer, and Roche (paid to the institution). Funding was also received (non-financial/medical writing) from Daiichi Sankyo, Gilead, Novartis, Pfizer, Roche, and Seagen (paid to the institution). GBG Forschungs GmbH has royalties in VM Scope and patents pending: EP14153692.0, EP21152186.9, and EP15702464.7. T.K. reports a patent WO2020109570A1 pending. S.R. declares to be a GBG Forschungs GmbH employee. GBG Forschungs GmbH received funding for research grants from AbbVie, Amgen, AstraZeneca, BMS, Daiichi Sankyo, Gilead, Molecular Health, Novartis, Pfizer, and Roche (paid to the institution). J.H. reports research funding from Lilly; honoraria from Lilly, Novartis, Roche, Pfizer, AstraZeneca, Seagen, Gilead, and Daiichi; consulting and advisory relationships with Lilly, Novartis, Roche, Pfizer, AstraZeneca, Gilead, and Daiichi; travel expenses from Roche, Novartis, Daiichi, and Gilead. P.J. reports research funding and travel expenses from Gilead Sciences GmbH. C.H. reports an advisory role and speakers bureau role for AstraZeneca, Roche, Novartis, and Aristo Pharma. B.V.S. is an employee of BioNTech SE and reports a patent WO2020109570A1 pending. J.-U.B. reports consultation fees, honoraria, and reimbursement for attending symposia from AstraZeneca, Amgen, Daiichi Sankyo, Eisai, Gilead, Lilly, MSD, Novartis, Pfizer, Roche, and Seagen. M.v.M. reports personal fees, honoraria, or travel grants from Amgen, AstraZeneca, Daiichi Sankyo, Genomic Health, GSK, Lilly, Molecular Health, MSD, Mylan, Novartis, Pfizer, Pierre Fabre, Roche, and Seagen. C. Schem reports speaker activities for Roche, Pfizer, Novartis, Celgen, Novartis, Exact Sciences, MSD, AstraZeneca, Lilly, and Seagen, as well as advisory boards for Roche, Astra Zeneca, Pfizer, Novartis, MSD, Amgen, Exact Sciences, Stemline, Lilly, and Novartis. T.L. reports personal fees from Amgen, Roche, Teva, Clovis, Tesaro, MSD, Novartis, Pfizer, Lilly, GSK, Gilead, AstraZeneca, Daiichi Sankyo, Stemline, and Seagen outside of the submitted work. T.L. participates in advisory boards from Amgen, MSD, Tesaro, Roche, Pfizer, Lilly, Myriad, Esai, GSK, Gilead, Daiichi Sankyo, Roche, and AstraZeneca outside of the submitted work and T.L. received travel support from Pfizer, PharmaMar, MSD, Celgene, Roche, AstraZeneca, Gilead, Daiichi Sankyo, Stemline, and Clovis outside of the submitted work. P.S. reports grants, personal fees, and non-financial support from AbbVie, Amgen, AstraZeneca, Bristol Myers Squibb Company, MSD, Incyte, Janssen-Cilag, Novartis, Takeda, Pfizer, and Roche. V.M. received speaker honoraria from AstraZeneca, Daiichi Sankyo, Eisai, GSK, Pfizer, MSD, Medac, Novartis, Roche, Seagen, Onkowissen, high5 Oncology, Medscape, Gilead, and Pierre Fabre; consultancy honoraria from Roche, Pierre Fabre, Amgen, ClinSol, Novartis, MSD, Daiichi Sankyo, Eisai, Lilly, Sanofi, Seagen, Gilead, and Stemline; institutional research support from Novartis, Roche, Seagen, and Genentech; and travel grants from Roche, Pfizer, Daiichi Sankyo, and Gilead. L.P. has received consulting fees and honoraria for advisory board participation from Pfizer, AstraZeneca, Merck, Novartis, Bristol Myers Squibb, GlaxoSmithKline, Genentech/Roche, Personalis, Daiichi, Natera, and Exact Sciences and institutional research funding from Seagen, GlaxoSmithKline, AstraZeneca, Merck, Pfizer, and Bristol Myers Squibb. C.E.G. reports the following competing interests: Exact Sciences, Advisory Board, personal; AbbVie, Steering Committee member, institutional, co-chair of SC for BrighTNess; Daiichi Sankyo, member SC, institutional, co-chair of SC for DESTINY-Breast05; Genentech/Roche, SC member, institutional, co-chair of SC for lidERA; Genentech/Roche, coordinating PI, institutional, NSABP B-59/GeparDouze; and Genentech/Roche, SC member, institutional, co-chair of SC for KATHERINE. C.J. reports honoraria from AstraZeneca, Amgen, Daiichi Sankyo, Lilly, Roche, Pfizer, MSD Oncology, Pierre Fabre, Sanofi-Aventis, Seagen, Gilead, and Novartis and has a consulting or advisory role for Amgen, Lilly, Roche, Pfizer, Pierre Fabre, Novartis, MSD Oncology, Agendia, Seagen, Gilead, Lilly, Stemline, and Medac. V.N. declares to be a GBG Forschungs GmbH employee. GBG Forschungs GmbH received funding for research grants from AbbVie, AstraZeneca, BMS, Daiichi Sankyo, Gilead, Novartis, Pfizer, and Roche (paid to the institution). GBG Forschungs GmbH received other funding from Daiichi Sankyo, Gilead, Novartis, Pfizer, Roche, and Seagen (paid to the institution). GBG Forschungs GmbH has the following royalties/patents: EP14153692.0, EP21152186.9, EP15702464.7, EP19808852.8, and VM Scope GmbH. M.U. reports honoraria from AstraZeneca, Amgen, Daiichi Sankyo, Lilly, Roche, Pfizer, MSD Oncology, Pierre Fabre, Sanofi-Aventis, Myriad, Seagen, Gilead, and Novartis; has a consulting or advisory role for Amgen, Lilly, Roche, Pfizer, Pierre Fabre, Novartis, MSD Oncology, Agendia, Seagen, Gilead, Lily, Stemline, Genzyme, and Medac; and all honoraria and fees are paid to the employer/institution. S.L. reports grants and other funding from AbbVie; other funding from Amgen; grants and other funding from AstraZeneca; other funding from BMS; grants and other funding from Celgene; grants, non-financial support, and other funding from Daiichi Sankyo; other funding from EirGenix; other funding from Eisai Europe Ltd; other funding from GSK; grants, non-financial support, and other funding from Immunomedics/Gilead; other funding from Lilly; other funding from Merck; grants from Molecular Health; grants, non-financial support, and other funding from Novartis; grants, non-financial support, and other funding from Pfizer; other funding from Pierre Fabre; other funding from Relay Therapeutics; grants, non-financial support, and other funding from Roche; other funding from Sanofi; non-financial support and other funding from Seagen; and other funding from Olema Pharmaceuticals, outside the submitted work. In addition, S.L. has a patent EP14153692.0 pending, a patent EP21152186.9 pending, a patent EP15702464.7 issued, a patent EP19808852.8 pending, and a patent Digital Ki67 Evaluator with royalties paid., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
122. Comparative analysis of Ficoll-Hypaque and CytoLyt techniques for blood removal in breast cancer malignant effusions: effects on RNA quality and sequencing outcomes.
- Author
-
Sura GH, Tran K, Trevarton AJ, Marczyk M, Fu C, Du L, Qu J, Lau R, Tasto A, Gould RE, Tinnirello A, Sinn BV, Pusztai L, Hatzis C, and Symmans WF
- Abstract
Introduction: To optimize RNA sequencing (RNA-seq) outcomes, we investigated preanalytical variables in malignant effusions containing metastatic breast cancer. We compared 2 processing methods-Ficoll-Hypaque density gradient enrichment and CytoLyt hemolysis-focusing on their effects on RNA quality, transcript abundance, and variant detection from cytospin slides, relative to fresh-frozen samples. Additionally, we compared read-based and Unique Molecular Identifier (UMI)-based library preparation methods., Materials and Methods: Thirteen malignant effusion specimens from metastatic breast cancer were processed using both the Ficoll-Hypaque and Cytolyt methods. RNA was extracted from fresh-frozen samples stored in RNA preservative and from cytospin slides fixed in Carnoy's solution. RNA quality was evaluated using RNA integrity number (RIN) and the percentage of fragments >200 bases (DV200). Sequencing was conducted with both read- and UMI-based methods., Results: Purified RNA was more fragmented by the Cytolyt method (mean RIN: 3.56, DV200: 78.97%), compared to the Ficoll-Hypaque method (mean RIN: 6.29, DV200: 88.08%). Sequencing data had high concordance correlation coefficient (CCC) for measurements of gene expression, whether from Cytolyt or Ficoll-Hypaque treated samples, and whether using the UMI- or read-based sequencing methods (read-based mean CCC: 0.967 from Cytolyt versus 0.974 from Ficoll-Hypaque, UMI-based mean CCC: 0.972 from Cytolyt versus 0.977 from Ficoll-Hypaque)., Conclusions: Despite the increased RNA fragmentation with the Cytolyt, RNA-seq data quality was comparable across Cytolyt and Ficoll-Hypaque methods. Both clearing methods are viable for short-read RNA-seq analysis, with read and UMI-based approaches performing similarly., (Copyright © 2024 American Society of Cytopathology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
123. Clinical Management of Ovarian Function Suppression in Premenopausal Women With Breast Cancer: A Survey of Members of ASCO.
- Author
-
Kelly CM, Bennett KE, Cahir C, Eisen A, and Pusztai L
- Abstract
Purpose: Ovarian function suppression (OFS) with gonadotropin-releasing hormone agonists (GnRHas) is a standard of care for premenopausal patients with high-risk stage II/III hormone receptor-positive breast cancer (BC). Practical guidance on the optimal choice of GnRHa, timing, schedule, and monitoring is limited. Our aim was to determine how oncologists use OFS in routine care., Methods: We designed a questionnaire to determine the choice of GnRHa, schedule, duration, initiation, use of bone modifiers, and monitoring of estradiol (E2). The questionnaire was sent to oncologists treating BC, in practice for >1 year and participating in the ASCO Research Survey Pool (RSP). It was also forwarded by investigators to oncologists meeting these criteria. The survey was open between November 14, 2023, and January 5, 2024., Results: Of 996 oncologists participating in the ASCO RSP, 178 (18%) completed the survey. An additional 56 oncologists contacted by investigators responded. Respondents were from the United States (57%), Asia (15%), and Europe (14%). Goserelin (54%) and leuprolide (39%) were the most frequently used GnRHas and were administered once every month by 46%. Approaches to starting GnRHas were varied. Most continued them for the duration of aromatase inhibitor therapy (57%). Estradiol monitoring was performed regularly, sometimes, or never by 43%, 27%, and 27%, respectively. The E2 assays used were standard (65%), ultrasensitive (16%), and unknown (14%). Interpreting E2 assay results were considered difficult by 55%; however, 62% of oncologists changed treatment on the basis of them. A total of 92% of respondents would like ASCO guidance on the practical use of OFS., Conclusion: Considerable practice variation exists for similar clinical scenarios in OFS administration. Respondents would welcome ASCO guidance on all aspects of OFS.
- Published
- 2024
- Full Text
- View/download PDF
124. Peripheral blood immune parameters, response, and adverse events after neoadjuvant chemotherapy plus durvalumab in early-stage triple-negative breast cancer.
- Author
-
Foldi J, Blenman KRM, Marczyk M, Gunasekharan V, Polanska A, Gee R, Davis M, Kahn AM, Silber A, and Pusztai L
- Subjects
- Humans, Female, Middle Aged, Adult, Aged, Receptors, Antigen, T-Cell genetics, Receptors, Antigen, T-Cell metabolism, Neoplasm Staging, Receptors, Antigen, B-Cell metabolism, Treatment Outcome, Neoadjuvant Therapy adverse effects, Neoadjuvant Therapy methods, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms blood, Triple Negative Breast Neoplasms immunology, Triple Negative Breast Neoplasms pathology, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Antibodies, Monoclonal adverse effects, Antibodies, Monoclonal therapeutic use, Antibodies, Monoclonal administration & dosage, Cytokines blood
- Abstract
Purpose: We evaluated T- and B-cell receptor (TCR and BCR) repertoire diversity and 38 serum cytokines in pre- and post-treatment peripheral blood of 66 patients with triple-negative breast cancer (TNBC) who received neoadjuvant chemotherapy plus durvalumab and assessed associations with pathologic response and immune-related adverse events (irAEs) during treatment., Methods: Genomic DNA was isolated from buffy coat for TCR and BCR clonotype profiling using the Immunoseq platform and diversity was quantified with Pielou's evenness index. MILLIPLEX MAP Human Cytokine/Chemokine Magnetic Bead Panel was used to measure serum cytokine levels, which were compared between groups using moderated t-statistic with Benjamini-Hochberg correction for multiple testing., Results: TCR and BCR diversity was high (Pielou's index > 0.75) in all samples. Baseline receptor diversities and change in diversity pre- and post-treatment were not associated with pathologic response or irAE status, except for BCR diversity that was significantly lower post-treatment in patients who developed irAE (unadjusted p = 0.0321). Five cytokines increased after treatment in patients with pathologic complete response (pCR) but decreased in patients with RD, most prominently IL-8. IFNγ, IL-7, and GM-CSF levels were higher in pre-treatment than in post-treatment samples of patients who developed irAEs but were lower in those without irAEs., Conclusion: Baseline peripheral blood cytokine levels may predict irAEs in patients treated with immune checkpoint inhibitors and chemotherapy, and increased post-treatment B-cell clonal expansion might mediate irAEs., (© 2024. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
125. The Immune-Related 27-Gene Signature DetermaIO Predicts Response to Neoadjuvant Atezolizumab plus Chemotherapy in Triple-Negative Breast Cancer.
- Author
-
Dugo M, Huang CS, Egle D, Bermejo B, Zamagni C, Seitz RS, Nielsen TJ, Thill M, Antón-Torres A, Russo S, Ciruelos EM, Schweitzer BL, Ross DT, Galbardi B, Greil R, Semiglazov V, Gyorffy B, Colleoni M, Kelly CM, Mariani G, Del Mastro L, Blasi O, Callari M, Pusztai L, Valagussa P, Viale G, Gianni L, and Bianchini G
- Subjects
- Humans, Female, Middle Aged, Biomarkers, Tumor genetics, Carboplatin administration & dosage, Carboplatin therapeutic use, Transcriptome, Aged, Adult, Prognosis, Treatment Outcome, Gene Expression Regulation, Neoplastic, Gene Expression Profiling, Albumins, Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized therapeutic use, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms pathology, Neoadjuvant Therapy methods, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Paclitaxel administration & dosage, Paclitaxel therapeutic use
- Abstract
Purpose: We assessed the 27-gene RT-qPCR-based DetermaIO assay and the same score calculated from RNA sequencing (RNA-seq) data as predictors of sensitivity to immune checkpoint therapy in the neoTRIPaPDL1 randomized trial that compared neoadjuvant carboplatin/nab-paclitaxel chemotherapy (CT) plus atezolizumab with CT alone in stage II/III triple-negative breast cancer. We also assessed the predictive function of the immuno-oncology (IO) score in expression data of patients treated with pembrolizumab plus paclitaxel (N = 29) or CT alone (N = 56) in the I-SPY2 trial., Experimental Design: RNA-seq data were obtained from pretreatment core biopsies from 242 (93.8%) of the 258 patients in the per-protocol-population. The DetermaIO RT-qPCR test, performed in the CAP/CLIA-accredited laboratory of Oncocyte Corp., was available for 220 patients (85.3%). A previously established threshold was used to assign DetermaIO-positive versus DetermaIO-negative status. Publicly available microarray data were used from I-SPY2., Results: IO scores calculated from RNA-seq and RT-qPCR data were highly concordant. In neoTRIPaPDL1, DetermaIO-positive cancers (N = 92, 41.8%) had pathologic complete response (pCR) rates of 69.8% and 46.9% in the CT + atezolizumab and CT arms, respectively. In DetermaIO-negative cases, pCR rates were similar in both arms (44.6% vs. 49.2%; interaction test P = 0.04). PDL1 protein expression and stromal tumor-infiltrating lymphocyte count were not predictive of differential benefit from atezolizumab. In I-SPY2, IO-positive cancers (45.9%) had pCR rates of 85.7% and 16%, with and without immunotherapy, respectively. In IO-negative cancers, pCR rates were 46.7% versus 16.1%., Conclusions: DetermaIO identified patients who benefited from neoadjuvant immunotherapy resulting in improved pCR rate, independently of PDL1., (©2024 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
126. Academic Uphill Battle to Personalize Treatment for Patients With Stage II/III Triple-Negative Breast Cancer.
- Author
-
Kok M, Gielen RJ, Adams S, Lennerz JK, Sharma P, Loibl S, Reardon E, Sonke G, Linn S, Delaloge S, Lacombe D, Robinson T, Badve S, Martin M, Balko JM, Ignatiadis M, Curigliano G, Wolff AC, Mittendorf EA, Loi S, Pusztai L, Tolaney SM, and Salgado R
- Published
- 2024
- Full Text
- View/download PDF
127. Neoadjuvant pembrolizumab plus chemotherapy/adjuvant pembrolizumab for early-stage triple-negative breast cancer: quality-of-life results from the randomized KEYNOTE-522 study.
- Author
-
Dent R, Cortés J, Pusztai L, McArthur H, Kümmel S, Bergh J, Denkert C, Park YH, Hui R, Harbeck N, Takahashi M, Untch M, Fasching PA, Cardoso F, Haiderali A, Jia L, Nguyen AM, Pan W, O'Shaughnessy J, and Schmid P
- Subjects
- Humans, Female, Middle Aged, Chemotherapy, Adjuvant, Aged, Adult, Paclitaxel administration & dosage, Cyclophosphamide administration & dosage, Carboplatin administration & dosage, Neoplasm Staging, Patient Reported Outcome Measures, Epirubicin administration & dosage, Doxorubicin administration & dosage, Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized therapeutic use, Quality of Life, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms pathology, Neoadjuvant Therapy methods, Antineoplastic Combined Chemotherapy Protocols therapeutic use
- Abstract
Background: In KEYNOTE-522 (NCT03036488), neoadjuvant pembrolizumab plus chemotherapy and then adjuvant pembrolizumab significantly improved pathological complete response and event-free survival vs neoadjuvant chemotherapy in early-stage triple-negative breast cancer (TNBC). We report patient-reported outcomes (PROs) from KEYNOTE-522., Methods: Patients were randomized 2:1 to neoadjuvant pembrolizumab 200 mg or placebo every 3 weeks, plus 4 cycles of paclitaxel plus carboplatin and then 4 cycles of doxorubicin (or epirubicin) plus cyclophosphamide. After surgery, patients received adjuvant pembrolizumab or placebo for up to 9 cycles. European Organisation for Research and Treatment of Cancer Quality of Life Questionnaire-Core 30 (EORTC QLQ-C30) and EORTC Breast Cancer-Specific Quality of Life Questionnaire (EORTC QLQ-BR23) were prespecified secondary objectives. Between-group differences in least squares (LS) mean change from baseline (day 1 of cycle 1 in both neoadjuvant and adjuvant phases) to the prespecified latest time point with at least 60% completion and at least 80% compliance were assessed using a longitudinal model (no alpha error assigned)., Results: Week 21 (neoadjuvant phase) and week 24 (adjuvant phase) were the latest time points at which completion/compliance rates were ≥60%/80%. In the neoadjuvant phase, between-group differences (pembrolizumab plus chemotherapy [n = 762] vs placebo plus chemotherapy [n = 383]) in LS mean change from baseline to week 21 in QLQ-C30 global health status/quality of life (GHS/QoL), emotional functioning, and physical functioning were -1.04 (95% confidence interval = -3.46 to 1.38), -0.69 (95% CI = -3.13 to 1.75), and -2.85 (95% CI = -5.11 to -0.60), respectively. In the adjuvant phase, between-group differences (pembrolizumab [n = 539] vs placebo [n = 308]) in LS mean change from baseline to week 24 were -0.41 (95% CI = -2.60 to 1.77), -0.60 (95% CI = -2.99 to 1.79), and -1.57 (95% CI = -3.36 to 0.21)., Conclusions: No substantial differences in PRO assessments were observed between neoadjuvant pembrolizumab plus chemotherapy followed by adjuvant pembrolizumab vs neoadjuvant placebo plus chemotherapy in early-stage TNBC., Trial Registration: ClinicalTrials.gov, NCT03036488., (© The Author(s) 2024. Published by Oxford University Press.)
- Published
- 2024
- Full Text
- View/download PDF
128. Trends in breast cancer specific death by clinical stage at diagnoses between 2000-2017.
- Author
-
Marczyk M, Kahn A, Silber A, Rosenblit M, Digiovanna MP, Lustberg M, and Pusztai L
- Abstract
Background: Approximately 40,000 individuals die from metastatic breast cancer each year. We examined what fractions of annual breast cancer-specific death (BCSD) are due to stage I, II, III, IV disease and if these proportions changed over time., Methods: We used data from SEER covering 1975 to 2017. After filtering for female sex at birth, one primary tumor type, surgery, AJCC (6th edition) stage > 0, no bilateral cancer, and survival data available, the final analysis included 972,763 patients. Temporal trends were assessed using a linear model and ANOVA test., Results: The contribution of stage I and II cancers to BCSD increased significantly from 16.2% to 23.1%, and from 30.7% to 39.5%, respectively between 2000 and 2017. The contribution of stages III and IV cancers decreased from 36.4% to 30.3%, and from 16.7% to 7.1%. In 2000, 0.92%, 4.0% and 10.7% of BCSD were due to T1a, T1b, and T1c node-negative cancers which increased significantly to 1.9%, 5.8%, and 14.7% by 2017. These temporal trends were similar for hormone receptor-positive and -negative cancers. The contribution of BCSD to all-cause mortality declined from 23.9% to 16.6% for stage I, and from 47.7% to 36.9% for stage II cancers by 2017., Conclusions: Patients with stage I/II breast cancers have excellent prognosis, yet these cancers account for over 60% of current BCSD because of their large absolute numbers. To further reduce breast cancer death strategies are needed to identify and treat patients with stage I/II disease who remain at risk for recurrence., (© The Author(s) 2024. Published by Oxford University Press. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2024
- Full Text
- View/download PDF
129. Phase III Randomized, Placebo-Controlled Trial of Endocrine Therapy ± 1 Year of Everolimus in Patients With High-Risk, Hormone Receptor-Positive, Early-Stage Breast Cancer.
- Author
-
Chavez-MacGregor M, Miao J, Pusztai L, Goetz MP, Rastogi P, Ganz PA, Mamounas EP, Paik S, Bandos H, Razaq W, O'Dea A, Kaklamani V, Silber ALM, Flaum LE, Andreopoulou E, Wendt AG, Carney JF, Sharma P, Gralow JR, Lew DL, Barlow WE, and Hortobagyi GN
- Subjects
- Humans, Female, Middle Aged, Aged, Adult, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Antineoplastic Combined Chemotherapy Protocols adverse effects, Receptors, Estrogen metabolism, Receptors, Estrogen analysis, Receptor, ErbB-2 metabolism, Receptors, Progesterone metabolism, Neoplasm Staging, Chemotherapy, Adjuvant, Antineoplastic Agents, Hormonal therapeutic use, Antineoplastic Agents, Hormonal adverse effects, Everolimus therapeutic use, Everolimus adverse effects, Everolimus administration & dosage, Breast Neoplasms drug therapy, Breast Neoplasms pathology, Breast Neoplasms mortality
- Abstract
Purpose: Phosphatidylinositol 3-kinase/AKT-serine threonine kinase/mammalian target of rapamycin (mTOR) pathway abnormalities contribute to endocrine resistance. Everolimus, an mTOR inhibitor, improved progression-free survival in hormone receptor-positive metastatic breast cancer (BC) when combined with endocrine therapy (ET). In this phase III randomized, placebo-controlled trial, we assessed the efficacy of everolimus + ET as adjuvant therapy in high-risk, hormone receptor-positive, human epidermal growth factor receptor 2-negative BC after adjuvant/neoadjuvant chemotherapy., Methods: Patients were randomly assigned 1:1 to physician's choice ET and 1 year of everolimus (10 mg orally once daily) or placebo stratified by risk group. The primary end point was invasive disease-free survival (IDFS) evaluated by a stratified log-rank test with the hazard ratio (HR) estimated by Cox regression. Subset analyses included preplanned evaluation by risk group and exploratory analyses by menopausal status and age. Secondary end points included overall survival (OS) and safety. Everolimus did not improve IDFS/OS when added to ET in patients with early-stage high-risk, hormone receptor-positive BC., Results: One thousand and nine hundred thirty-nine patients were randomly assigned with 1,792 eligible for analysis. Overall, no benefit of everolimus was seen for IDFS (HR, 0.94 [95% CI, 0.77 to 1.14]) or OS (HR, 0.97 [95% CI, 0.75 to 1.26]). The assumption of proportional hazards was not met suggesting significant variability in the HR over time since the start of treatment. In an unplanned subgroup analysis among postmenopausal patients (N = 1,221), no difference in IDFS (HR, 1.08 [95% CI, 0.86 to 1.36]) or OS (HR, 1.19 [95% CI, 0.89 to 1.60]) was seen. In premenopausal patients (N = 571), everolimus improved both IDFS (HR, 0.64 [95% CI, 0.44 to 0.94]) and OS (HR, 0.49 [95% CI, 0.28 to 0.86]). Treatment completion rates were lower in the everolimus arm compared with placebo (48% v 73%) with higher grade 3 and 4 adverse events (35% v 7%)., Conclusion: One year of adjuvant everolimus + ET did not improve overall outcomes. Subset analysis suggests mTOR inhibition as a possible target for patients who remain premenopausal after chemotherapy.
- Published
- 2024
- Full Text
- View/download PDF
130. Associations amongst genes, molecules, cells, and organs in breast cancer metastasis.
- Author
-
Nathanson SD, Dieterich LC, Zhang XH, Chitale DA, Pusztai L, Reynaud E, Wu YH, and Ríos-Hoyo A
- Subjects
- Humans, Female, Bone Neoplasms secondary, Bone Neoplasms genetics, Neoplasm Metastasis, Lymphatic Metastasis pathology, Endothelial Cells pathology, Breast Neoplasms pathology, Breast Neoplasms genetics
- Abstract
This paper is a cross fertilization of ideas about the importance of molecular aspects of breast cancer metastasis by basic scientists, a pathologist, and clinical oncologists at the Henry Ford Health symposium. We address four major topics: (i) the complex roles of lymphatic endothelial cells and the molecules that stimulate them to enhance lymph node and systemic metastasis and influence the anti-tumor immunity that might inhibit metastasis; (ii) the interaction of molecules and cells when breast cancer spreads to bone, and how bone metastases may themselves spread to internal viscera; (iii) how molecular expression and morphologic subtypes of breast cancer assist clinicians in determining which patients to treat with more or less aggressive therapies; (iv) how the outcomes of patients with oligometastases in breast cancer are different from those with multiple metastases and how that could justify the aggressive treatment of these patients with the hope of cure., (© 2023. The Author(s), under exclusive licence to Springer Nature B.V.)
- Published
- 2024
- Full Text
- View/download PDF
131. Neoadjuvant Chemotherapy and Immunotherapy for Estrogen Receptor-Positive Human Epidermal Growth Factor 2-Negative Breast Cancer.
- Author
-
Ríos-Hoyo A, Cobain E, Huppert LA, Beitsch PD, Buchholz TA, Esserman L, van 't Veer LJ, Rugo HS, and Pusztai L
- Subjects
- Humans, Female, Immunotherapy methods, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Chemotherapy, Adjuvant, Breast Neoplasms drug therapy, Breast Neoplasms immunology, Breast Neoplasms therapy, Neoadjuvant Therapy, Receptor, ErbB-2 metabolism, Receptors, Estrogen metabolism
- Published
- 2024
- Full Text
- View/download PDF
132. Pre-treatment amino acids and risk of paclitaxel-induced peripheral neuropathy in SWOG S0221.
- Author
-
Chen CS, Zirpoli G, Budd GT, Barlow WE, Pusztai L, Hortobagyi GN, Albain KS, Godwin AK, Thompson A, Henry NL, Ambrosone CB, Stringer KA, and Hertz DL
- Subjects
- Adult, Aged, Female, Humans, Middle Aged, Antineoplastic Agents, Phytogenic adverse effects, Antineoplastic Agents, Phytogenic administration & dosage, Severity of Illness Index, Amino Acids blood, Breast Neoplasms drug therapy, Paclitaxel adverse effects, Paclitaxel administration & dosage, Peripheral Nervous System Diseases chemically induced
- Abstract
Background: Chemotherapy-induced peripheral neuropathy (CIPN) is a treatment-limiting and debilitating neurotoxicity of many commonly used anti-cancer agents, including paclitaxel. The objective of this study was to confirm the previously found inverse association between pre-treatment blood concentrations of histidine and CIPN occurrence and examine relationships of other amino acids with CIPN severity., Methods: Pre-treatment serum concentrations of 20 amino acids were measured in the SWOG S0221 (NCT00070564) trial of patients with early-stage breast cancer receiving paclitaxel. The associations between amino acids and CIPN severity were tested in regression analysis adjusted for paclitaxel schedule, age, self-reported race, and body mass index with Bonferroni correction. The network of metabolic pathways of amino acids was analyzed using over-representation analysis. The partial correlation network of amino acids was evaluated using a debiased sparse partial correlation algorithm., Results: In the primary analysis, histidine concentration was not associated with CIPN occurrence (odds ratio (OR) = 0.97 [0.83, 1.13], p = 0.72). In secondary analyses, higher concentrations of four amino acids, glutamate (β = 0.58 [0.23, 0.93], p = 0.001), phenylalanine (β = 0.54 [0.19, 0.89], p = 0.002), tyrosine (β = 0.57 [0.23, 0.91], p = 0.001), and valine (β = 0.58 [0.24, 0.92], p = 0.001) were associated with more severe CIPN, but none of these associations retained significance after adjustment. In the over-representation analysis, no amino acid metabolic pathways were significantly enriched (all FDR > 0.05). In the network of enriched pathways, glutamate metabolism had the highest centrality., Conclusions: This analysis showed that pre-treatment serum amino acid concentrations are not strongly predictive of CIPN severity. Prospectively designed studies that assess non-amino acid metabolomics predictors are encouraged., (© 2024. The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
133. Completion Rate and Positive Results Reporting Among Immunotherapy Trials in Breast Cancer, 2004-2023.
- Author
-
Mariani M, Viale G, Galbardi B, Licata L, Bosi C, Dugo M, Notini G, Naldini MM, Callari M, Criscitiello C, Pusztai L, and Bianchini G
- Subjects
- Humans, Female, Cross-Sectional Studies, Breast Neoplasms therapy, Breast Neoplasms drug therapy, Immunotherapy methods, Immunotherapy statistics & numerical data, Clinical Trials as Topic
- Abstract
Importance: Clinical trials are the path to test and introduce new therapies in the clinic. Trials that are unable to produce results represent inefficiency in the system and may also undermine patient confidence in the new drug development process., Objectives: To survey the immunotherapy clinical trial landscape of breast cancer between January 2004 and April 2023 and examine what fraction of trials with primary completion date up to November 30, 2022, failed to report outcome, assessing the proportion of trials that yielded positive results and describing trial features associated with these 2 outcomes., Design, Setting, and Participants: This cross-sectional study included breast cancer immunotherapy trials identified in ClinicalTrials.gov. Trial details and results were retrieved in December 2023. Google Scholar, PubMed, and LARVOL CLIN websites were also searched for reports., Main Outcomes and Measures: Trial outcome reported as abstract or manuscript. Reported trials were categorized as positive (ie, met its end point) or negative. Association between reporting and trial features were tested using Fisher exact test., Results: A total of 331 immuno-oncology trials were initiated in breast cancer by April 2023; 242 trials were phase II, 47 were phase I, and 42 phase III. By setting, 212 studies (64.0%) were conducted in metastatic, 94 (28.4%) in neoadjuvant, and 25 (7.6%) in adjuvant settings. Among phase II and III trials, 168 (59.2%) were nonrandomized. One hundred twenty trials had primary completion dates up to November 30, 2022, of which 30 (25.0%; enrolling a combined 2428 patients) failed to report their outcomes; 7 phase I trials (31.8%), 21 phase II trials (23.6%), and 2 phase III trials (22.2%) were unreported. Single-center studies were significantly more likely to be unreported than multicenter studies (19 of 54 [35.2%] vs 9 of 60 [15.0%]; P = .02). Of the 90 reported trials, 47 (52.2%) and 43 (47.8%) were positive and negative, respectively. Seventeen of 19 (89.5%) of the reported randomized trials (accruing a total of 4189 patients) were negative., Conclusions and Relevance: In this cross-sectional study of immunotherapy breast cancer trials, the large number of trials yielded modest clinical impact. Single-center trials commonly failed to report their outcomes and many phase II studies have not translated into corresponding successful phase III trials.
- Published
- 2024
- Full Text
- View/download PDF
134. Navigating practical challenges in immunotherapy for metastatic triple negative breast cancer.
- Author
-
Licata L, Dieci MV, De Angelis C, Marchiò C, Miglietta F, Cortesi L, Fabi A, Schmid P, Cortes J, Pusztai L, Bianchini G, and Curigliano G
- Subjects
- Humans, Female, Triple Negative Breast Neoplasms therapy, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms immunology, Immunotherapy methods, Immune Checkpoint Inhibitors therapeutic use
- Abstract
Immunotherapy has revolutionized cancer therapy and now represents a standard of care for many tumor types, including triple-negative breast cancer. Despite the positive results that have led to the approval of immunotherapy in both early- and advanced-stage triple-negative breast cancer, pivotal clinical trials cannot address the myriad questions arising in everyday clinical practice, often falling short in delivering all the information that clinicians require. In this manuscript, we aim to address some of these practical questions, with the purpose of providing clinicians with a guide for optimizing the use of immune checkpoint inhibitors in the management of breast cancer patients and identifying opportunities for future research to clarify unresolved questions., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
135. Evaluation of large language models as a diagnostic aid for complex medical cases.
- Author
-
Ríos-Hoyo A, Shan NL, Li A, Pearson AT, Pusztai L, and Howard FM
- Abstract
Background: The use of large language models (LLM) has recently gained popularity in diverse areas, including answering questions posted by patients as well as medical professionals., Objective: To evaluate the performance and limitations of LLMs in providing the correct diagnosis for a complex clinical case., Design: Seventy-five consecutive clinical cases were selected from the Massachusetts General Hospital Case Records, and differential diagnoses were generated by OpenAI's GPT3.5 and 4 models., Results: The mean number of diagnoses provided by the Massachusetts General Hospital case discussants was 16.77, by GPT3.5 30 and by GPT4 15.45 ( p < 0.0001). GPT4 was more frequently able to list the correct diagnosis as first (22% versus 20% with GPT3.5, p = 0.86), provide the correct diagnosis among the top three generated diagnoses (42% versus 24%, p = 0.075). GPT4 was better at providing the correct diagnosis, when the different diagnoses were classified into groups according to the medical specialty and include the correct diagnosis at any point in the differential list (68% versus 48%, p = 0.0063). GPT4 provided a differential list that was more similar to the list provided by the case discussants than GPT3.5 (Jaccard Similarity Index 0.22 versus 0.12, p = 0.001). Inclusion of the correct diagnosis in the generated differential was correlated with PubMed articles matching the diagnosis (OR 1.40, 95% CI 1.25-1.56 for GPT3.5, OR 1.25, 95% CI 1.13-1.40 for GPT4), but not with disease incidence., Conclusions and Relevance: The GPT4 model was able to generate a differential diagnosis list with the correct diagnosis in approximately two thirds of cases, but the most likely diagnosis was often incorrect for both models. In its current state, this tool can at most be used as an aid to expand on potential diagnostic considerations for a case, and future LLMs should be trained which account for the discrepancy between disease incidence and availability in the literature., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 Ríos-Hoyo, Shan, Li, Pearson, Pusztai and Howard.)
- Published
- 2024
- Full Text
- View/download PDF
136. Event-free survival by residual cancer burden with pembrolizumab in early-stage TNBC: exploratory analysis from KEYNOTE-522.
- Author
-
Pusztai L, Denkert C, O'Shaughnessy J, Cortes J, Dent R, McArthur H, Kümmel S, Bergh J, Park YH, Hui R, Harbeck N, Takahashi M, Untch M, Fasching PA, Cardoso F, Zhu Y, Pan W, Tryfonidis K, and Schmid P
- Subjects
- Humans, Female, Middle Aged, Carboplatin administration & dosage, Neoadjuvant Therapy methods, Neoplasm Staging, Cyclophosphamide administration & dosage, Cyclophosphamide therapeutic use, Cyclophosphamide adverse effects, Aged, Adult, Doxorubicin therapeutic use, Doxorubicin administration & dosage, Epirubicin administration & dosage, Epirubicin therapeutic use, Progression-Free Survival, Chemotherapy, Adjuvant methods, Antineoplastic Agents, Immunological therapeutic use, Antineoplastic Agents, Immunological adverse effects, Antineoplastic Agents, Immunological administration & dosage, Double-Blind Method, Antibodies, Monoclonal, Humanized therapeutic use, Antibodies, Monoclonal, Humanized administration & dosage, Antibodies, Monoclonal, Humanized adverse effects, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms pathology, Triple Negative Breast Neoplasms mortality, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Neoplasm, Residual pathology, Paclitaxel administration & dosage, Paclitaxel therapeutic use, Paclitaxel adverse effects
- Abstract
Background: KEYNOTE-522 demonstrated statistically significant improvements in pathological complete response (pCR) with neoadjuvant pembrolizumab plus chemotherapy and event-free survival (EFS) with neoadjuvant pembrolizumab plus chemotherapy followed by adjuvant pembrolizumab in patients with high-risk, early-stage triple-negative breast cancer (TNBC). Prior studies have shown the prognostic value of the residual cancer burden (RCB) index to quantify the extent of residual disease after neoadjuvant chemotherapy. In this preplanned exploratory analysis, we assessed RCB distribution and EFS within RCB categories by treatment group., Patients and Methods: A total of 1174 patients with stage T1c/N1-2 or T2-4/N0-2 TNBC were randomized 2 : 1 to pembrolizumab 200 mg or placebo every 3 weeks given with four cycles of paclitaxel + carboplatin, followed by four cycles of doxorubicin or epirubicin + cyclophosphamide. After surgery, patients received pembrolizumab or placebo for nine cycles or until recurrence or unacceptable toxicity. Primary endpoints are pCR and EFS. RCB is a prespecified exploratory endpoint. The association between EFS and RCB was assessed using a Cox regression model., Results: Pembrolizumab shifted patients into lower RCB categories across the entire spectrum compared with placebo. There were more patients in the pembrolizumab group with RCB-0 (pCR), and fewer patients in the pembrolizumab group with RCB-1, RCB-2, and RCB-3. The corresponding hazard ratios (95% confidence intervals) for EFS were 0.70 (0.38-1.31), 0.92 (0.39-2.20), 0.52 (0.32-0.82), and 1.24 (0.69-2.23). The most common first EFS events were distant recurrences, with fewer in the pembrolizumab group across all RCB categories. Among patients with RCB-0/1, more than half [21/38 (55.3%)] of all events were central nervous system recurrences, with 13/22 (59.1%) in the pembrolizumab group and 8/16 (50.0%) in the placebo group., Conclusions: Addition of pembrolizumab to chemotherapy resulted in fewer EFS events in the RCB-0, RCB-1, and RCB-2 categories, with the greatest benefit in RCB-2. These findings demonstrate that pembrolizumab not only increased pCR rates, but also improved EFS among most patients who do not have a pCR., Competing Interests: Disclosure All authors’ institutions received research funding from Merck Sharp & Dohme LLC, a subsidiary of Merck & Co., Inc., Rahway, NJ, USA, for the conduct of this study. YZ, WP, and KT are full-time employees of Merck Sharp & Dohme LLC, a subsidiary of Merck & Co., Inc., Rahway, NJ, USA and hold stock and/or restricted stock units in Merck & Co., Inc., Rahway, NJ, USA., (Copyright © 2024 European Society for Medical Oncology. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
137. TROPION-Breast03: a randomized phase III global trial of datopotamab deruxtecan ± durvalumab in patients with triple-negative breast cancer and residual invasive disease at surgical resection after neoadjuvant therapy.
- Author
-
Bardia A, Pusztai L, Albain K, Ciruelos EM, Im SA, Hershman D, Kalinsky K, Isaacs C, Loirat D, Testa L, Tokunaga E, Wu J, Dry H, Barlow W, Kozarski R, Maxwell M, Harbeck N, and Sharma P
- Abstract
Background: Despite advances in the treatment of early triple-negative breast cancer (TNBC), patients with residual invasive disease after neoadjuvant therapy have a high risk of disease recurrence and worse survival outcomes than those who have pathological complete response (pCR). Improving outcomes in early TNBC remains an unmet need requiring new adjuvant treatment approaches. Datopotamab deruxtecan (Dato-DXd) is an antibody-drug conjugate comprising a humanized anti-trophoblast cell-surface antigen 2 immunoglobulin G1 (IgG1) monoclonal antibody attached via a plasma-stable, cleavable linker to a potent topoisomerase I inhibitor payload, with activity observed in advanced TNBC., Objectives: TROPION-Breast03 is an ongoing phase III study evaluating the efficacy and safety of Dato-DXd alone or combined with durvalumab versus standard-of-care therapy as adjuvant treatment in patients with stage I-III TNBC with residual invasive disease at surgical resection following neoadjuvant treatment., Methods and Design: Eligible patients, aged ⩾18 years, will be randomized in a 2:1:2 ratio to receive Dato-DXd [6 mg/kg intravenously (IV) every 3 weeks (Q3W); eight cycles] and durvalumab (1120 mg IV Q3W; nine cycles), Dato-DXd monotherapy (6 mg/kg IV Q3W), or investigator's choice of therapy (ICT; capecitabine, pembrolizumab, or capecitabine and pembrolizumab). The primary endpoint is invasive disease-free survival (iDFS) for Dato-DXd and durvalumab versus ICT. Key secondary endpoints include safety, distant disease-free survival, and overall survival for Dato-DXd and durvalumab versus ICT and iDFS for Dato-DXd monotherapy versus ICT., Ethics: TROPION-Breast03 will be approved by the independent ethics committees or institutional review boards at each study site. All study participants will provide written informed consent., Discussion: TROPION-Breast03 will help define the potential role of Dato-DXd in the treatment of patients with early-stage TNBC who do not have pCR after neoadjuvant therapy., Trial Registration: ClinicalTrials.gov identifier: NCT05629585 (registration date: 29 November 2022)., Competing Interests: A.B. reports consulting fees from Pfizer, Novartis, Genentech, Merck, Radius Health, Immunomedics/Gilead, Sanofi, Daiichi Pharma/AstraZeneca, Phillips, Eli Lilly, and Foundation Medicine; grants or funds from Genentech, Novartis, Pfizer, Merck, Sanofi, Radius Health, Immunomedics/Gilead, Daiichi Pharma/AstraZeneca, and Eli Lilly. L.P. reports membership on the board of directors for the HOPE Foundation; consulting fees from Pfizer, AstraZeneca, Merck, Novartis, Bristol Myers Squibb, GlaxoSmithKline, Roche/Genentech, Personalis, Daiichi, Natera, and Exact Sciences; and institutional research funding from Seagen, GlaxoSmithKline, AstraZeneca, Merck, Pfizer, and Bristol Myers Squibb. K.A. reports institutional research funding from Quantum Leap (I-SPY 2); and non-financial conflicts with IDMC (member) and Seattle Genetics. EMC reports consulting fees from Pfizer, MSD, Gilead, Roche, Eli Lilly, Daiichi-Sankyo, Novartis, and AstraZeneca; and institutional research funding from Roche. S.-A.I. reports advisory council or committee participation with AstraZeneca, Novartis, Hanmi, Pfizer, Eisai, Roche, Eli Lilly, GlaxoSmithKline, MSD, Daiichi-Sankyo, Idience, and Bertis; and grants or funds from AstraZeneca, Pfizer, Eisai, Roche, Daewoong Pharm, Boryung Pharm, and Daiichi-Sankyo. D.H. declares no conflicts of interest. K.K. reports consulting fees from Merck, Eli Lilly, Novartis, AstraZeneca, Roche/Genentech, Immunomedics, Seattle Genetics, Oncosec, 4D pharma, Daiichi-Sankyo, Puma Biotechnology, Mersna, Menarini Silicon Biosystems, Myovant Sciences, and Takeda; and grants or funds from Novartis, Ascentage, Roche/Genentech, Eli Lilly, Seattle Genetics, AstraZeneca, and Daiichi-Sankyo. C.I. reports consulting fees from Genentech, Puma Biotechnology, Seattle Genetics, AstraZeneca, Novartis, Pfizer, ION, and Gilead; institutional research funding from Tesaro/GlaxoSmithKline, Seattle Genetics, Pfizer, AstraZeneca, Bristol Myers Squibb, Genentech, and Novartis; and other financial relationships with Wolters Kluwer (UptoDate) and McGraw Hill (Goodman and Gillman). D.L. reports advisory council or committee participation with MSD, Seagen, Novartis, Eli Lilly, AstraZeneca, and Exact Sciences; honoraria from MSD, Seagen, Pfizer, Novartis, Eli Lilly, AstraZeneca, Roche, Exact Sciences, and Daiichi-Sankyo; and consulting fees from MSD and AstraZeneca. L.T. reports advisory council or committee participation with AstraZeneca, Daichii-Sankyo, MSD, Eli Lilly, Novartis, and Pfizer; consulting fees from AstraZeneca, Daiichi-Sankyo, MSD, Novartis, and Pfizer; grants or funds from Novartis; and provision of educational support for Roche, Pfizer, AstraZeneca, and Gilead. E.T. reports honoraria from Daiichi-Sankyo, AstraZeneca, and Eli Lilly. J.W. reports research funding from the National Natural Science Foundation and F. Hoffmann-La Roche Ltd; committee participation with Eli Lilly, AstraZeneca; honoraria from Novartis. H.D. reports employment by AstraZeneca and ownership of stock/shares in AstraZeneca. W.B. declares no conflicts of interest. R.K. reports employment by AstraZeneca. M.M. reports employment by AstraZeneca and ownership of stock/shares in AstraZeneca. N.H. reports membership on the board of directors of the West German Study Group (WSG); status as the ESMO Director of Education; honoraria from Amgen, AstraZeneca, Daiichi-Sankyo, EPG Communication, Gilead, Eli Lilly, Medscape, MSD, Novartis, Pierre-Fabre, Pfizer, Roche, Sandoz, Sanofi, Seagen, Springer, Viatris, and Zuellig Pharma; consulting fees from Gilead, Roche, Sandoz, Sanofi, and Seagen. P.S. reports consulting fees from Pfizer, Merck, Gilead, Genzyme (Sanofi), Novartis, AstraZeneca, and GlaxoSmithKline; institutional research funding from Novartis, Merck, and Gilead; and royalties from UpToDate., (© The Author(s), 2024.)
- Published
- 2024
- Full Text
- View/download PDF
138. Outstanding Properties of the Hydration Shell around β-d-Glucose: A Computational Study.
- Author
-
Bakó I, Pusztai L, and Pothoczki S
- Abstract
Ab initio molecular dynamics (AIMD) simulations have been performed on aqueous solutions of four simple sugars, α-d-glucose, β-d-glucose, α-d-mannose, and α-d-galactose. Hydrogen-bonding (HB) properties, such as the number of donor- and acceptor-type HB-s, and the lengths and strengths of hydrogen bonds between sugar and water molecules, have been determined. Related electronic properties, such as the dipole moments of water molecules and partial charges of the sugar O atoms, have also been calculated. The hydrophilic and hydrophobic shells were characterized by means of spatial distribution functions. β-d-Glucose was found to form the highest number of hydrophilic and the smallest number of hydrophobic connections to neighboring water molecules. The average sugar-water H-bond length was the shortest for β-d-glucose, which suggests that these are the strongest such H-bonds. Furthermore, β-d-glucose appears to stand out in terms of the symmetry properties of both its hydrophilic and hydrophobic hydration shells. In summary, in all aspects considered here, there seems to be a correlation between the distinct characteristics of β-d-glucose reported here and its outstanding solubility in water. Admittedly, our findings represent only some of the important factors that influence the solubility., Competing Interests: The authors declare no competing financial interest., (© 2024 The Authors. Published by American Chemical Society.)
- Published
- 2024
- Full Text
- View/download PDF
139. Image-based multiplex immune profiling of cancer tissues: translational implications. A report of the International Immuno-oncology Biomarker Working Group on Breast Cancer.
- Author
-
Jahangir CA, Page DB, Broeckx G, Gonzalez CA, Burke C, Murphy C, Reis-Filho JS, Ly A, Harms PW, Gupta RR, Vieth M, Hida AI, Kahila M, Kos Z, van Diest PJ, Verbandt S, Thagaard J, Khiroya R, Abduljabbar K, Acosta Haab G, Acs B, Adams S, Almeida JS, Alvarado-Cabrero I, Azmoudeh-Ardalan F, Badve S, Baharun NB, Bellolio ER, Bheemaraju V, Blenman KR, Botinelly Mendonça Fujimoto L, Burgues O, Chardas A, Cheang MCU, Ciompi F, Cooper LA, Coosemans A, Corredor G, Dantas Portela FL, Deman F, Demaria S, Dudgeon SN, Elghazawy M, Fernandez-Martín C, Fineberg S, Fox SB, Giltnane JM, Gnjatic S, Gonzalez-Ericsson PI, Grigoriadis A, Halama N, Hanna MG, Harbhajanka A, Hart SN, Hartman J, Hewitt S, Horlings HM, Husain Z, Irshad S, Janssen EA, Kataoka TR, Kawaguchi K, Khramtsov AI, Kiraz U, Kirtani P, Kodach LL, Korski K, Akturk G, Scott E, Kovács A, Laenkholm AV, Lang-Schwarz C, Larsimont D, Lennerz JK, Lerousseau M, Li X, Madabhushi A, Maley SK, Manur Narasimhamurthy V, Marks DK, McDonald ES, Mehrotra R, Michiels S, Kharidehal D, Minhas FUAA, Mittal S, Moore DA, Mushtaq S, Nighat H, Papathomas T, Penault-Llorca F, Perera RD, Pinard CJ, Pinto-Cardenas JC, Pruneri G, Pusztai L, Rajpoot NM, Rapoport BL, Rau TT, Ribeiro JM, Rimm D, Vincent-Salomon A, Saltz J, Sayed S, Hytopoulos E, Mahon S, Siziopikou KP, Sotiriou C, Stenzinger A, Sughayer MA, Sur D, Symmans F, Tanaka S, Taxter T, Tejpar S, Teuwen J, Thompson EA, Tramm T, Tran WT, van der Laak J, Verghese GE, Viale G, Wahab N, Walter T, Waumans Y, Wen HY, Yang W, Yuan Y, Bartlett J, Loibl S, Denkert C, Savas P, Loi S, Specht Stovgaard E, Salgado R, Gallagher WM, and Rahman A
- Subjects
- Humans, Female, Biomarkers, Tumor genetics, Prognosis, Phenotype, United Kingdom, Tumor Microenvironment, Breast Neoplasms
- Abstract
Recent advances in the field of immuno-oncology have brought transformative changes in the management of cancer patients. The immune profile of tumours has been found to have key value in predicting disease prognosis and treatment response in various cancers. Multiplex immunohistochemistry and immunofluorescence have emerged as potent tools for the simultaneous detection of multiple protein biomarkers in a single tissue section, thereby expanding opportunities for molecular and immune profiling while preserving tissue samples. By establishing the phenotype of individual tumour cells when distributed within a mixed cell population, the identification of clinically relevant biomarkers with high-throughput multiplex immunophenotyping of tumour samples has great potential to guide appropriate treatment choices. Moreover, the emergence of novel multi-marker imaging approaches can now provide unprecedented insights into the tumour microenvironment, including the potential interplay between various cell types. However, there are significant challenges to widespread integration of these technologies in daily research and clinical practice. This review addresses the challenges and potential solutions within a structured framework of action from a regulatory and clinical trial perspective. New developments within the field of immunophenotyping using multiplexed tissue imaging platforms and associated digital pathology are also described, with a specific focus on translational implications across different subtypes of cancer. © 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland., (© 2024 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.)
- Published
- 2024
- Full Text
- View/download PDF
140. Neoadjuvant Trebananib plus Paclitaxel-based Chemotherapy for Stage II/III Breast Cancer in the Adaptively Randomized I-SPY2 Trial-Efficacy and Biomarker Discovery.
- Author
-
Albain KS, Yau C, Petricoin EF, Wolf DM, Lang JE, Chien AJ, Haddad T, Forero-Torres A, Wallace AM, Kaplan H, Pusztai L, Euhus D, Nanda R, Elias AD, Clark AS, Godellas C, Boughey JC, Isaacs C, Tripathy D, Lu J, Yung RL, Gallagher RI, Wulfkuhle JD, Brown-Swigart L, Krings G, Chen YY, Potter DA, Stringer-Reasor E, Blair S, Asare SM, Wilson A, Hirst GL, Singhrao R, Buxton M, Clennell JL, Sanil A, Berry S, Asare AL, Matthews JB, DeMichele AM, Hylton NM, Melisko M, Perlmutter J, Rugo HS, Symmans WF, Van't Veer LJ, Yee D, Berry DA, and Esserman LJ
- Subjects
- Female, Humans, Antineoplastic Combined Chemotherapy Protocols adverse effects, Bayes Theorem, Neoadjuvant Therapy, Paclitaxel adverse effects, Receptor, ErbB-2 metabolism, Receptor, TIE-2, Trastuzumab adverse effects, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Breast Neoplasms pathology, Recombinant Fusion Proteins
- Abstract
Purpose: The neutralizing peptibody trebananib prevents angiopoietin-1 and angiopoietin-2 from binding with Tie2 receptors, inhibiting angiogenesis and proliferation. Trebananib was combined with paclitaxel±trastuzumab in the I-SPY2 breast cancer trial., Patients and Methods: I-SPY2, a phase II neoadjuvant trial, adaptively randomizes patients with high-risk, early-stage breast cancer to one of several experimental therapies or control based on receptor subtypes as defined by hormone receptor (HR) and HER2 status and MammaPrint risk (MP1, MP2). The primary endpoint is pathologic complete response (pCR). A therapy "graduates" if/when it achieves 85% Bayesian probability of success in a phase III trial within a given subtype. Patients received weekly paclitaxel (plus trastuzumab if HER2-positive) without (control) or with weekly intravenous trebananib, followed by doxorubicin/cyclophosphamide and surgery. Pathway-specific biomarkers were assessed for response prediction., Results: There were 134 participants randomized to trebananib and 133 to control. Although trebananib did not graduate in any signature [phase III probabilities: Hazard ratio (HR)-negative (78%), HR-negative/HER2-positive (74%), HR-negative/HER2-negative (77%), and MP2 (79%)], it demonstrated high probability of superior pCR rates over control (92%-99%) among these subtypes. Trebananib improved 3-year event-free survival (HR 0.67), with no significant increase in adverse events. Activation levels of the Tie2 receptor and downstream signaling partners predicted trebananib response in HER2-positive disease; high expression of a CD8 T-cell gene signature predicted response in HR-negative/HER2-negative disease., Conclusions: The angiopoietin (Ang)/Tie2 axis inhibitor trebananib combined with standard neoadjuvant therapy increased estimated pCR rates across HR-negative and MP2 subtypes, with probabilities of superiority >90%. Further study of Ang/Tie2 receptor axis inhibitors in validated, biomarker-predicted sensitive subtypes is warranted., (©2023 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
141. Early breast cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up.
- Author
-
Loibl S, André F, Bachelot T, Barrios CH, Bergh J, Burstein HJ, Cardoso MJ, Carey LA, Dawood S, Del Mastro L, Denkert C, Fallenberg EM, Francis PA, Gamal-Eldin H, Gelmon K, Geyer CE, Gnant M, Guarneri V, Gupta S, Kim SB, Krug D, Martin M, Meattini I, Morrow M, Janni W, Paluch-Shimon S, Partridge A, Poortmans P, Pusztai L, Regan MM, Sparano J, Spanic T, Swain S, Tjulandin S, Toi M, Trapani D, Tutt A, Xu B, Curigliano G, and Harbeck N
- Subjects
- Humans, Female, Follow-Up Studies, Breast Neoplasms diagnosis, Breast Neoplasms epidemiology, Breast Neoplasms therapy
- Published
- 2024
- Full Text
- View/download PDF
142. Core Needle Biopsies as an Alternative Source for Ex Vivo Expanded TIL for Adoptive Cell Therapy in Triple-Negative Breast Cancer.
- Author
-
Coman MM, Pusztai L, Hooley R, Andreveja L, Kim L, Joshi N, Bersenev A, Krause D, and Park TS
- Subjects
- Humans, Biopsy, Large-Core Needle, Lymphocytes, Tumor-Infiltrating pathology, Phenotype, Immunotherapy, Adoptive, Triple Negative Breast Neoplasms therapy
- Abstract
Adoptive transfer of ex vivo expanded tumor-infiltrating lymphocytes (TILs) have produced long-term response in metastatic cancers. TILs have traditionally been expanded from surgically resected specimens. Ultrasound-guided core needle biopsy (CNB) is an alternative method that avoids the morbidity of surgery and have added benefits which may include patients not amenable to surgery as well as the potential to produce TILs from multiple lesions in the same patient. We assessed the ability to produce and expand TILs from primary triple-negative breast cancer tumors from CNB (n=7) and demonstrate comparable expansion, phenotype and cytokine secretion after phorbol myristate acetate-ionomycin stimulation to TILs expanded from surgery (n=6). T cell Receptor clonality and diversity were also comparable between the two cohorts throughout the TIL culture. CNB is a safe and feasible method to obtain tumor tissue for TIL generation in patients with triple-negative breast cancer., (Copyright © 2023 Wolters Kluwer Health, Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
143. Breast cancers with high proliferation and low ER-related signalling have poor prognosis and unique molecular features with implications for therapy.
- Author
-
Licata L, Barreca M, Galbardi B, Dugo M, Viale G, Győrffy B, Karn T, Pusztai L, Gianni L, Callari M, and Bianchini G
- Subjects
- Humans, Female, Biomarkers, Disease-Free Survival, Cell Proliferation, Cyclins therapeutic use, Neoadjuvant Therapy, Prognosis, Breast Neoplasms drug therapy, Breast Neoplasms genetics, Breast Neoplasms pathology
- Abstract
Background: Luminal breast cancers with high proliferation (MKS
hi ) and low ER-related signalling (ERSlo ) have a poor prognosis. We investigated treatment responses and molecular features of MKShi /ERSlo tumours to inform potential therapies., Methods: Gene expression data from patients who received neoadjuvant chemotherapy (NAC) without (MDACC, N = 199) or with pembrolizumab (I-SPY2, N = 40), or endocrine therapy (NET) without (POETIC, N = 172) or with palbociclib (NeoPalAna, N = 32) were analyzed to assess treatment response by MKS/ERS-subgroups. TCGA was used to assess the mutational landscape and biomarkers associated with palbociclib-resistance (Cyclin-E, RBsig, IRPR) and immunotherapy-response (TMB, TILs, T-cell inflamed) by MKS/ERS-subgroups., Results: Compared to MKShi /ERShi tumours, MKShi /ERSlo tumours had higher pathological response rates to NAC (22% vs 8%, p = 0.06) but a higher recurrence risk (4-year metastasis-free survival 70% vs 94%, p = 0.01). MKShi /ERSlo tumours frequently harboured TP53 (34%) and PIK3CA (33%) mutations, and showed high expression of Cyclin-E, RBsig and IRPR, high TMB and elevated TIL and T-cell inflamed metagene expression. MKShi /ERSlo tumours retained high proliferation after NET with or without palbociclib but had higher pathological complete response rates when pembrolizumab was added to NAC (42% vs 21%, p = 0.07)., Conclusions: MKShi /ERSlo tumours have dismal outcomes and are enriched in chemotherapy-sensitive but ET- and palbociclib-resistant tumours. Biomarker analysis and clinical data suggest a potential role for immunotherapy in this group., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
144. Pan-cancer analysis of antibody-drug conjugate targets and putative predictors of treatment response.
- Author
-
Bosi C, Bartha Á, Galbardi B, Notini G, Naldini MM, Licata L, Viale G, Mariani M, Pistilli B, Ali HR, André F, Piras M, Callari M, Barreca M, Locatelli A, Viganò L, Criscitiello C, Pusztai L, Curigliano G, Győrffy B, Dugo M, and Bianchini G
- Subjects
- Humans, Immunoconjugates therapeutic use, Antineoplastic Agents therapeutic use, Lung Neoplasms drug therapy
- Abstract
Background: Antibody-drug conjugates (ADCs) are a rapidly expanding class of compounds in oncology. Our goal was to assess the expression of ADC targets and potential downstream determining factors of activity across pan-cancer and normal tissues., Materials and Methods: ADCs in clinical trials (n = 121) were identified through ClinicalTrials.gov, corresponding to 54 targets. Genes potentially implicated in treatment response were identified in the literature. Gene expression from The Cancer Genome Atlas (9000+ cancers of 31 cancer types), the Genotype-Tissue Expression database (n = 19,000 samples from 31 normal tissue types), and the TNMplot.com (n = 12,494 unmatched primary and metastatic samples) were used in this analysis. To compare relative expression across and within tumour types we used pooled normal tissues as reference., Results: For most ADC targets, mRNA levels correlated with protein expression. Pan-cancer target expression distributions identified appealing cancer types for each ADC development. Co-expression of multiple targets was common and suggested opportunities for ADC combinations. Expression levels of genes potentially implicated in ADC response downstream of the target might provide additional information (e.g. TOP1 was highly expressed in many tumour types, including breast and lung cancers). Metastatic compared to primary tissues overexpressed some ADCs targets. Single sample "targetgram" plots were generated to visualise the expression of potentially competing ADC targets and resistance/sensitivity markers highlighting high inter-patient heterogeneity. Off-cancer target expression only partially explains adverse events, while expression of determinants of payload activity explained more of the observed toxicities., Conclusion: Our findings draw attention to new therapeutic opportunities for ADCs that can be tested in the clinic and our web platform (https://tnmplot.com) can assist in prioritising upcoming ADC targets for clinical development., Competing Interests: Declaration of Competing Interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: C.B.: Financial interests: personal consulting fees from Kardo srl (personal); travel support for attending meetings from Daiichi-Sankyo and Lilly. G.N.: Travel support for attending meetings from Lilly and Sanofi. M.M.: Travel support for attending meetings from Lilly. L.L.: Financial interests: Consulting fees from Exact Sciences, Helsinn, EISAI; honoraria for speakers’ bureaus from Gilead, Exact Sciences, Helsinn; support for attending meetings from Lilly and Gilead; advisory board for Lilly, Exact Sciences, AstraZeneca, Italfarmaco, Accord, Seagen and Daiichi Sankyo (all personal and financial). G.V.: Financial interests: Advisory board for Gilead; speakers’ bureaus: Novartis, Lilly; support for attending meetings: Pfizer, Lilly (all personal and financial). B.P.: Financial interests: Consulting fees from AstraZeneca (institutional), Seagen (institutional), Gilead (institutional), Novartis (institutional), Lilly (institutional), MSD (institutional), Pierre Fabre (personal), Daiichi-Sankyo (institutional/personal); research funding (to the institution): Astra Zeneca, Daiichi-Sankyo, Gilead, Seagen, MSD; travel support: Astra Zeneca; Pierre Fabre; MSD; Daiichi-Sankyo, Pfizer. F.A.: Financial interests: grants or speaker/Advisory compensated to hospital: AstraZeneca, Daiichi Sankyo, Pfizer, Lilly, Relay; honorarium: Lilly. M.P.: Financial interests: Travel support for attending meetings from Gilead and Novartis. C.C.: Financial interests: Personal fees for consulting, advisory role, and speakers’ bureau from Lilly, Roche, Novartis, MSD, Seagen, Gilead, Daiichi Sankyo, AstraZeneca, and Pfizer. L.P.: Financial interests: Consulting fees and honoraria for advisory board participation from Pfizer, Astra Zeneca, Merck, Novartis, Bristol-Myers Squibb, Stemline-Menarini, GlaxoSmithKline, Genentech/Roche, Personalis, Daiichi, Natera, Exact Sciences (personal), and institutional research funding from Seagen, GlaxoSmithKline, AstraZeneca, Merck, Pfizer and Bristol Myers Squibb. G.C.: Financial Interests: AstraZeneca, Invited Speaker, Personal; AstraZeneca, Advisory Board, Personal, BMS, Advisory Board, Personal; Celcuity, Advisory Board, Personal; Daiichi Sankyo, Invited Speaker, personal; Daiichi Sankyo, Advisory Board, Personal; Exact Sciences, Advisory Board, Personal; Gilead, Advisory Board, Personal, Advisory Board; Lilly, Advisory Board, Personal; Menarini, Advisory Board, Personal, Advisory Board; Merck, Advisory Board, Personal; Novartis, Invited Speaker, Personal; Pfizer, Writing Engagement, Personal; Pfizer, Advisory Board, Personal; Pfizer, Invited Speaker, Personal; Roche, Advisory Board, Personal; Roche, Invited Speaker, Personal; Veracyte, Advisory Board, Personal; Ellipsis, Other, Personal, Advisory Board; Astellas, Funding, Institutional, Financial interest, Phase I studies; AstraZeneca, Funding, Institutional, Financial interest, Phase I studies; Blueprint Medicine, Funding, Institutional, Financial interest, Phase I studies; BMS, Funding, Institutional, Financial interest, Phase I studies; Daiichi Sankyo, Funding, Institutional, Financial interest, Phase I studies; Kymab, Funding, Institutional, Financial interest, Phase I studies; Merck, Research Grant, Institutional, Financial interest, Investigator Initiated Trial; Novartis, Funding, Institutional, Financial interest, Phase I studies; Philogen, Funding, Institutional, Financial interest, Phase I studies; Relay Therapeutics, Coordinating PI, Institutional, Financial interest, Phase I clinical basket trial; Roche, Funding, Institutional, Financial interest, Phase I studies; Sanofi, Funding, Institutional, Financial interest, Phase I studies. Non-financial interests: Consiglio Superiore di Sanità, Officer, Italian National Health Council as Advisor for Ministry of Health; ESMO, Officer, ESMO Clinical Practice Guidelines Chair; ESMO, Member of Board of Directors, Chair of Clinical Practice Guidelines Committee; Europa Donna, Advisory Role, Member of the Scientific Council. Patient advocacy association; EUSOMA, Officer, Member of the Advisory Council; Fondazione Beretta, Advisory Role, Cancer Research Foundation; Lega Italiana Lotta ai Tumori, Member of Board of Directors, No compensation for this role. This a public national company for cancer prevention. G.B.: Financial Interests: AstraZeneca, Advisory Board, Personal; AstraZeneca, Other, Personal, Consultancy; Daiichi Sankyo, Advisory Board, Personal; Daiichi Sankyo, Other, Personal, Consultancy; Lilly, Advisory Board, Personal; Lilly, Invited Speaker, Personal; Novartis, Advisory Board, Personal; Pfizer, Advisory Board, Personal; Roche, Other, Personal, Consultancy; MSD, Other, Personal, Consultancy; Gilead, Other, Personal, Consultancy; Sanofi, Other, Personal, Consultancy; Roche, Invited Speaker, Personal; AstraZeneca, Invited Speaker, Personal; Daiichi Sankyo, Invited Speaker, Personal; MSD, Invited Speaker, Personal; Chugai, Invited Speaker, Personal; EISAI, Invited Speaker, Personal; Gilead, Invited Speaker, Personal; Seagen, Invited Speaker, Personal; Neopharm Israel, Invited Speaker, Personal; Roche, Other, Personal, Support for attending meetings and/or travel; Pfizer, Other, Personal, Support for attending meetings and/or travel; MSD, Other, Personal, Support for attending meetings and/or travel; Chugai, Other, Personal, Support for attending meetings and/or travel; Novartis, Other, Personal, Support for attending meetings and/or travel; Roche, Advisory Board, Personal; Amgen, Advisory Board, Personal; MSD, Advisory Board, Personal; Chugai, Advisory Board, Personal; EISAI, Advisory Board, Personal; Gilead, Advisory Board, Personal; Seagen, Advisory Board, Personal; Exact Science, Advisory Board, Personal; Roche, Advisory Board, Personal; MSD, Advisory Board, Personal; Gilead, Advisory Board, Personal; Gilead, Other, Personal, Support for attending meetings and/or travel; Daiichi Sankyo, Other, Personal, Support for attending meetings and/or travel; Roche, Steering Committee Member, Financial interest, Personal and Institutional; Novartis, Steering Committee Member, Financial interest, Personal and Institutional; Lilly, Steering Committee Member, Financial interest, Personal and Institutional; AstraZeneca, Steering Committee Member, Financial interest, Personal and Institutional; Gilead, Local PI, Financial interest, Institutional; Pfizer, Local PI, Financial interest, Institutional; Daiichi Sankyo, Local PI, Financial interest, Institutional; Lilly, Local PI, Financial interest, Institutional; MSD, Local PI, Financial interest, Institutional; Novartis, Local PI, Financial interest, Institutional; Non-Financial Interests: Fondazione Michelangelo, Leadership Role, Head of Traslational Research. A.B., B.Ga, H.R.A., M.C., M.B., A.L., L.V., M.D., M.M.N., and B.G. declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2023 The Author(s). Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
145. De Novo Oligometastatic Breast Cancer.
- Author
-
Pusztai L, Rozenblit M, Dubsky P, Bachelot T, Kirby AM, Linderholm BK, White JR, Chmura SJ, Carey LA, Chua BH, and Miller KD
- Subjects
- Female, Humans, Breast Neoplasms pathology, Neoplasm Metastasis
- Published
- 2023
- Full Text
- View/download PDF
146. Race, Gene Expression Signatures, and Clinical Outcomes of Patients With High-Risk Early Breast Cancer.
- Author
-
Kyalwazi B, Yau C, Campbell MJ, Yoshimatsu TF, Chien AJ, Wallace AM, Forero-Torres A, Pusztai L, Ellis ED, Albain KS, Blaes AH, Haley BB, Boughey JC, Elias AD, Clark AS, Isaacs CJ, Nanda R, Han HS, Yung RL, Tripathy D, Edmiston KK, Viscusi RK, Northfelt DW, Khan QJ, Asare SM, Wilson A, Hirst GL, Lu R, Symmans WF, Yee D, DeMichele AM, van 't Veer LJ, Esserman LJ, and Olopade OI
- Subjects
- Female, Humans, Retrospective Studies, Transcriptome, Pathologic Complete Response, Disease-Free Survival, Breast Neoplasms genetics, Racial Groups
- Abstract
Importance: There has been little consideration of genomic risk of recurrence by breast cancer subtype despite evidence of racial disparities in breast cancer outcomes., Objective: To evaluate associations between clinical trial end points, namely pathologic complete response (pCR) and distant recurrence-free survival (DRFS), and race and examine whether gene expression signatures are associated with outcomes by race., Design, Setting, and Participants: This retrospective cohort study used data from the Investigation of Serial Studies to Predict Your Therapeutic Response With Imaging and Molecular Analysis 2 (I-SPY 2) multicenter clinical trial of neoadjuvant chemotherapy with novel agents and combinations for patients with previously untreated stage II/III breast cancer. Analyses were conducted of associations between race and short- and long-term outcomes, overall and by receptor subtypes, and their association with 28 expression biomarkers. The trial enrolled 990 female patients between March 30, 2010, and November 5, 2016, with a primary tumor size of 2.5 cm or greater and clinical or molecular high risk based on MammaPrint or hormone receptor (HR)-negative/ERBB2 (formerly HER2 or HER2/neu)-positive subtyping across 9 arms. This data analysis was performed between June 10, 2021, and October 20, 2022., Exposure: Race, tumor receptor subtypes, and genomic biomarker expression of early breast cancer., Main Outcomes and Measures: The primary outcomes were pCR and DRFS assessed by race, overall, and by tumor subtype using logistic regression and Cox proportional hazards regression models. The interaction between 28 expression biomarkers and race, considering pCR and DRFS overall and within subtypes, was also evaluated., Results: The analytic sample included 974 participants (excluding 16 self-reporting as American Indian or Alaska Native, Native Hawaiian or Other Pacific Islander, or multiple races due to small sample sizes), including 68 Asian (7%), 120 Black (12%), and 786 White (81%) patients. Median (range) age at diagnosis was 47 (25-71) years for Asian, 49 (25-77) for Black, and 49 (23-73) years for White patients. The pCR rates were 32% (n = 22) for Asian, 30% for Black (n = 36), and 32% for White (n = 255) patients (P = .87). Black patients with HR-positive/ERBB2-negative tumors not achieving pCR had significantly worse DRFS than their White counterparts (hazard ratio, 2.28; 95% CI, 1.24-4.21; P = .01), with 5-year DRFS rates of 55% (n = 32) and 77% (n = 247), respectively. Black patients with HR-positive/ERBB2-negative tumors, compared with White patients, had higher expression of an interferon signature (mean [SD], 0.39 [0.87] and -0.10 [0.99]; P = .007) and, compared with Asian patients, had a higher mitotic score (mean [SD], 0.07 [1.08] and -0.69 [1.06]; P = .01) and lower estrogen receptor/progesterone receptor signature (mean [SD], 0.31 [0.90] and 1.08 [0.95]; P = .008). A transforming growth factor β signature had a significant association with race relative to pCR and DRFS, with a higher signature associated with lower pCR and worse DRFS outcomes among Black patients only., Conclusions and Relevance: The findings show that women with early high-risk breast cancer who achieve pCR have similarly good outcomes regardless of race, but Black women with HR-positive/ERBB2-negative tumors without pCR may have worse DRFS than White women, highlighting the need to develop and test novel biomarker-informed therapies in diverse populations.
- Published
- 2023
- Full Text
- View/download PDF
147. Immunological and clinicopathological features predict HER2-positive breast cancer prognosis in the neoadjuvant NeoALTTO and CALGB 40601 randomized trials.
- Author
-
Rediti M, Fernandez-Martinez A, Venet D, Rothé F, Hoadley KA, Parker JS, Singh B, Campbell JD, Ballman KV, Hillman DW, Winer EP, El-Abed S, Piccart M, Di Cosimo S, Symmans WF, Krop IE, Salgado R, Loi S, Pusztai L, Perou CM, Carey LA, and Sotiriou C
- Subjects
- Female, Humans, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Hormones, Neoadjuvant Therapy, Randomized Controlled Trials as Topic, Receptor, ErbB-2 genetics, Receptor, ErbB-2 therapeutic use, Trastuzumab therapeutic use, Treatment Outcome, Tumor Microenvironment, Breast Neoplasms drug therapy, Breast Neoplasms genetics
- Abstract
The identification of prognostic markers in patients receiving neoadjuvant therapy is crucial for treatment optimization in HER2-positive breast cancer, with the immune microenvironment being a key factor. Here, we investigate the complexity of B and T cell receptor (BCR and TCR) repertoires in the context of two phase III trials, NeoALTTO and CALGB 40601, evaluating neoadjuvant paclitaxel with trastuzumab and/or lapatinib in women with HER2-positive breast cancer. BCR features, particularly the number of reads and clones, evenness and Gini index, are heterogeneous according to hormone receptor status and PAM50 subtypes. Moreover, BCR measures describing clonal expansion, namely evenness and Gini index, are independent prognostic factors. We present a model developed in NeoALTTO and validated in CALGB 40601 that can predict event-free survival (EFS) by integrating hormone receptor and clinical nodal status, breast pathological complete response (pCR), stromal tumor-infiltrating lymphocyte levels (%) and BCR repertoire evenness. A prognostic score derived from the model and including those variables, HER2-EveNT, allows the identification of patients with 5-year EFS > 90%, and, in those not achieving pCR, of a subgroup of immune-enriched tumors with an excellent outcome despite residual disease., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
148. Pembrolizumab Plus Chemotherapy Followed by Pembrolizumab in Patients With Early Triple-Negative Breast Cancer: A Secondary Analysis of a Randomized Clinical Trial.
- Author
-
Takahashi M, Cortés J, Dent R, Pusztai L, McArthur H, Kümmel S, Denkert C, Park YH, Im SA, Ahn JH, Mukai H, Huang CS, Chen SC, Kim MH, Jia L, Li XT, Tryfonidis K, Karantza V, Iwata H, and Schmid P
- Subjects
- Adult, Humans, Female, Middle Aged, B7-H1 Antigen, Antibodies, Monoclonal, Humanized therapeutic use, Asia, Adjuvants, Immunologic, Triple Negative Breast Neoplasms drug therapy
- Abstract
Importance: In the phase 3 KEYNOTE-522 study, addition of pembrolizumab to neoadjuvant chemotherapy followed by adjuvant pembrolizumab significantly increased pathologic complete response (pCR) and event-free survival (EFS) vs neoadjuvant chemotherapy in patients with early triple-negative breast cancer., Objective: To evaluate efficacy and safety outcomes for patients enrolled in East/Southeast Asia (Asia) in KEYNOTE-522., Design, Setting, and Participants: KEYNOTE-522, a multicenter, double-blind, randomized clinical trial, enrolled 1174 patients between March 7, 2017, and September 13, 2018. For interim EFS and overall survival (OS) analyses (data cutoff, March 23, 2021), median follow-up was 39.8 months (range, 30.4-46.9 months) for pembrolizumab plus chemotherapy and 40.8 months (range, 30.1-46.9 months) for placebo plus chemotherapy. Data cutoff for pCR analysis was September 24, 2018. This secondary analysis included adults enrolled in Asia with newly diagnosed, previously untreated, nonmetastatic triple-negative breast cancer (tumor stage T1c and nodal stage N1-2 or tumor stage T2-4 and nodal stage N0-2) and Eastern Cooperative Oncology Group performance status of 0 to 1, regardless of programmed cell death ligand 1 (PD-L1) status., Intervention: Patients were randomized 2:1 to 4 cycles of pembrolizumab (200 mg every 3 weeks) or placebo plus carboplatin and paclitaxel and another 4 cycles of pembrolizumab or placebo plus doxorubicin or epirubicin and cyclophosphamide before surgery. After definitive surgery, patients received pembrolizumab or placebo every 3 weeks for 9 cycles or until recurrence or unacceptable toxic effects., Main Outcomes and Measures: The main outcome was pCR (no evidence of primary tumor after neoadjuvant therapy or carcinoma in situ after neoadjuvant therapy and no regional lymph node involvement after neoadjuvant therapy) at the time of definitive surgery and EFS., Results: A total of 216 of 1174 randomized patients (all female; median [range] age, 46.0 [24.0-71.0] years) were from Korea, Japan, Taiwan, and Singapore (136 in the pembrolizumab plus chemotherapy group and 80 in the placebo plus chemotherapy group). Of these patients, 104 (76.5%) in the pembrolizumab plus chemotherapy group and 60 (75.0%) in the placebo plus chemotherapy group had a tumor PD-L1 combined positive score of 1 or greater. Pathologic complete response was 58.7% (95% CI, 46.7%-69.9%) with pembrolizumab plus chemotherapy and 40.0% (95% CI, 26.4%-54.8%) with placebo plus chemotherapy; benefit was observed regardless of PD-L1 status. Thirteen patients (9.6%) in the pembrolizumab plus chemotherapy group and 20 patients (25.0%) in the placebo plus chemotherapy group had EFS events (hazard ratio, 0.35; 95% CI, 0.17-0.71). The 36-month EFS rate was 91.2% (95% CI, 85.0%-94.9%) with pembrolizumab plus chemotherapy and 77.2% (95% CI, 66.3%-85.0%) with placebo plus chemotherapy. Grade 3 to 4 treatment-related adverse events occurred in 109 patients (80.1%) receiving pembrolizumab plus chemotherapy and 64 patients (81.0%) receiving placebo plus chemotherapy., Conclusions and Relevance: In this subgroup analysis of patients enrolled in Asia in KEYNOTE-522, neoadjuvant pembrolizumab plus chemotherapy followed by adjuvant pembrolizumab led to clinically meaningful improvements in pCR and EFS vs neoadjuvant chemotherapy alone. These findings support the use of neoadjuvant pembrolizumab plus chemotherapy followed by adjuvant pembrolizumab as a standard-of-care therapy for patients in Asian countries with early triple-negative breast cancer., Trial Registration: ClinicalTrials.gov Identifier: NCT03036488.
- Published
- 2023
- Full Text
- View/download PDF
149. Vitamin D Insufficiency as a Risk Factor for Paclitaxel-Induced Peripheral Neuropathy in SWOG S0221.
- Author
-
Chen CS, Zirpoli G, Barlow WE, Budd GT, McKiver B, Pusztai L, Hortobagyi GN, Albain KS, Damaj MI, Godwin AK, Thompson A, Henry NL, Ambrosone CB, Stringer KA, and Hertz DL
- Subjects
- Humans, Female, Animals, Mice, Paclitaxel adverse effects, Prospective Studies, Vitamin D therapeutic use, Risk Factors, Peripheral Nervous System Diseases chemically induced, Peripheral Nervous System Diseases epidemiology, Breast Neoplasms complications, Breast Neoplasms drug therapy, Vitamin D Deficiency complications, Vitamin D Deficiency epidemiology, Antineoplastic Agents therapeutic use
- Abstract
Background: Prior work suggests that patients with vitamin D insufficiency may have a higher risk of chemotherapy-induced peripheral neuropathy (CIPN) from paclitaxel. The objective of this study was to validate vitamin D insufficiency as a CIPN risk factor., Methods: We used data and samples from the prospective phase III SWOG S0221 (ClinicalTrials.gov identifier: NCT00070564) trial that compared paclitaxel-containing chemotherapy regimens for early-stage breast cancer. We quantified pretreatment 25-hydroxy-vitamin D in banked serum samples using a liquid chromatography-tandem mass spectrometry targeted assay. We tested the association between vitamin D insufficiency (≤20 ng/mL) and grade ≥3 sensory CIPN via multiple logistic regression and then adjusted for self-reported race, age, body mass index, and paclitaxel schedule (randomization to weekly or every-2-week dosing). We also tested the direct effect of vitamin D deficiency on mechanical hypersensitivity in mice randomized to a regular or vitamin D-deficient diet., Results: Of the 1,191 female patients in the analysis, 397 (33.3%) had pretreatment vitamin D insufficiency, and 195 (16.4%) developed grade ≥3 CIPN. Patients with vitamin D insufficiency had a higher incidence of grade ≥3 CIPN than those who had sufficient vitamin D (20.7% vs 14.2%; odds ratio [OR], 1.57; 95% CI, 1.14-2.15; P=.005). The association retained significance after adjusting for age and paclitaxel schedule (adjusted OR, 1.65; 95% CI, 1.18-2.30; P=.003) but not race (adjusted OR, 1.39; 95% CI, 0.98-1.97; P=.066). In the mouse experiments, the vitamin D-deficient diet caused mechanical hypersensitivity and sensitized mice to paclitaxel (both P<.05)., Conclusions: Pretreatment vitamin D insufficiency is the first validated potentially modifiable predictive biomarker of CIPN from paclitaxel. Prospective trials are needed to determine whether vitamin D supplementation prevents CIPN and improves treatment outcomes in patients with breast and other cancer types.
- Published
- 2023
- Full Text
- View/download PDF
150. Molecular Characterization of HER2-Low Invasive Breast Carcinoma by Quantitative RT-PCR Using Oncotype DX Assay.
- Author
-
Lin HK, Can T, Kahn A, Flannery CA, Hoag J, Akkunuri A, Bailey H, Baehner R, Pusztai L, and Rozenblit M
- Subjects
- Humans, Female, Reverse Transcriptase Polymerase Chain Reaction, Reproducibility of Results, Receptors, Estrogen genetics, Neoplasm Recurrence, Local genetics, Neoplasm Recurrence, Local pathology, Biomarkers, Tumor genetics, Biomarkers, Tumor analysis, Prognosis, Receptor, ErbB-2 genetics, Breast Neoplasms genetics, Breast Neoplasms pathology
- Abstract
Background: HER2 immunohistochemistry (IHC) reproducibility is suboptimal for HER-low cases (IHC 1+ or 2+)., Methods: The Yale cohort included 214 stages I-II estrogen receptor positive breast cancers with IHC scores 0, 1+, and 2+ and routine Oncotype DX Recurrence Score (RS) results. The Exact Sciences (ES) cohort included 9 57 624 patients who had an Oncotype DX RS assay that assigns HER2-negative, equivocal, or positive status based on HER2 mRNA levels., Results: HER2 mRNA levels varied across IHC categories but with increasing medians of 9.10 (n = 89), 9.20 (n = 71), and 9.45 (n = 54) in IHC 0, 1+, and 2+, respectively. 22.4% of HER2-low (1+/2+) cancer had RS > 25. Over 98% of HER-low cancers were HER2-negative by Oncotype DX assignment., Conclusions: Cancers with higher mRNA levels exist within IHC 0 and low categories, most of the HER2-low patients by IHC have low RS indicating no benefit from current adjuvant chemotherapies., (© The Author(s) 2023. Published by Oxford University Press.)
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.