1
|
Paiva CE, Guimarães VS, Silva ATF, Maia YCDP, Paiva BSR, Reinert T, LeVasseur N. Navigating Endocrine Sensitivity Assessment in Nonmetastatic Breast Cancer Through Early On-Treatment Ki67 Understanding. Clin Breast Cancer 2025:S1526-8209(25)00111-9. [PMID: 40413085 DOI: 10.1016/j.clbc.2025.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 04/04/2025] [Accepted: 04/28/2025] [Indexed: 05/27/2025]
Abstract
In the landscape of breast cancer management, precise assessment of endocrine sensitivity significantly influences therapeutic strategies. The utilization of Ki67, an immunohistochemical marker of tumor proliferation, in tumor rebiopsy holds paramount importance in unraveling breast cancer's response to endocrine therapy. This narrative review aims to elucidate the pivotal role of Ki67 in endocrine sensitivity assessment. We explore the nuanced process of Ki67 evaluation, its timing, and its implications for clinical outcomes in breast cancer patients. From a clinical perspective to pathological response and risk of recurrence, we navigate the spectrum of Ki67's impact. By delving into these facets, we underscore Ki67's potential to guide personalized treatment strategies, shedding light on the intricate interplay between endocrine therapy and clinical outcomes in breast cancer patients.
Collapse
Affiliation(s)
- Carlos Eduardo Paiva
- Department of Clinical Oncology, Barretos Cancer Hospital, Barretos, SP, Brazil; Palliative Care and Quality of Life Research Group (GPQual), Barretos Cancer Hospital, Barretos, SP, Brazil.
| | - Vitor Souza Guimarães
- Palliative Care and Quality of Life Research Group (GPQual), Barretos Cancer Hospital, Barretos, SP, Brazil
| | - Alinne Tatiane Faria Silva
- Palliative Care and Quality of Life Research Group (GPQual), Barretos Cancer Hospital, Barretos, SP, Brazil
| | - Yara Cristina de Paiva Maia
- Nutrition and Molecular Biology Research Group, School of Medicine, Federal University of Uberlandia, Minas Gerais, Uberlandia, Brazil
| | - Bianca Sakamoto Ribeiro Paiva
- Palliative Care and Quality of Life Research Group (GPQual), Barretos Cancer Hospital, Barretos, SP, Brazil; Learning and Research Institute, Barretos Cancer Hospital, Barretos, SP, Brazil
| | - Tomás Reinert
- Centro de Oncologia Família Irineu Boff, Hospital Nora Teixeira, Santa Casa de Porto Alegre, Porto Alegre, RS, Brazil; Grupo Brasileiro de Estudos em Câncer de Mama (GBECAM), Porto Alegre, RS, Brazil
| | - Nathalie LeVasseur
- Division of Medical Oncology, BC Cancer, Vancouver, BC, Canada; Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
2
|
Zheng C, Zheng D, Zhang Y, Lin R, Zheng Z. A population‑based propensity score matching analysis of neoadjuvant compared to adjuvant chemotherapy in luminal breast cancer. Sci Rep 2025; 15:9568. [PMID: 40113956 PMCID: PMC11926370 DOI: 10.1038/s41598-025-93934-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
This study evaluated the long-term prognosis of patients with luminal (HR+/HER2-) breast cancer who underwent either neoadjuvant or adjuvant chemotherapy. Between January 2014 and December 2018, 1065 h+/HER2- breast cancer patients were retrospectively analyzed. Each patient receiving neoadjuvant chemotherapy was matched with two patients receiving adjuvant chemotherapy using a 1:2 propensity score matching (PSM) approach. After matching, 47 neoadjuvant chemotherapy patients and 89 adjuvant chemotherapy patients were included. The clinical and pathological characteristics of both groups were compared, and risk factors for postoperative events were assessed alongside a survival analysis. Following propensity score matching, the characteristics of the two groups were well balanced. The study identified lower progesterone receptor (PR) expression, histological grade III, and lymph node metastasis as independent risk factors for recurrence-free survival (RFS). No significant difference in RFS was observed between neoadjuvant and adjuvant chemotherapy. It is recommended that patients with HR+/HER2- breast cancer who exhibit a poor response to neoadjuvant chemotherapy should undergo early surgery, with personalized treatment decisions based on postoperative pathological findings.
Collapse
Affiliation(s)
- Chenhui Zheng
- Department of Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Danni Zheng
- Department of Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Yuzhu Zhang
- Department of Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Renzhi Lin
- Department of Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Zhibao Zheng
- Department of Surgical Oncology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China.
| |
Collapse
|
3
|
Das S, Pattnaik G, Pattanaik S, Jena BR, Satapathy BS, Pradhan A. Envisioning Clinical Management of Breast Cancer: a Comprehensive Review. Curr Drug Discov Technol 2025; 22:e290424229495. [PMID: 38685777 DOI: 10.2174/0115701638300812240417055802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/29/2024] [Accepted: 03/12/2024] [Indexed: 05/02/2024]
Abstract
BACKGROUND Background: Coming to the edge of disease manufacturing in the twenty-- first-century, breast cancer occupies a terrifying scenario in the globe, especially in adult women. Its curiosity endeavours remarkable advances made during the past decennaries for cancer treatment and diagnosis. OBJECTIVE It accounts for the fifth leading cause of transience, killing approximately 570,000 people per annum. To reduce the prognosis of clinical oncological development with the application of a new chemical entity, some of the critical challenges, like active pharmaceutical ingredients with high chemical resistance, extreme side effects, and high treatment costs are some of the limitations in the curbing aspects of breast melanoma. METHODS In cancer research, hence, the development of drugs that are safe, efficient, and cost-effective remains a 'Holy Grail' that may be considered as a boon to target the malignant tissues with novel therapeutics devices. RESULTS Through the findings on overcoming the drawbacks of traditional methods, researchers have given special attention to cancer-preventive and theranostic approaches based on some novel drug delivery systems. CONCLUSION The present study forecasts the wide-ranging modern applications, and on developing some novel liposomal drug delivery therapy against breast cancer.
Collapse
Affiliation(s)
- Shubhashree Das
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Jatani, 752050, Odisha, India
| | - Gurudutta Pattnaik
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Jatani, 752050, Odisha, India
| | - Sovan Pattanaik
- School of Pharmaceutical Sciences, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, Jatani, 752050, Odisha, India
| | - Bikash Ranjan Jena
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Jatani, 752050, Odisha, India
| | - Bhabani Sankar Satapathy
- School of Pharmaceutical Sciences, Siksha 'O' Anusandhan Deemed to be University, Bhubaneswar, Jatani, 752050, Odisha, India
| | - Ayushi Pradhan
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Jatani, 752050, Odisha, India
| |
Collapse
|
4
|
Xu P, Luo W, Hu J, Ma X, Hao Q, Hui W, Zhou Z, Lin S, Wang M, Wu H, Dai Z, Kang H. Favorable outcome of neoadjuvant endocrine treatment than surgery-first in female HR-positive/HER2-negative breast cancer patients-A NCDB analysis (2010-2016). Cancer Med 2024; 13:e7244. [PMID: 38859692 PMCID: PMC11165171 DOI: 10.1002/cam4.7244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 03/03/2024] [Accepted: 04/27/2024] [Indexed: 06/12/2024] Open
Abstract
PURPOSE To assess the efficacy of neoadjuvant endocrine therapy in female HR-positive/HER2-negative breast cancer patients. DATA AND METHODS We identified female patients aged ≥18 years with cT1-4N0-XM0, HR(+), and HER2(-) breast cancer from the National Cancer Database. The patients who underwent surgery first were categorized as "surgery-first," while those who received NET before surgery were classified as "NET." Propensity score-matching, Cox proportional-hazard model, variance inflation factors, and interaction analysis were employed to estimate the correlation between NET and survival outcomes. RESULTS Among 432,387 cases, 2914 NET patients and 2914 surgery-first patients were matched. Compared with the surgery-first group, the NET group received less adjuvant chemotherapy (p < 0.001). Furthermore, the NET group exhibited higher survival probabilities compared with the surgery-first group (3 years: 91.4% vs. 82.1%; 5 years: 82.1% vs. 66.8%). Multivariate Cox analysis indicated that NET was associated with improved OS (surgery-first vs. NET: HR 2.17, 95% CI: 1.93-2.44). Age over 55 years old, having public insurance, higher CDCC score, higher NSBR grade, ER(+)PR(-), and advanced clinical stage were related to worse OS (all p < 0.05). There was an interaction between age, race, income, and home and treatment regimen (all p < 0.05). CONCLUSION NET may be a more effective treatment procedure than surgery-first in female HR-positive/HER2-negative, non-metastatic breast cancer patients. Future clinical studies with more detailed data will provide higher-level evidence-based data.
Collapse
Affiliation(s)
- Peng Xu
- The Comprehensive Breast Care CenterThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Wen Luo
- The Comprehensive Breast Care CenterThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Jingjing Hu
- Massachusetts General Cancer CenterBostonMassachusettsUSA
| | - Xiaobin Ma
- The Comprehensive Breast Care CenterThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Qian Hao
- The Comprehensive Breast Care CenterThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Wentao Hui
- The Comprehensive Breast Care CenterThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Zhangjian Zhou
- The Comprehensive Breast Care CenterThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Shuai Lin
- The Comprehensive Breast Care CenterThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Meng Wang
- The Comprehensive Breast Care CenterThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| | - Hao Wu
- Department of Biophysics, School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to DiseasesXi'an Jiaotong University Health Science CenterXi'anShaanxiChina
| | - Zhijun Dai
- Department of Breast SurgeryThe First Affiliated Hospital, College of Medicine, Zhejiang UniversityHangzhouChina
| | - Huafeng Kang
- The Comprehensive Breast Care CenterThe Second Affiliated Hospital of Xi'an Jiaotong UniversityXi'anShaanxiChina
| |
Collapse
|
5
|
Jayaraman S, Wu X, Kalari KR, Tang X, Kuffel MJ, Bruinsma ES, Jalali S, Peterson KL, Correia C, Kudgus RA, Kaufmann SH, Renuse S, Ingle JN, Reid JM, Ames MM, Fields AP, Schellenberg MJ, Hawse JR, Pandey A, Goetz MP. Endoxifen downregulates AKT phosphorylation through protein kinase C beta 1 inhibition in ERα+ breast cancer. NPJ Breast Cancer 2023; 9:101. [PMID: 38114522 PMCID: PMC10730845 DOI: 10.1038/s41523-023-00606-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 12/01/2023] [Indexed: 12/21/2023] Open
Abstract
Endoxifen, a secondary tamoxifen metabolite, is a potent antiestrogen exhibiting estrogen receptor alpha (ERα) binding at nanomolar concentrations. Phase I/II clinical trials identified clinical activity of Z-endoxifen (ENDX), in endocrine-refractory metastatic breast cancer as well as ERα+ solid tumors, raising the possibility that ENDX may have a second, ERα-independent, mechanism of action. An unbiased mass spectrometry approach revealed that ENDX concentrations achieved clinically with direct ENDX administration (5 µM), but not low concentrations observed during tamoxifen treatment (<0.1 µM), profoundly altered the phosphoproteome of the aromatase expressing MCF7AC1 cells with limited impact on the total proteome. Computational analysis revealed protein kinase C beta (PKCβ) and protein kinase B alpha or AKT1 as potential kinases responsible for mediating ENDX effects on protein phosphorylation. ENDX more potently inhibited PKCβ1 kinase activity compared to other PKC isoforms, and ENDX binding to PKCβ1 was confirmed using Surface Plasma Resonance. Under conditions that activated PKC/AKT signaling, ENDX induced PKCβ1 degradation, attenuated PKCβ1-activated AKTSer473 phosphorylation, diminished AKT substrate phosphorylation, and induced apoptosis. ENDX's effects on AKT were phenocopied by siRNA-mediated PKCβ1 knockdown or treatment with the pan-AKT inhibitor, MK-2206, while overexpression of constitutively active AKT diminished ENDX-induced apoptosis. These findings, which identify PKCβ1 as an ENDX target, indicate that PKCβ1/ENDX interactions suppress AKT signaling and induce apoptosis in breast cancer.
Collapse
Affiliation(s)
| | - Xinyan Wu
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Krishna R Kalari
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Xiaojia Tang
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, 55905, USA
| | - Mary J Kuffel
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Elizabeth S Bruinsma
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Shahrzad Jalali
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | | | - Cristina Correia
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Rachel A Kudgus
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Scott H Kaufmann
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Santosh Renuse
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - James N Ingle
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Joel M Reid
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Matthew M Ames
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Alan P Fields
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL, 32224, USA
| | - Matthew J Schellenberg
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - John R Hawse
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Cancer Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Matthew P Goetz
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
6
|
Mueller S, Grote I, Bartels S, Kandt L, Christgen H, Lehmann U, Gluz O, Graeser M, Kates R, Harbeck N, Kreipe H, Christgen M. p53 Expression in Luminal Breast Cancer Correlates With TP53 Mutation and Primary Endocrine Resistance. Mod Pathol 2023; 36:100100. [PMID: 36788081 DOI: 10.1016/j.modpat.2023.100100] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 01/02/2023] [Accepted: 01/05/2023] [Indexed: 01/13/2023]
Abstract
TP53 mutation is associated with primary endocrine resistance in luminal breast cancer (BC). Nuclear accumulation of p53, as determined by immunohistochemistry (IHC), is a surrogate marker for TP53 mutation. The immunohistochemical p53 index that defines a p53-positive status is not well established. This study determined the optimal p53 index cutoff to identify luminal BCs harboring TP53 mutations. In total, 364 luminal BCs from the West German Study Group ADAPT trial (NCT01779206) were analyzed for TP53 mutations by next-generation sequencing and for p53 expression by IHC (DO-7 antibody). P53 indices were determined by automated image analysis. All tumors were from patients treated with short-term preoperative endocrine therapy (pET; tamoxifen or aromatase inhibitor) before tumor resection. IHC evaluation included needle biopsies before therapy (baseline) and resections specimens after therapy (post-pET). Optimal p53 index cutoffs were defined with Youden statistics. TP53 mutations were detected in 16.3% of BC cases. The median p53 indices were significantly higher in TP53-mutated BCs compared to BCs harboring wild-type TP53 (baseline: 47.0% vs 6.4%, P < .001; post-pET: 50.1% vs 1.1%, P < .001). Short-term pET decreased p53 indices in BCs harboring wild-type TP53 (P < .001) but not in TP53-mutated BCs (P = .102). For baseline biopsies, the optimal p53 index cutoff was ≥34.6% (specificity 0.92, sensitivity 0.63, Youden index 0.54, accuracy: 0.87). For post-pET specimens, the optimal cutoff was ≥25.3% (specificity 0.95, sensitivity 0.65, Youden index 0.60, accuracy: 0.90). Using these cutoffs to define the p53 status, p53-positive BCs were >2-fold more common in pET nonresponders compared to pET responders (baseline: 37/162, 22.8% vs 18/162, 11.1%, P = .007; post-pET: 36/179, 20.1% vs 16/179, 8.9%, P = .004). In summary, IHC for p53 identifies TP53-mutated luminal BCs with high specificity and accuracy. Optimal cutoffs are ≥35% and ≥25% for treatment-naïve and endocrine-pretreated patients, respectively.
Collapse
Affiliation(s)
- Sophie Mueller
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Isabel Grote
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Stephan Bartels
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Leonie Kandt
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | | | - Ulrich Lehmann
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Oleg Gluz
- West German Study Group, Moenchengladbach, Germany; Ev. Bethesda Hospital, Moenchengladbach, Germany; Women's Clinic and Breast Center, University Clinics Cologne, Cologne, Germany
| | - Monika Graeser
- West German Study Group, Moenchengladbach, Germany; Ev. Bethesda Hospital, Moenchengladbach, Germany; Department of Gynecology, University Medical Center Hamburg, Hamburg, Germany
| | - Ron Kates
- West German Study Group, Moenchengladbach, Germany
| | - Nadia Harbeck
- West German Study Group, Moenchengladbach, Germany; Department of OB&GYN and CCC Munich, Breast Center, LMU University Hospital, Munich, Germany
| | - Hans Kreipe
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | | |
Collapse
|
7
|
Martínez-Pérez C, Turnbull AK, Kay C, Dixon JM. Neoadjuvant endocrine therapy in postmenopausal women with HR+/HER2- breast cancer. Expert Rev Anticancer Ther 2023; 23:67-86. [PMID: 36633402 DOI: 10.1080/14737140.2023.2162043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/20/2022] [Indexed: 01/13/2023]
Abstract
INTRODUCTION While endocrine therapy is the standard-of-care adjuvant treatment for hormone receptor-positive (HR+) breast cancers, there is also extensive evidence for the role of pre-operative (or neoadjuvant) endocrine therapy (NET) in HR+ postmenopausal women. AREAS COVERED We conducted a thorough review of the published literature, to summarize the evidence to date, including studies of how NET compares to neoadjuvant chemotherapy, which NET agents are preferable, and the optimal duration of NET. We describe the importance of on-treatment assessment of response, the different predictors available (including Ki67, PEPI score, and molecular signatures) and the research opportunities the pre-operative setting offers. We also summarize recent combination trials and discuss how the COVID-19 pandemic led to increases in NET use for safe management of cases with deferred surgery and adjuvant treatments. EXPERT OPINION NET represents a safe and effective tool for the management of postmenopausal women with HR+/HER2- breast cancer, enabling disease downstaging and a wider range of surgical options. Aromatase inhibitors are the preferred NET, with evidence suggesting that longer regimens might yield optimal results. However, NET remains currently underutilised in many territories and institutions. Further validation of predictors for treatment response and benefit is needed to help standardise and fully exploit the potential of NET in the clinic.
Collapse
Affiliation(s)
- Carlos Martínez-Pérez
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
- Edinburgh Breast Cancer Now Research Team, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
| | - Arran K Turnbull
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
- Edinburgh Breast Cancer Now Research Team, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
| | - Charlene Kay
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
- Edinburgh Breast Cancer Now Research Team, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
| | - J Michael Dixon
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
- Edinburgh Breast Cancer Now Research Team, MRC Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland
- Edinburgh Breast Unit, Western General Hospital, Edinburgh, Scotland
| |
Collapse
|
8
|
Bergamino MA, López-Knowles E, Morani G, Tovey H, Kilburn L, Schuster EF, Alataki A, Hills M, Xiao H, Holcombe C, Skene A, Robertson JF, Smith IE, Bliss JM, Dowsett M, Cheang MCU. HER2-enriched subtype and novel molecular subgroups drive aromatase inhibitor resistance and an increased risk of relapse in early ER+/HER2+ breast cancer. EBioMedicine 2022; 83:104205. [PMID: 35985932 PMCID: PMC9482930 DOI: 10.1016/j.ebiom.2022.104205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 07/22/2022] [Accepted: 07/22/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Oestrogen receptor positive/ human epidermal growth factor receptor positive (ER+/HER2+) breast cancers (BCs) are less responsive to endocrine therapy than ER+/HER2- tumours. Mechanisms underpinning the differential behaviour of ER+HER2+ tumours are poorly characterised. Our aim was to identify biomarkers of response to 2 weeks' presurgical AI treatment in ER+/HER2+ BCs. METHODS All available ER+/HER2+ BC baseline tumours (n=342) in the POETIC trial were gene expression profiled using BC360™ (NanoString) covering intrinsic subtypes and 46 key biological signatures. Early response to AI was assessed by changes in Ki67 expression and residual Ki67 at 2 weeks (Ki672wk). Time-To-Recurrence (TTR) was estimated using Kaplan-Meier methods and Cox models adjusted for standard clinicopathological variables. New molecular subgroups (MS) were identified using consensus clustering. FINDINGS HER2-enriched (HER2-E) subtype BCs (44.7% of the total) showed poorer Ki67 response and higher Ki672wk (p<0.0001) than non-HER2-E BCs. High expression of ERBB2 expression, homologous recombination deficiency (HRD) and TP53 mutational score were associated with poor response and immune-related signatures with High Ki672wk. Five new MS that were associated with differential response to AI were identified. HER2-E had significantly poorer TTR compared to Luminal BCs (HR 2.55, 95% CI 1.14-5.69; p=0.0222). The new MS were independent predictors of TTR, adding significant value beyond intrinsic subtypes. INTERPRETATION Our results show HER2-E as a standardised biomarker associated with poor response to AI and worse outcome in ER+/HER2+. HRD, TP53 mutational score and immune-tumour tolerance are predictive biomarkers for poor response to AI. Lastly, novel MS identify additional non-HER2-E tumours not responding to AI with an increased risk of relapse. FUNDING Cancer Research UK (CRUK/07/015).
Collapse
Affiliation(s)
- Milana A Bergamino
- Clinical Trials and Statistics Unit (ICR-CTSU)- Division of Clinical Studies, The Institute of Cancer Research, London, UK
| | - Elena López-Knowles
- Royal Marsden Hospital, London, UK; The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Gabriele Morani
- Clinical Trials and Statistics Unit (ICR-CTSU)- Division of Clinical Studies, The Institute of Cancer Research, London, UK
| | - Holly Tovey
- Clinical Trials and Statistics Unit (ICR-CTSU)- Division of Clinical Studies, The Institute of Cancer Research, London, UK
| | - Lucy Kilburn
- Clinical Trials and Statistics Unit (ICR-CTSU)- Division of Clinical Studies, The Institute of Cancer Research, London, UK
| | - Eugene F Schuster
- Royal Marsden Hospital, London, UK; The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Anastasia Alataki
- Royal Marsden Hospital, London, UK; The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | | | - Hui Xiao
- Clinical Trials and Statistics Unit (ICR-CTSU)- Division of Clinical Studies, The Institute of Cancer Research, London, UK; Royal Marsden Hospital, London, UK
| | - Chris Holcombe
- Liverpool University Hospitals Foundation Trust, Liverpool, UK
| | | | - John F Robertson
- Faculty of Medicine & Health Sciences, Queen's Medical Centre, Nottingham, UK
| | | | - Judith M Bliss
- Clinical Trials and Statistics Unit (ICR-CTSU)- Division of Clinical Studies, The Institute of Cancer Research, London, UK
| | | | - Maggie C U Cheang
- Clinical Trials and Statistics Unit (ICR-CTSU)- Division of Clinical Studies, The Institute of Cancer Research, London, UK.
| |
Collapse
|
9
|
Alataki A, Dowsett M. Human epidermal growth factor receptor-2 and endocrine resistance in hormone-dependent breast cancer. Endocr Relat Cancer 2022; 29:R105-R122. [PMID: 35613334 PMCID: PMC9254309 DOI: 10.1530/erc-21-0293] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 05/25/2022] [Indexed: 12/27/2022]
Abstract
Endocrine therapies are the main treatment strategies for the clinical management of hormone-dependent breast cancer. Despite prolonged time to recurrence in the adjuvant setting and the initial clinical responses in the metastatic setting, many patients eventually encounter tumour relapse due to acquired resistance to these agents. Other patients experience a lack of tumour regression at the beginning of treatment indicating de novo resistance that significantly limits its efficacy in the clinic. There is compelling evidence that human epidermal growth factor receptor-2 (HER2) overexpression contributes to resistance to endocrine therapies in oestrogen receptor-positive (ER+) breast cancer. ER+/HER2+ tumours comprise about 10% of all breast cancer cases and about 60% of the whole set of HER2+ tumours. Most patients with primary ER+/HER2+ disease will receive antibody-based HER2-targeted therapy, but this is generally for no more than one year while endocrine treatment is usually for at least 5 years. A number of HER2-kinase inhibitors are also now in clinical use or in clinical trials, and the interaction of these with endocrine treatment may differ from that of antibody treatment. In this review article, we aim to summarise knowledge on molecular mechanisms of breast cancer resistance to endocrine therapies attributable to the impact of HER2 signalling on endocrine sensitivity, to discuss data from clinical trials addressing the role of HER2 in the development of endocrine resistance in the metastatic, neoadjuvant and adjuvant settings and to explore rational new therapeutic strategies.
Collapse
Affiliation(s)
- Anastasia Alataki
- Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital and The Institute of Cancer Research, London, UK
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
- Correspondence should be addressed to A Alataki:
| | - Mitch Dowsett
- Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital and The Institute of Cancer Research, London, UK
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| |
Collapse
|
10
|
Estrogen receptor inhibition mediates radiosensitization of ER-positive breast cancer models. NPJ Breast Cancer 2022; 8:31. [PMID: 35273179 PMCID: PMC8913671 DOI: 10.1038/s41523-022-00397-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 02/03/2022] [Indexed: 11/28/2022] Open
Abstract
Endocrine therapy (ET) is an effective first-line therapy for women with estrogen receptor-positive (ER + ) breast cancers. While both ionizing radiation (RT) and ET are used for the treatment of women with ER+ breast cancer, the most effective sequencing of therapy and the effect of ET on tumor radiosensitization remains unclear. Here we sought to understand the effects of inhibiting estrogen receptor (ER) signaling in combination with RT in multiple preclinical ER+ breast cancer models. Clonogenic survival assays were performed using variable pre- and post-treatment conditions to assess radiosensitization with estradiol, estrogen deprivation, tamoxifen, fulvestrant, or AZD9496 in ER+ breast cancer cell lines. Estrogen stimulation was radioprotective (radiation enhancement ratios [rER]: 0.51–0.82). Conversely, when given one hour prior to RT, ER inhibition or estrogen depletion radiosensitized ER+ MCF-7 and T47D cells (tamoxifen rER: 1.50–1.60, fulvestrant rER: 1.76–2.81, AZD9496 rER: 1.33–1.48, estrogen depletion rER: 1.47–1.51). Combination treatment resulted in an increase in double-strand DNA (dsDNA) breaks as a result of inhibition of non-homologous end joining-mediated dsDNA break repair with no effect on homologous recombination. Treatment with tamoxifen or fulvestrant in combination with RT also increased the number of senescent cells but did not affect apoptosis or cell cycle distribution. Using an MCF-7 xenograft model, concurrent treatment with tamoxifen and RT was synergistic and resulted in a significant decrease in tumor volume and a delay in time to tumor doubling without significant toxicity. These findings provide preclinical evidence that concurrent treatment with ET and RT may be an effective radiosensitization strategy.
Collapse
|
11
|
Dowsett M, Kilburn L, Rimawi MF, Osborne CK, Pogue-Geile K, Liu Y, Jacobs SA, Finnigan M, Puhalla S, Dodson A, Martins V, Cheang M, Perry S, Holcombe C, Turner N, Swift C, Bliss JM, Johnston S, on behalf of the PALLET trialists. Biomarkers of Response and Resistance to Palbociclib Plus Letrozole in Patients With ER +/HER2 - Breast Cancer. Clin Cancer Res 2022; 28:163-174. [PMID: 34645649 PMCID: PMC9632606 DOI: 10.1158/1078-0432.ccr-21-1628] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/13/2021] [Accepted: 10/07/2021] [Indexed: 01/07/2023]
Abstract
PURPOSE To determine (i) the relationship between candidate biomarkers of the antiproliferative (Ki67) response to letrozole and palbociclib alone and combined in ER+/HER2- breast cancer; and (ii) the pharmacodynamic effect of the agents on the biomarkers. EXPERIMENTAL DESIGN 307 postmenopausal women with ER+/HER2- primary breast cancer were randomly assigned to neoadjuvant treatment with letrozole for 14 weeks; letrozole for 2 weeks, then letrozole+palbociclib to 14 weeks; palbociclib for 2 weeks, then letrozole+palbociclib to 14 weeks; or letrozole+palbociclib for 14 weeks. Biopsies were taken at baseline, 2 and 14 weeks and surgery at varying times after stopping palbociclib. Immunohistochemical analyses were conducted for Ki67, c-PARP, ER, PgR, RB1, CCNE1, and CCND1. RESULTS Higher baselines ER and PgR were significantly associated with a greater chance of complete cell-cycle arrest (CCCA: Ki67 <2.7%) at 14 weeks and higher baseline Ki67, c-PARP, and CCNE1 with a lower chance. The interaction with treatment was significant only for c-PARP. CCND1 levels were decreased c.20% by letrozole at 2 and 14 weeks but showed a tendency to increase with palbociclib. CCNE1 levels fell 82% (median) in tumors showing CCCA but were unchanged in those with no CCCA. Only 2/9 tumors showed CCCA 3-9 days after stopping palbociclib. ESR1 mutations were found in 2/4 tumors for which surgery took place ≥6 months after starting treatment. CONCLUSIONS High CCNE1 levels were confirmed as a biomarker of resistance to letrozole+palbociclib. Ki67 recovery within 3-9 days of discontinuing palbociclib indicates incomplete suppression of proliferation during the "off" week of its schedule.
Collapse
Affiliation(s)
- Mitch Dowsett
- Royal Marsden Hospital, London, United Kingdom.,Breast Cancer Now Toby Robins Center for Breast Cancer Research, Institute of Cancer Research, London, United Kingdom.,Corresponding Author: Mitch Dowsett, Royal Marsden Hospital, London SW3 6JJ, UK. Phone: 44-207-808-2884; E-mail:
| | - Lucy Kilburn
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, Sutton, United Kingdom
| | | | | | | | | | | | | | - Shannon Puhalla
- NSABP Foundation, Pittsburgh, Pennsylvania.,University of Pittsburgh Medical Center Cancer Center, Pittsburgh, Pennsylvania
| | | | | | - Maggie Cheang
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, Sutton, United Kingdom
| | - Sophie Perry
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, Sutton, United Kingdom
| | - Chris Holcombe
- Royal Liverpool and Broadgreen University Hospitals National Health Service Trust, Liverpool, United Kingdom
| | - Nick Turner
- Royal Marsden Hospital, London, United Kingdom.,Breast Cancer Now Toby Robins Center for Breast Cancer Research, Institute of Cancer Research, London, United Kingdom
| | - Claire Swift
- Royal Marsden Hospital, London, United Kingdom.,Breast Cancer Now Toby Robins Center for Breast Cancer Research, Institute of Cancer Research, London, United Kingdom
| | - Judith M. Bliss
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, Sutton, United Kingdom
| | | | | |
Collapse
|
12
|
Integrating CDK4/6 inhibitors in the treatment of patients with early breast cancer. Breast 2021; 62 Suppl 1:S70-S79. [PMID: 34930649 PMCID: PMC9097805 DOI: 10.1016/j.breast.2021.12.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 12/07/2021] [Accepted: 12/12/2021] [Indexed: 12/13/2022] Open
Abstract
CDK4/6 inhibitors have an established role in the treatment of hormone receptor positive HER2-negative advanced breast cancer. All studies conducted in metastatic breast cancer showed a benefit in delaying progression when added to standard endocrine therapy, regardless of therapy line, pretreatment, menopausal status, site of metastasis, CDK4/6 inhibitor used and associated endocrine therapy. A benefit in overall survival has also been demonstrated. In early breast cancer, only the MonarchE study has shown an improved invasive disease-free survival with abemaciclib taken for 2 years, whereas the Penelope-B did not meet the primary endpoint and the PALLAS study was terminated early for futility. Studies conducted in the neoadjuvant setting might help to explain the discordant results.
CDK4/6 inhibitors increase PFS in advanced breast cancer in all subgroups. 2-years abemaciclib added to endocrine therapy improves invasive disease-free survival in high-risk breast cancer. Palbociclib did not improve invasive disease-free survival in early breast cancer.
Collapse
|
13
|
Grote I, Bartels S, Kandt L, Bollmann L, Christgen H, Gronewold M, Raap M, Lehmann U, Gluz O, Nitz U, Kuemmel S, Zu Eulenburg C, Braun M, Aktas B, Grischke EM, Schumacher C, Luedtke-Heckenkamp K, Kates R, Wuerstlein R, Graeser M, Harbeck N, Christgen M, Kreipe H. TP53 mutations are associated with primary endocrine resistance in luminal early breast cancer. Cancer Med 2021; 10:8581-8594. [PMID: 34779146 PMCID: PMC8633262 DOI: 10.1002/cam4.4376] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 09/10/2021] [Accepted: 09/13/2021] [Indexed: 12/19/2022] Open
Abstract
Background Whereas the genomic landscape of endocrine‐resistant breast cancer has been intensely characterized in previously treated cases with local or distant recurrence, comparably little is known about genomic alterations conveying primary non‐responsiveness to endocrine treatment in luminal early breast cancer. Methods In this study, 622 estrogen receptor‐expressing breast cancer cases treated with short‐term preoperative endocrine therapy (pET) from the WSG‐ADAPT trial (NCT01779206) were analyzed for genetic alterations associated with impaired endocrine proliferative response (EPR) to 3‐week pET with tamoxifen or aromatase inhibitors. EPR was categorized as optimal (post‐pET Ki67 <10%) versus slightly, moderately, and severely impaired (post‐pET Ki67 10%–19%, 20%–34%, and ≥35%, respectively). Recently described gene mutations frequently found in previously treated advanced breast cancer were analyzed (ARID1A, BRAF, ERBB2, ESR1, GATA3, HRAS, KRAS, NRAS, PIK3CA, and TP53) by next‐generation sequencing. Amplifications of CCND1, FGFR1, ERBB2, and PAK1 were determined by digital PCR or fluorescence in situ hybridization. Results ERBB2 amplification (p = 0.0015) and mutations of TP53 (p < 0.0001) were significantly associated with impaired EPR. Impaired EPR in TP53‐mutated breast cancer cases was independent from the Oncotype DX Recurrence Score group and was seen both with tamoxifen‐ and aromatase inhibitor‐based pET (p = 0.0005 each). Conclusion We conclude that impaired EPR to pET is suitable to identify cases with primary endocrine resistance in early luminal breast cancer and that TP53‐mutated luminal cancers might not be sufficiently treated by endocrine therapy alone.
Collapse
Affiliation(s)
- Isabel Grote
- Hannover Medical School, Institute of Pathology, Hannover, Germany
| | - Stephan Bartels
- Hannover Medical School, Institute of Pathology, Hannover, Germany
| | - Leonie Kandt
- Hannover Medical School, Institute of Pathology, Hannover, Germany
| | - Laura Bollmann
- Hannover Medical School, Institute of Pathology, Hannover, Germany
| | | | - Malte Gronewold
- Hannover Medical School, Institute of Pathology, Hannover, Germany
| | - Mieke Raap
- Hannover Medical School, Institute of Pathology, Hannover, Germany
| | - Ulrich Lehmann
- Hannover Medical School, Institute of Pathology, Hannover, Germany
| | - Oleg Gluz
- West German Study Group, Moenchengladbach, Germany.,Ev. Bethesda Hospital, Moenchengladbach, Germany.,University Clinics Cologne, Women's Clinic and Breast Center, Cologne, Germany
| | - Ulrike Nitz
- West German Study Group, Moenchengladbach, Germany.,Ev. Bethesda Hospital, Moenchengladbach, Germany
| | - Sherko Kuemmel
- West German Study Group, Moenchengladbach, Germany.,Clinics Essen-Mitte, Breast Unit, Essen, Germany.,Charité, Women's Clinic, Berlin, Germany
| | | | | | - Bahriye Aktas
- University Clinics Essen, Women's Clinic, Essen, Germany.,University Clinics Leipzig, Women's Clinic, Leipzig, Germany
| | | | | | | | - Ronald Kates
- West German Study Group, Moenchengladbach, Germany
| | - Rachel Wuerstlein
- Department OB&GYN and CCC Munich, LMU University Hospital, Breast Center, Munich, Germany
| | - Monika Graeser
- West German Study Group, Moenchengladbach, Germany.,Ev. Bethesda Hospital, Moenchengladbach, Germany.,Department of Gynecology, University Medical Center Hamburg, Hamburg, Germany
| | - Nadia Harbeck
- West German Study Group, Moenchengladbach, Germany.,Department OB&GYN and CCC Munich, LMU University Hospital, Breast Center, Munich, Germany
| | | | - Hans Kreipe
- Hannover Medical School, Institute of Pathology, Hannover, Germany
| |
Collapse
|
14
|
Adtani PN, Narasimhan M, Girija DM. In vitro anticancer activity of a pentacyclic triterpenoid via the mitochondrial pathway in bone-invasive oral squamous cell carcinoma. J Oral Maxillofac Pathol 2021; 25:313-321. [PMID: 34703127 PMCID: PMC8491335 DOI: 10.4103/0973-029x.325234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 06/17/2021] [Accepted: 07/06/2021] [Indexed: 11/27/2022] Open
Abstract
Context: Oral cancer is the most dreadful cancer worldwide with a 5-year survival rate of approximately 50%. Anticancer therapies such as chemotherapy and radiotherapy result in severe side effects. Aim: We aimed to evaluate the in vitro anticancer activity of Asiatic acid (AA) on bone-invasive oral squamous cell carcinoma (BHY) cell line. Settings and Design: This was an in vitro laboratory setting. Materials and Methods: BHY cell lines were used for the experiment. Confocal microscopy was used to observe cellular alterations. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay was performed to determine the IC50 concentration of AA and flow cytometry to analyze the percentage of cells in each phase of the cell cycle post treatment. Immunoblot assays and semiquantitative reverse transcriptase-polymerase chain reaction (rt-PCR) were used to study the expression level of genes involved. Statistical Analysis Used: Student's t-test and one-way analysis of variance were used for statistical analysis. Results: IC50 concentration of AA was 15.6 μM. On flow cytometry analysis, treatment with 15.6 μM and 31.25 μM of AA for 24 h increased the percentage of cells in the G2/M phase to 45.63% and 53.12%, respectively, compared to 9.62% in control group. Immunoblot analysis and semiquantitative rt-PCR demonstrated an upregulation of p53, cyclin-dependent kinase inhibitors (p21 and p27), caspase-3, caspase-9, cytochrome c and Bax in a time-dependent manner and downregulation of cyclins and anti-apoptotic protein Bcl-2 (**P < 0.05, ***P < 0.001 versus control) post AA treatment. Conclusion: AA induces apoptosis via the mitochondrial-dependent pathway and causes cell cycle arrest at the G2/M phase in BHY cell line.
Collapse
Affiliation(s)
- Pooja Narain Adtani
- Department of Basic Medical and Dental Sciences, College of Dentistry, Gulf Medical University, Ajman, U.A.E
| | - Malathi Narasimhan
- Department of Oral Pathology and Microbiology, Faculty of Dental Sciences, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | - Dinesh M Girija
- Department of Cell Biology, Research and Development, Vopec Pharmaceuticals Pvt., Ltd., Chennai, Tamil Nadu, India
| |
Collapse
|
15
|
Hong K, Yao L, Sheng X, Ye D, Guo Y. Neoadjuvant Therapy of Cyclin-Dependent Kinase 4/6 Inhibitors Combined with Endocrine Therapy in HR+/HER2- Breast Cancer: A Systematic Review and Meta-Analysis. Oncol Res Treat 2021; 44:557-567. [PMID: 34515204 DOI: 10.1159/000518573] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 07/19/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Cyclin-dependent kinase (CDK) 4/6 inhibitors have been advocated for adjuvant therapy of metastatic hormone receptor (HR)+/human epidermal growth factor receptor 2 (HER2)- breast cancer (BC). However, the efficiency of adding CDK 4/6 inhibitors to neoadjuvant therapy was not unequivocal. OBJECTIVE The aim of the study was to evaluate the efficiency and toxicity of neoadjuvant CDK 4/6 inhibitors + endocrine therapy (ET) versus neoadjuvant endocrine monotherapy or standard neoadjuvant chemotherapy in HR+/HER2- BC. METHOD We searched PubMed, the Cochrane Library, Web of Science, and Embase online databases for randomized controlled trials and single-arm studies written in English until April 2021. RESULTS Five studies comparing CDK 4/6 inhibitors + ET as neoadjuvant treatments to ET alone and 2 studies comparing neoadjuvant CDK 4/6 inhibitors + ET to neoadjuvant chemotherapy were analysed. Neoadjuvant CDK 4/6 inhibitors + ET improved the rate of complete cell cycle arrest (CCCA: central Ki67 < 2.7%, odds ratio [OR] = 7.91, 95% confidence interval [CI] = 4.81-13.03, p < 0.001), increased the risk of adverse events (AEs; especially ≥3 AEs; AEs of all grades: OR = 9.10, 95% CI = 2.39-34.58, p = 0.001; AEs ≥3: OR = 12.24, 95% CI = 4.17-35.88, p < 0.001), led to no significant differences in pathological complete response (pCR) in patients with BC (OR = 0.34, 95% CI = 0.04-2.85, p = 0.318) compared to endocrine monotherapy. Moreover, subgroup analysis showed that the 3 types of CDK 4/6 inhibitors all improved the rate of CCCA (ribociclib: OR = 10.31, 95% CI = 3.59-29.61, p < 0.001; palbociclib: OR = 7.39, 95% CI = 1.26-43.40, p = 0.027, and abemaciclib: OR = 8.28, 95% CI = 3.41-20.11, p < 0.001). Compared to neoadjuvant chemotherapy, neoadjuvant CDK 4/6 inhibitors plus ET decreased the risk of AEs ≥3 (OR = 0.50, 95% CI = 0.29-0.87, p = 0.015) and showed similar ability to reach pCR (OR = 0.50, 95% CI = 0.12-2.07, p = 0.342) and reduce the residual cancer burden (RCB, RCB 0-1: OR = 0.47, 95% CI = 0.18-1.22, p = 0.121; RCB 2-3: OR = 2.30, 95% CI = 0.89-5.91, p = 0.084). CONCLUSIONS The results suggested that combination therapy had increased efficacy and toxicity compared to endocrine monotherapy and showed similar efficacy to and better safety than neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Kai Hong
- Medicine School, Ningbo University, Ningbo, China
| | - Lingli Yao
- Medicine School, Ningbo University, Ningbo, China
| | - Xianneng Sheng
- Department of Thyroid and Breast Surgery, Ningbo City First Hospital, Ningbo, China
| | - Dan Ye
- Medicine School, Ningbo University, Ningbo, China
| | - Yu Guo
- Department of Thyroid and Breast Surgery, Ningbo City First Hospital, Ningbo, China
| |
Collapse
|
16
|
Smith I, Robertson J, Kilburn L, Wilcox M, Evans A, Holcombe C, Horgan K, Kirwan C, Mallon E, Sibbering M, Skene A, Vidya R, Cheang M, Banerji J, Morden J, Sidhu K, Dodson A, Bliss JM, Dowsett M. Long-term outcome and prognostic value of Ki67 after perioperative endocrine therapy in postmenopausal women with hormone-sensitive early breast cancer (POETIC): an open-label, multicentre, parallel-group, randomised, phase 3 trial. Lancet Oncol 2020; 21:1443-1454. [PMID: 33152284 PMCID: PMC7606901 DOI: 10.1016/s1470-2045(20)30458-7] [Citation(s) in RCA: 191] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/16/2020] [Accepted: 07/16/2020] [Indexed: 12/25/2022]
Abstract
Background Preoperative and perioperative aromatase inhibitor (POAI) therapy has the potential to improve outcomes in women with operable oestrogen receptor-positive primary breast cancer. It has also been suggested that tumour Ki67 values after 2 weeks (Ki672W) of POAI predicts individual patient outcome better than baseline Ki67 (Ki67B). The POETIC trial aimed to test these two hypotheses. Methods POETIC was an open-label, multicentre, parallel-group, randomised, phase 3 trial (done in 130 UK hospitals) in which postmenopausal women aged at least 50 years with WHO performance status 0–1 and hormone receptor-positive, operable breast cancer were randomly assigned (2:1) to POAI (letrozole 2·5 mg per day orally or anastrozole 1 mg per day orally) for 14 days before and following surgery or no POAI (control). Adjuvant treatment was given as per UK standard local practice. Randomisation was done centrally by computer-generated permuted block method (variable block size of six or nine) and was stratified by hospital. Treatment allocation was not masked. The primary endpoint was time to recurrence. A key second objective explored association between Ki67 (dichotomised at 10%) and disease outcomes. The primary analysis for clinical endpoints was by modified intention to treat (excluding patients who withdrew consent). For Ki67 biomarker association and endpoint analysis, the evaluable population included all randomly assigned patients who had paired Ki67 values available. This study is registered with ClinicalTrials.gov, NCT02338310; the European Clinical Trials database, EudraCT2007-003877-21; and the ISRCTN registry, ISRCTN63882543. Recruitment is complete and long-term follow-up is ongoing. Findings Between Oct 13, 2008, and April 16, 2014, 4480 women were recruited and randomly assigned to POAI (n=2976) or control (n=1504). On Feb 6, 2018, median follow-up was 62·9 months (IQR 58·1–74·1). 434 (10%) of 4480 women had a breast cancer recurrence (280 [9%] POAI; 154 [10%] control), hazard ratio 0·92 (95% CI 0·75–1·12); p=0·40 with the proportion free from breast cancer recurrence at 5 years of 91·0% (95% CI 89·9–92·0) for patients in the POAI group and 90·4% (88·7–91·9) in the control group. Within the POAI-treated HER2-negative subpopulation, 5-year recurrence risk in women with low Ki67B and Ki672W (low–low) was 4·3% (95% CI 2·9–6·3), 8·4% (6·8–10·5) with high Ki67B and low Ki672W (high–low) and 21·5% (17·1–27·0) with high Ki67B and Ki672W (high–high). Within the POAI-treated HER2-positive subpopulation, 5-year recurrence risk in the low–low group was 10·1% (95% CI 3·2–31·3), 7·7% (3·4–17·5) in the high–low group, and 15·7% (10·1–24·4) in the high–high group. The most commonly reported grade 3 adverse events were hot flushes (20 [1%] of 2801 patients in the POAI group vs six [<1%] of 1400 in the control group) and musculoskeletal pain (29 [1%] vs 13 [1%]). No treatment-related deaths were reported. Interpretation POAI has not been shown to improve treatment outcome, but can be used without detriment to help select appropriate adjuvant therapy based on tumour Ki67. Most patients with low Ki67B or low POAI-induced Ki672W do well with adjuvant standard endocrine therapy (giving consideration to clinical–pathological factors), whereas those whose POAI-induced Ki672W remains high might benefit from further adjuvant treatment or trials of new therapies. Funding Cancer Research UK.
Collapse
Affiliation(s)
- Ian Smith
- The Royal Marsden NHS Foundation Trust, London, UK; The Institute of Cancer Research, London, UK.
| | - John Robertson
- University of Nottingham, Nottingham, UK; University Hospitals of Derby and Burton, Derby, UK
| | | | | | | | - Chris Holcombe
- Liverpool University Hospitals Foundation Trust, Liverpool, UK
| | | | - Cliona Kirwan
- University of Manchester and Manchester University NHS Foundation Trust, Manchester, UK
| | | | | | - Anthony Skene
- Royal Bournemouth and Christchurch NHS Foundation Trust, Bournemouth, UK
| | - Raghavan Vidya
- University of Birmingham and Royal Wolverhampton NHS Trust, Wolverhampton, UK
| | | | | | | | - Kally Sidhu
- The Royal Marsden NHS Foundation Trust, London, UK
| | | | | | - Mitch Dowsett
- The Royal Marsden NHS Foundation Trust, London, UK; The Institute of Cancer Research, London, UK
| |
Collapse
|
17
|
Madigan LI, Dinh P, Graham JD. Neoadjuvant endocrine therapy in locally advanced estrogen or progesterone receptor-positive breast cancer: determining the optimal endocrine agent and treatment duration in postmenopausal women-a literature review and proposed guidelines. Breast Cancer Res 2020; 22:77. [PMID: 32690069 PMCID: PMC7370425 DOI: 10.1186/s13058-020-01314-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/06/2020] [Indexed: 12/20/2022] Open
Abstract
Introduction For patients with locally advanced estrogen receptor or progesterone receptor-positive breast cancer, neoadjuvant endocrine therapy (NET) facilitates down-staging of the tumor and increased rates of breast-conserving surgery. However, NET remains under-utilized, and there are very limited clinical guidelines governing which therapeutic agent to use, or the optimal duration of treatment in postmenopausal women. This literature review aims to discuss the evidence surrounding (1) biomarkers for patient selection for NET, (2) the optimal neoadjuvant endocrine agent for postmenopausal women with locally advanced breast cancer, and (3) the optimal duration of NET. In addition, we make initial recommendations towards developing a clinical guideline for the prescribing of NET. Method A wide-ranging search of online electronic databases was conducted using a truncated PIC search strategy to identify articles that were relevant to these aims and revealed a number of key findings. Results Randomized trials have consistently demonstrated that aromatase inhibitors are more effective than tamoxifen, in terms of objective response rate and rate of BCS, and should be used as first-line NET. The three available aromatase inhibitors have so far been demonstrated to be biologically equivalent, with the choice of aromatase inhibitor not having been shown to affect clinical outcomes. There is increasing evidence for extending the duration of NET beyond 3 to 4 months, to at least 6 months or until maximal clinical response is achieved. While on-treatment levels of the proliferation marker Ki67 are predictive of long-term outcome, the choice of adjuvant therapy in patients who have received NET and then surgery is best guided by the preoperative endocrine prognostic index, or PEPI, which incorporates Ki67 with other clinical parameters. Conclusion This study reveals that in appropriately selected patients, NET can provide equivalent clinical benefit to neoadjuvant chemotherapy in the same cohort, if suitable treatments and durations are chosen. Our findings highlight the need for better defined biomarkers both for guiding patient selection and for measuring outcomes. Development of standard guidelines for the prescribing of NET has the potential to improve both clinical outcomes and quality of life in this patient cohort.
Collapse
Affiliation(s)
- Lauren I Madigan
- Sydney Medical School - Westmead, The University of Sydney, Sydney, Australia.,Present Address: South Eastern Sydney Local Health District, and St. George and Sutherland Clinical Schools, UNSW Medicine, Sydney, Australia
| | - Phuong Dinh
- Sydney Medical School - Westmead, The University of Sydney, Sydney, Australia.,Westmead Breast Cancer Institute, Westmead Hospital, Westmead, Australia
| | - J Dinny Graham
- Sydney Medical School - Westmead, The University of Sydney, Sydney, Australia. .,Westmead Breast Cancer Institute, Westmead Hospital, Westmead, Australia. .,The Westmead Institute for Medical Research, The University of Sydney, Westmead, Australia.
| |
Collapse
|
18
|
Chopra N, Tovey H, Pearson A, Cutts R, Toms C, Proszek P, Hubank M, Dowsett M, Dodson A, Daley F, Kriplani D, Gevensleben H, Davies HR, Degasperi A, Roylance R, Chan S, Tutt A, Skene A, Evans A, Bliss JM, Nik-Zainal S, Turner NC. Homologous recombination DNA repair deficiency and PARP inhibition activity in primary triple negative breast cancer. Nat Commun 2020; 11:2662. [PMID: 32471999 PMCID: PMC7260192 DOI: 10.1038/s41467-020-16142-7] [Citation(s) in RCA: 184] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 04/03/2020] [Indexed: 12/17/2022] Open
Abstract
Triple negative breast cancer (TNBC) encompasses molecularly different subgroups, with a subgroup harboring evidence of defective homologous recombination (HR) DNA repair. Here, within a phase 2 window clinical trial, RIO trial (EudraCT 2014-003319-12), we investigate the activity of PARP inhibitors in 43 patients with untreated TNBC. The primary end point, decreased Ki67, occured in 12% of TNBC. In secondary end point analyses, HR deficiency was identified in 69% of TNBC with the mutational-signature-based HRDetect assay. Cancers with HRDetect mutational signatures of HR deficiency had a functional defect in HR, assessed by impaired RAD51 foci formation on end of treatment biopsy. Following rucaparib treatment there was no association of Ki67 change with HR deficiency. In contrast, early circulating tumor DNA dynamics identified activity of rucaparib, with end of treatment ctDNA levels suppressed by rucaparib in mutation-signature HR-deficient cancers. In ad hoc analysis, rucaparib induced expression of interferon response genes in HR-deficient cancers. The majority of TNBCs have a defect in DNA repair, identifiable by mutational signature analysis, that may be targetable with PARP inhibitors.
Collapse
Affiliation(s)
- Neha Chopra
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, CB2 0XZ, United Kingdom
| | - Holly Tovey
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Alex Pearson
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, CB2 0XZ, United Kingdom
| | - Ros Cutts
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, CB2 0XZ, United Kingdom
| | - Christy Toms
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Paula Proszek
- The Centre for Molecular Pathology, The Royal Marsden Hospital, 15 Cotswold Road, Sutton, SM2 5NG, Surrey, United Kingdom
| | - Michael Hubank
- The Centre for Molecular Pathology, The Royal Marsden Hospital, 15 Cotswold Road, Sutton, SM2 5NG, Surrey, United Kingdom
| | - Mitch Dowsett
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, CB2 0XZ, United Kingdom
- Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital, London, United Kingdom
| | - Andrew Dodson
- Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital, London, United Kingdom
| | - Frances Daley
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, CB2 0XZ, United Kingdom
| | - Divya Kriplani
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, CB2 0XZ, United Kingdom
| | - Heidi Gevensleben
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, CB2 0XZ, United Kingdom
| | - Helen Ruth Davies
- Department of Medical Genetics, The Clinical School, Box 238, Level 6 Addenbrooke's Treatment Centre, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, United Kingdom
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Box 197, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, United Kingdom
| | - Andrea Degasperi
- Department of Medical Genetics, The Clinical School, Box 238, Level 6 Addenbrooke's Treatment Centre, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, United Kingdom
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Box 197, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, United Kingdom
| | - Rebecca Roylance
- University College London Hospitals NHS Foundation Trust, NIHR University College London Hospitals Biomedical Research Centre, London, United Kingdom
| | - Stephen Chan
- Nottingham University Hospital Trust (City Campus), Nottingham, United Kingdom
| | - Andrew Tutt
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, CB2 0XZ, United Kingdom
- Breast Cancer Now Research Unit, Cancer Centre, Guy's Hospital, King's College London, London, United Kingdom
| | - Anthony Skene
- Royal Bournemouth Hospital, Bournemouth, United Kingdom
| | - Abigail Evans
- Poole Hospital NHS Foundation Trust, Poole, United Kingdom
| | - Judith M Bliss
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, United Kingdom
| | - Serena Nik-Zainal
- Department of Medical Genetics, The Clinical School, Box 238, Level 6 Addenbrooke's Treatment Centre, Cambridge Biomedical Campus, Cambridge, CB2 0QQ, United Kingdom
- MRC Cancer Unit, Hutchison/MRC Research Centre, University of Cambridge, Box 197, Cambridge Biomedical Campus, Cambridge, CB2 0XZ, United Kingdom
| | - Nicholas C Turner
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, CB2 0XZ, United Kingdom.
- Breast Unit, The Royal Marsden Hospital, Fulham Road, London, United Kingdom.
| |
Collapse
|
19
|
Expression of Nrf2 and NF-κB transcription factors in breast cancer and breast fibroadenoma: Insights for a new therapeutic approach. Oncotarget 2020; 11:1629-1636. [PMID: 32405338 PMCID: PMC7210014 DOI: 10.18632/oncotarget.27574] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 04/03/2020] [Indexed: 11/30/2022] Open
Abstract
Background: Cancer and fibroadenoma are the most common breast tumors in women of reproductive age. Nuclear factor erythroid 2-related factor 2 (Nrf2) and the nuclear factor kappaB (NF-κB) transcription factor play an important role in the inflammatory process and in cell proliferation. However, few studies have analyzed these markers in breast cancer and fibroadenoma in women of reproductive age.
Results: Light microscopy showed a higher concentration of anti-Nrf2 and anti-NF-κB-stained nuclei in breast cancer than in fibroadenoma. The mean percentage of stained nuclei for Nrf2 was 7.12 ± 5.2 and 43.21 ± 19.83 in the control and study groups, respectively (p < 0.0001). The mean percentage of anti-NF-κB was 10.75 ± 7.09 and 56.14 ± 21.19 (mean ± standard deviation) in the control and study groups, respectively (p < 0.0001). Histological grade 3 tumors showed a significantly higher expression of Nrf2 and NF-κB than grade 1 tumors (p < 0.05).
Material and methods: This study was approved by the Institutional Review Board of Federal University of Piaui and all patients assigned an inform consent term prior to the study initiation. Nrf2 and NF-κB expression was evaluated by immunohistochemistry in 66 patients, divided into two groups, control (fibroadenoma, n = 36) and study (cancer, n = 30). The data were analyzed using ANOVA test and the statistical significance was established at p < 0.05.
Conclusion: Nrf2 and NF-κB expression was significantly higher in breast cancer than in fibroadenoma, in addition to having a greater association with more aggressive tumors.
Collapse
|
20
|
McDonald ES, Doot RK, Young AJ, Schubert EK, Tchou J, Pryma DA, Farwell MD, Nayak A, Ziober A, Feldman MD, DeMichele A, Clark AS, Shah PD, Lee H, Carlin SD, Mach RH, Mankoff DA. Breast Cancer 18F-ISO-1 Uptake as a Marker of Proliferation Status. J Nucl Med 2019; 61:665-670. [PMID: 31836680 DOI: 10.2967/jnumed.119.232363] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 09/16/2019] [Indexed: 12/13/2022] Open
Abstract
The σ2 receptor is a potential in vivo target for measuring proliferative status in cancer. The feasibility of using N-(4-(6,7-dimethoxy-3,4-dihydroisoquinolin-2(1H)-yl)butyl)-2-(2-18F-fluoroethoxy)-5-methylbenzamide (18F-ISO-1) to image solid tumors in lymphoma, breast cancer, and head and neck cancer has been previously established. Here, we report the results of the first dedicated clinical trial of 18F-ISO-1 in women with primary breast cancer. Our study objective was to determine whether 18F-ISO-1 PET could provide an in vivo measure of tumor proliferative status, and we hypothesized that uptake would correlate with a tissue-based assay of proliferation, namely Ki-67 expression. Methods: Twenty-eight women with 29 primary invasive breast cancers were prospectively enrolled in a clinical trial (NCT02284919) between March 2015 and January 2017. Each received an injection of 278-527 MBq of 18F-ISO-1 and then underwent PET/CT imaging of the breasts 50-55 min later. In vivo uptake of 18F-ISO-1 was quantitated by SUVmax and distribution volume ratios and was compared with ex vivo immunohistochemistry for Ki-67. Wilcoxon rank-sum tests assessed uptake differences across Ki-67 thresholds, and Spearman correlation tested associations between uptake and Ki-67. Results: Tumor SUVmax (median, 2.0 g/mL; range, 1.3-3.3 g/mL), partial-volume-corrected SUVmax, and SUV ratios were tested against Ki-67. Tumors stratified into the high-Ki-67 (≥20%) group had SUVmax greater than the low-Ki-67 (<20%) group (P = 0.02). SUVmax exhibited a positive correlation with Ki-67 across all breast cancer subtypes (ρ = 0.46, P = 0.01, n = 29). Partial-volume-corrected SUVmax was positively correlated with Ki-67 for invasive ductal carcinoma (ρ = 0.51, P = 0.02, n = 21). Tumor-to-normal-tissue ratios and tumor distribution volume ratio did not correlate with Ki-67 (P > 0.05). Conclusion: 18F-ISO-1 uptake in breast cancer modestly correlates with an in vitro assay of proliferation.
Collapse
Affiliation(s)
- Elizabeth S McDonald
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert K Doot
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Anthony J Young
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Erin K Schubert
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Julia Tchou
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Daniel A Pryma
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael D Farwell
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Anupma Nayak
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Amy Ziober
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Michael D Feldman
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Angela DeMichele
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Amy S Clark
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Payal D Shah
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Hsiaoju Lee
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sean D Carlin
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert H Mach
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David A Mankoff
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
21
|
Martins LM, de Melo Escorcio Dourado CS, Campos-Verdes LM, Sampaio FA, Revoredo CMS, Costa-Silva DR, da Conceição Barros-Oliveira M, de Jesus Nery Junior E, do Rego-Medeiros LM, Gebrim LH, Alves-Ribeiro FA, Rodrigues GP, Chagas DC, do Nascimento Marreiro D, da Silva BB. Expression of matrix metalloproteinase 2 and 9 in breast cancer and breast fibroadenoma: a randomized, double-blind study. Oncotarget 2019; 10:6879-6884. [PMID: 31839881 PMCID: PMC6901341 DOI: 10.18632/oncotarget.27347] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 11/06/2019] [Indexed: 11/25/2022] Open
Abstract
Background Matrix metalloproteinases (MMPs) 2 and 9 may play an important role in cell proliferation and dissemination of cancer. However, few studies have compared the expression of these proteins between breast cancer and fibroadenoma. Material and methods A randomized, double-blind study was carried out in 66 premenopausal women, aged 20-49 years, who had been diagnosed with fibroadenoma or breast cancer. The patients were divided into two groups: Group A, control (fibroadenoma, n=36) and Group B, study (cancer, n=30). Immunohistochemical analysis was performed using tissue samples of fibroadenoma and breast cancer to assess MMP-2 and MMP-9 antigen expression. Cells were considered positive if exhibiting brown cytoplasmic staining. Fisher’s exact test was used to compare the percentage of cases with cells expressing MMP-2 and MMP-9 in control and study groups (p < 0.05). Results Light microscopy showed a higher concentration of cells with positive cytoplasmic staining for MMP-2 and MMP-9 expression in breast cancer than in fibroadenoma. The percentage of cases with cells expressing MMP-2 in the control and study groups was 41.67% and 86.11%, respectively (p < 0.0009), whereas the percentage of cases with cells expressing MMP-9 in groups A and B was 66.67% and 93.33%, respectively (p<0.0138). MMP-2 and MMP-9 positive expression was significantly higher in moderately differentiated tumors compared to well and poorly differentiated tumors, p <0.005 and p<0.001, respectively. Conclusions The current study shows that MMP-2 and MMP-9 protein expression was significantly higher in the breast cancer than in the fibroadenoma and also in moderately differentiated breast cancer.
Collapse
Affiliation(s)
- Luana Mota Martins
- Postgraduate Program, Northeast Biotechnology Network (RENORBIO), Biotechnology in Health, Federal University of Piaui, Teresina, Piaui 64000-020, Brazil
| | - Carla Solange de Melo Escorcio Dourado
- Postgraduate Program, Northeast Biotechnology Network (RENORBIO), Biotechnology in Health, Federal University of Piaui, Teresina, Piaui 64000-020, Brazil
| | - Larysse Maira Campos-Verdes
- Postgraduate Program, Northeast Biotechnology Network (RENORBIO), Biotechnology in Health, Federal University of Piaui, Teresina, Piaui 64000-020, Brazil
| | - Fabiane Araújo Sampaio
- Postgraduate Program, Northeast Biotechnology Network (RENORBIO), Biotechnology in Health, Federal University of Piaui, Teresina, Piaui 64000-020, Brazil
| | - Camila Maria Simplício Revoredo
- Postgraduate Program, Northeast Biotechnology Network (RENORBIO), Biotechnology in Health, Federal University of Piaui, Teresina, Piaui 64000-020, Brazil
| | - Danylo Rafhael Costa-Silva
- Postgraduate Program, Northeast Biotechnology Network (RENORBIO), Biotechnology in Health, Federal University of Piaui, Teresina, Piaui 64000-020, Brazil
| | - Maria da Conceição Barros-Oliveira
- Postgraduate Program, Northeast Biotechnology Network (RENORBIO), Biotechnology in Health, Federal University of Piaui, Teresina, Piaui 64000-020, Brazil
| | - Elmo de Jesus Nery Junior
- Postgraduate Program, Northeast Biotechnology Network (RENORBIO), Biotechnology in Health, Federal University of Piaui, Teresina, Piaui 64000-020, Brazil
| | - Lucia Maria do Rego-Medeiros
- Facid / Wyden Differential Integral Medicine Faculty, Department of Mastology, Teresina, Piaui 64052-810, Brazill
| | - Luiz Henrique Gebrim
- Postgraduate Program, Northeast Biotechnology Network (RENORBIO), Biotechnology in Health, Federal University of Piaui, Teresina, Piaui 64000-020, Brazil
| | - Francisco Adelton Alves-Ribeiro
- Postgraduate Program, Northeast Biotechnology Network (RENORBIO), Biotechnology in Health, Federal University of Piaui, Teresina, Piaui 64000-020, Brazil
| | - Gilmara Péres Rodrigues
- Postgraduate Program, Northeast Biotechnology Network (RENORBIO), Biotechnology in Health, Federal University of Piaui, Teresina, Piaui 64000-020, Brazil
| | - Diego Cipriano Chagas
- Postgraduate Program, Northeast Biotechnology Network (RENORBIO), Biotechnology in Health, Federal University of Piaui, Teresina, Piaui 64000-020, Brazil
| | - Dilina do Nascimento Marreiro
- Postgraduate Program, Northeast Biotechnology Network (RENORBIO), Biotechnology in Health, Federal University of Piaui, Teresina, Piaui 64000-020, Brazil
| | - Benedito Borges da Silva
- Postgraduate Program, Northeast Biotechnology Network (RENORBIO), Biotechnology in Health, Federal University of Piaui, Teresina, Piaui 64000-020, Brazil.,Facid / Wyden Differential Integral Medicine Faculty, Department of Mastology, Teresina, Piaui 64052-810, Brazill
| |
Collapse
|
22
|
A case-control study of Metallothionein-1 expression in breast cancer and breast fibroadenoma. Sci Rep 2019; 9:7407. [PMID: 31092851 PMCID: PMC6520370 DOI: 10.1038/s41598-019-43565-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 04/27/2019] [Indexed: 11/08/2022] Open
Abstract
The overexpression of Metallothionein-1 (MT-1) may play an important role in breast cancer; however, few studies have compared MT-1 expression between breast cancer and fibroadenoma. A cross-sectional controlled study was performed in 66 premenopausal women, aged 20–49 years, who had been histologically diagnosed with breast fibroadenoma or breast cancer. The patients were divided into two groups: group A, control (fibroadenoma, n = 36) and group B, study (breast cancer, n = 30). Immunohistochemistry was performed on tissue samples of fibroadenoma and breast cancer patients to evaluate the expression of metallothionein using an anti-MT-1 polyclonal antibody (rabbit polyclonal anti-metallothionein-Catalog Number biorbyt-orb11042) at a dilution of 1:100. The data were analyzed using NOVA (p < 0.05). Microscopic analysis showed a higher concentration of anti-MT-1-stained nuclei in breast cancer tissues than in fibroadenoma tissues. The mean proportion of cells with anti-MT-1-stained nuclei was 26.93% and 9.10%, respectively, in the study and control groups (p < 0.001). Histological grade 3 tumors showed a significantly higher MT-1 expression than hitological grade 1 (p < 0.05), while breast tumors negative for estrogen-, progesterone- and HER2- receptors had a significantly higher MT-1 expression than positive breast tumors positive for these parameters (p < 0.05). MT-1 protein in women of reproductive age was significantly higher in breast cancer than in fibroadenoma in this study. Furthermore, there was higher MT-1 immunoreactivity in more aggressive tumors.
Collapse
|
23
|
Johnston S, Puhalla S, Wheatley D, Ring A, Barry P, Holcombe C, Boileau JF, Provencher L, Robidoux A, Rimawi M, McIntosh SA, Shalaby I, Stein RC, Thirlwell M, Dolling D, Morden J, Snowdon C, Perry S, Cornman C, Batten LM, Jeffs LK, Dodson A, Martins V, Modi A, Osborne CK, Pogue-Geile KL, Cheang MCU, Wolmark N, Julian TB, Fisher K, MacKenzie M, Wilcox M, Huang Bartlett C, Koehler M, Dowsett M, Bliss JM, Jacobs SA. Randomized Phase II Study Evaluating Palbociclib in Addition to Letrozole as Neoadjuvant Therapy in Estrogen Receptor-Positive Early Breast Cancer: PALLET Trial. J Clin Oncol 2019; 37:178-189. [PMID: 30523750 DOI: 10.1200/jco.18.01624] [Citation(s) in RCA: 135] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
PURPOSE CDK4/6 inhibitors are used to treat estrogen receptor (ER)-positive metastatic breast cancer (BC) in combination with endocrine therapy. PALLET is a phase II randomized trial that evaluated the effects of combination palbociclib plus letrozole as neoadjuvant therapy. PATIENTS AND METHODS Postmenopausal women with ER-positive primary BC and tumors greater than or equal to 2.0 cm were randomly assigned 3:2:2:2 to letrozole (2.5 mg/d) for 14 weeks (A); letrozole for 2 weeks, then palbociclib plus letrozole to 14 weeks (B); palbociclib for 2 weeks, then palbociclib plus letrozole to 14 weeks (C); or palbociclib plus letrozole for 14 weeks. Palbociclib 125 mg/d was administered orally on a 21-days-on, 7-days-off schedule. Core-cut biopsies were taken at baseline and 2 and 14 weeks. Coprimary end points for letrozole versus palbociclib plus letrozole groups (A v B + C + D) were change in Ki-67 (protein encoded by the MKI67 gene; immunohistochemistry) between baseline and 14 weeks and clinical response (ordinal and ultrasound) after 14 weeks. Complete cell-cycle arrest was defined as Ki-67 less than or equal to 2.7%. Apoptosis was characterized by cleaved poly (ADP-ribose) polymerase. RESULTS Three hundred seven patients were recruited. Clinical response was not significantly different between palbociclib plus letrozole and letrozole groups ( P = .20; complete response + partial response, 54.3% v 49.5%), and progressive disease was 3.2% versus 5.4%, respectively. Median log-fold change in Ki-67 was greater with palbociclib plus letrozole compared with letrozole (-4.1 v -2.2; P < .001) in the 190 evaluable patients (61.9%), corresponding to a geometric mean change of -97.4% versus -88.5%. More patients on palbociclib plus letrozole achieved complete cell-cycle arrest (90% v 59%; P < .001). Median log-fold change (suppression) of cleaved poly (ADP-ribose) polymerase was greater with palbociclib plus letrozole versus letrozole (-0.80 v -0.42; P < .001). More patients had grade 3 or greater toxicity on palbociclib plus letrozole (49.8% v 17.0%; P < .001) mainly because of asymptomatic neutropenia. CONCLUSION Adding palbociclib to letrozole significantly enhanced the suppression of malignant cell proliferation (Ki-67) in primary ER-positive BC, but did not increase the clinical response rate over 14 weeks, which was possibly related to a concurrent reduction in apoptosis.
Collapse
Affiliation(s)
- Stephen Johnston
- 1 The Royal Marsden National Health Service Foundation Trust, London, United Kingdom
| | - Shannon Puhalla
- 2 Univeristy of Pittsburgh Medical Center Cancer Center, Pittsburgh, PA
| | - Duncan Wheatley
- 3 Royal Cornwall Hospitals National Health Service Foundation Trust, Treliske, United Kingdom
| | - Alistair Ring
- 1 The Royal Marsden National Health Service Foundation Trust, London, United Kingdom
| | - Peter Barry
- 1 The Royal Marsden National Health Service Foundation Trust, London, United Kingdom
| | - Chris Holcombe
- 4 Royal Liverpool and Broadgreen University Hospitals National Health Service Trust, Liverpool, United Kingdom
| | | | - Louise Provencher
- 6 Centre Hospitalier Université de Quebec-Universite Laval, Quebec City, Quebec, Canada
| | - André Robidoux
- 7 Centre Hospitalier Université de Montréal, Montreal, Quebec, Canada
| | | | | | - Ibrahim Shalaby
- 10 Joe Arrington Cancer Research and Treatment Center, Lubbock, TX
| | - Robert C Stein
- 11 National Institute for Health Research University College London Hospitals Biomedical Research Centre, London, United Kingdom
- 12 University College London Hospitals National Health Service Foundation Trust, London, United Kingdom
| | | | - David Dolling
- 14 The Institute of Cancer Research, London, United Kingdom
| | - James Morden
- 14 The Institute of Cancer Research, London, United Kingdom
| | - Claire Snowdon
- 14 The Institute of Cancer Research, London, United Kingdom
| | - Sophie Perry
- 14 The Institute of Cancer Research, London, United Kingdom
| | - Chester Cornman
- 15 National Surgical Adjuvant Breast and Bowel Project Foundation, Pittsburgh, PA
| | - Leona M Batten
- 14 The Institute of Cancer Research, London, United Kingdom
| | - Lisa K Jeffs
- 14 The Institute of Cancer Research, London, United Kingdom
| | - Andrew Dodson
- 1 The Royal Marsden National Health Service Foundation Trust, London, United Kingdom
- 14 The Institute of Cancer Research, London, United Kingdom
| | - Vera Martins
- 1 The Royal Marsden National Health Service Foundation Trust, London, United Kingdom
| | - Arjun Modi
- 1 The Royal Marsden National Health Service Foundation Trust, London, United Kingdom
| | | | | | | | - Norman Wolmark
- 15 National Surgical Adjuvant Breast and Bowel Project Foundation, Pittsburgh, PA
| | - Thomas B Julian
- 16 Allegheny Health Network Cancer Institute, Pittsburgh, PA
| | - Kate Fisher
- 17 International Drug Development Institute, Brussels, Belgium
| | | | - Maggie Wilcox
- 18 Independent Cancer Patients Voice, London, United Kingdom
| | | | | | - Mitch Dowsett
- 1 The Royal Marsden National Health Service Foundation Trust, London, United Kingdom
- 14 The Institute of Cancer Research, London, United Kingdom
| | - Judith M Bliss
- 14 The Institute of Cancer Research, London, United Kingdom
| | - Samuel A Jacobs
- 15 National Surgical Adjuvant Breast and Bowel Project Foundation, Pittsburgh, PA
| |
Collapse
|
24
|
Denduluri N, Miller K, O'Regan RM. Using a Neoadjuvant Approach for Evaluating Novel Therapies for Patients With Breast Cancer. Am Soc Clin Oncol Educ Book 2018; 38:47-55. [PMID: 30231324 DOI: 10.1200/edbk_200719] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Preoperative systemic therapy, though primarily used to downstage breast cancers, can offer, using pathologic complete response (pCR) as an endpoint, a rapid assessment of efficacy of a given therapeutic approach, particularly in triple-negative (TNBC) and HER2-positive breast cancers. Recently, this approach resulted in the approval of pertuzumab for HER2-positive cancers, in a considerably quicker timeline than would have been possible with its assessment in the adjuvant setting. However, the use of preoperative systemic therapy remains controversial, as the higher response rates noted with newer approaches have not routinely translated into improved longer-term outcomes, nor have they been confirmed in larger adjuvant trials. Almost all trials have demonstrated that pCR is a robust prognostic marker in patients with TNBC and HER2-positive cancers, so part of this discrepancy may be due to inadequate power in the preoperative trials and/or due to the heterogeneous nature of breast cancers. PCR following preoperative chemotherapy is not prognostic in many hormone receptor (HR)-positive breast cancers, especially those with a luminal A phenotype, which typically has minimal response to chemotherapy. Given this lack of response to chemotherapy, there is considerable interest in the use of neoadjuvant endocrine therapy (NET). The rate of pCR to NET in HR-positive cancers is low, leading to the use of surrogate markers, including changes in Ki-67 and the preoperative endocrine prognostic index (PEPI) score, as biomarkers of efficacy. Overall, the use of neoadjuvant approaches offers a rapid assessment of efficacy of novel therapies and remains a useful research tool for drug evaluation.
Collapse
Affiliation(s)
- Neelima Denduluri
- From the Virginia Cancer Specialists, Fairfax, VA; Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; University of Wisconsin Carbone Cancer Center, Madison, WI
| | - Kathy Miller
- From the Virginia Cancer Specialists, Fairfax, VA; Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; University of Wisconsin Carbone Cancer Center, Madison, WI
| | - Ruth M O'Regan
- From the Virginia Cancer Specialists, Fairfax, VA; Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN; University of Wisconsin Carbone Cancer Center, Madison, WI
| |
Collapse
|
25
|
AlFakeeh A, Brezden-Masley C. Overcoming endocrine resistance in hormone receptor-positive breast cancer. ACTA ACUST UNITED AC 2018; 25:S18-S27. [PMID: 29910644 DOI: 10.3747/co.25.3752] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Endocrine therapy, a major modality in the treatment of hormone receptor (hr)-positive breast cancer (bca), has improved outcomes in metastatic and nonmetastatic disease. However, a limiting factor to the use of endocrine therapy in bca is resistance resulting from the development of escape pathways that promote the survival of cancer cells despite estrogen receptor (er)-targeted therapy. The resistance pathways involve extensive cross-talk between er and receptor tyrosine kinase growth factors [epidermal growth factor receptor, human epidermal growth factor receptor 2 (her2), and insulin-like growth factor 1 receptor] and their downstream signalling pathways-most notably pi3k/akt/mtor and mapk. In some cases, resistance develops as a result of genetic or epigenetic alterations in various components of the signalling pathways, such as overexpression of her2 and erα co-activators, aberrant expression of cell-cycle regulators, and PIK3CA mutations. By combining endocrine therapy with various molecularly targeted agents and signal transduction inhibitors, some success has been achieved in overcoming and modulating endocrine resistance in hr-positive bca. Established strategies include selective er downregulators, anti-her2 agents, mtor (mechanistic target of rapamycin) inhibitors, and inhibitors of cyclin-dependent kinases 4 and 6. Inhibitors of pi3ka are not currently a treatment option for women with hr-positive bca outside the context of clinical trial. Ongoing clinical trials are exploring more agents that could be combined with endocrine therapy, and biomarkers that would help to guide decision-making and maximize clinical efficacy. In this review article, we address current treatment strategies for endocrine resistance, and we highlight future therapeutic targets in the endocrine pathway of bca.
Collapse
Affiliation(s)
- A AlFakeeh
- Division of Hematology/Oncology, St. Michael's Hospital, University of Toronto, Toronto, ON.,King Fahad Medical City, Comprehensive Cancer Centre, Riyadh, Saudi Arabia
| | - C Brezden-Masley
- Division of Hematology/Oncology, St. Michael's Hospital, University of Toronto, Toronto, ON.,Keenan Research Centre, Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Toronto, ON
| |
Collapse
|
26
|
Ma CX, Suman V, Goetz MP, Northfelt D, Burkard ME, Ademuyiwa F, Naughton M, Margenthaler J, Aft R, Gray R, Tevaarwerk A, Wilke L, Haddad T, Moynihan T, Loprinzi C, Hieken T, Barnell EK, Skidmore ZL, Feng YY, Krysiak K, Hoog J, Guo Z, Nehring L, Wisinski KB, Mardis E, Hagemann IS, Vij K, Sanati S, Al-Kateb H, Griffith OL, Griffith M, Doyle L, Erlichman C, Ellis MJ. A Phase II Trial of Neoadjuvant MK-2206, an AKT Inhibitor, with Anastrozole in Clinical Stage II or III PIK3CA-Mutant ER-Positive and HER2-Negative Breast Cancer. Clin Cancer Res 2017; 23:6823-6832. [PMID: 28874413 PMCID: PMC6392430 DOI: 10.1158/1078-0432.ccr-17-1260] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 07/25/2017] [Accepted: 08/30/2017] [Indexed: 02/01/2023]
Abstract
Purpose: Hyperactivation of AKT is common and associated with endocrine resistance in estrogen receptor-positive (ER+) breast cancer. The allosteric pan-AKT inhibitor MK-2206 induced apoptosis in PIK3CA-mutant ER+ breast cancer under estrogen-deprived condition in preclinical studies. This neoadjuvant phase II trial was therefore conducted to test the hypothesis that adding MK-2206 to anastrozole induces pathologic complete response (pCR) in PIK3CA mutant ER+ breast cancer.Experimental Design: Potential eligible patients with clinical stage II/III ER+/HER2- breast cancer were preregistered and received anastrozole (goserelin if premenopausal) for 28 days in cycle 0 pending tumor PIK3CA sequencing. Patients positive for PIK3CA mutation in the tumor were eligible to start MK-2206 (150 mg orally weekly, with prophylactic prednisone) on cycle 1 day 2 (C1D2) and to receive a maximum of four 28-day cycles of combination therapy before surgery. Serial biopsies were collected at preregistration, C1D1 and C1D17.Results: Fifty-one patients preregistered and 16 of 22 with PIK3CA-mutant tumors received study drug. Three patients went off study due to C1D17 Ki67 >10% (n = 2) and toxicity (n = 1). Thirteen patients completed neoadjuvant therapy followed by surgery. No pCRs were observed. Rash was common. MK-2206 did not further suppress cell proliferation and did not induce apoptosis on C1D17 biopsies. Although AKT phosphorylation was reduced, PRAS40 phosphorylation at C1D17 after MK-2206 persisted. One patient acquired an ESR1 mutation at surgery.Conclusions: MK-2206 is unlikely to add to the efficacy of anastrozole alone in PIK3CA-mutant ER+ breast cancer and should not be studied further in the target patient population. Clin Cancer Res; 23(22); 6823-32. ©2017 AACR.
Collapse
Affiliation(s)
- Cynthia X Ma
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri.
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Vera Suman
- Department of Health Sciences Research, Mayo Clinic, Rochester, Minnesota
| | | | - Donald Northfelt
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, Arizona
| | - Mark E Burkard
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Foluso Ademuyiwa
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Michael Naughton
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Julie Margenthaler
- Section of Endocrine and Oncologic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Rebecca Aft
- Section of Endocrine and Oncologic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Richard Gray
- Department of General Surgery, Mayo Clinic, Phoenix, Arizona
| | - Amye Tevaarwerk
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Lee Wilke
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Tufia Haddad
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | | | | | - Tina Hieken
- Department of General Surgery, Mayo Clinic, Rochester, Minnesota
| | - Erica K Barnell
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri
| | - Zachary L Skidmore
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri
| | - Yan-Yang Feng
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri
| | - Kilannin Krysiak
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri
| | - Jeremy Hoog
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Zhanfang Guo
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Leslie Nehring
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Kari B Wisinski
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Elaine Mardis
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri
| | - Ian S Hagemann
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Kiran Vij
- Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Souzan Sanati
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Hussam Al-Kateb
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Obi L Griffith
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri
| | - Malachi Griffith
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri
| | - Laurence Doyle
- Cancer Therapy Evaluation Program, NCI, Bethesda, Maryland
| | | | - Matthew J Ellis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
27
|
Abstract
Purpose of Review Pre-operative endocrine therapy can be used to down-stage large or locally advanced breast cancers in ER+ disease. In the last four decades, it has evolved from a treatment perceived as an alternative to surgery for those too unfit to undergo surgery or chemotherapy, to the present day where it is a valuable and valid option in the treatment of postmenopausal women with ER-rich (Allred score 7–8, or > 50% staining for ER) breast cancer. Recent Findings Emerging data from the metastatic setting is translating into neoadjuvant trials, utilising dual endocrine targeting or combinations of endocrine agents and other targeted drugs, including those acting against components of the PI3K pathway and the cell cycle. The routine use of peri-operative endocrine therapy in all ER+ tumours may help to yield important long-term prognostic information, and guide adjuvant endocrine therapy. Summary Pre-operative endocrine therapy is an exciting and evolving area with emerging new approaches. In this review, established evidence and emerging data on its applications are discussed.
Collapse
|
28
|
Abstract
The goal of individualized and targeted treatment and precision medicine requires the assessment of potential therapeutic targets to direct treatment selection. The biomarkers used to direct precision medicine, often termed companion diagnostics, for highly targeted drugs have thus far been almost entirely based on in vitro assay of biopsy material. Molecular imaging companion diagnostics offer a number of features complementary to those from in vitro assay, including the ability to measure the heterogeneity of each patient's cancer across the entire disease burden and to measure early changes in response to treatment. We discuss the use of molecular imaging methods as companion diagnostics for cancer therapy with the goal of predicting response to targeted therapy and measuring early (pharmacodynamic) response as an indication of whether the treatment has "hit" the target. We also discuss considerations for probe development for molecular imaging companion diagnostics, including both small-molecule probes and larger molecules such as labeled antibodies and related constructs. We then describe two examples where both predictive and pharmacodynamic molecular imaging markers have been tested in humans: endocrine therapy for breast cancer and human epidermal growth factor receptor type 2-targeted therapy. The review closes with a summary of the items needed to move molecular imaging companion diagnostics from early studies into multicenter trials and into the clinic.
Collapse
Affiliation(s)
- David A Mankoff
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA.
| | - Christine E Edmonds
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Michael D Farwell
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Daniel A Pryma
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
29
|
Clinical and microarray analysis of breast cancers of all subtypes from two prospective preoperative chemotherapy studies. Br J Cancer 2016; 115:411-9. [PMID: 27415010 PMCID: PMC4985347 DOI: 10.1038/bjc.2016.184] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 05/08/2016] [Accepted: 05/19/2016] [Indexed: 02/08/2023] Open
Abstract
Background: We aimed to analyse clinical and gene expression profiles to predict pathologic complete response and disease-free survival using two consecutive, prospective, preoperative chemotherapy trial cohorts. Methods: Clinicopathological and gene expression data were evaluated in a cohort from two consecutive phase II preoperative studies that included patients with stage IIA–IIIC breast cancer of all subtypes. Analysed specimens were obtained before preoperative chemotherapy, and cDNA microarray analyses were performed using the Affymetrix Gene Chip U133 plus 2.0. Results: Between December 2005 and December 2010, 122 patients were analysed. The pathologic complete response rate was significantly higher in HER2+ and HR−/HER2− cancers. Age, pathologic complete response, HR−/HER2− status, and lymph node positivity (⩾4) were significant poor prognostic factors for disease-free survival. For the cDNA microarray analyses, sufficient tumour samples were available from 78 of the 107 patients (73%). An 8-gene signature predictive of pathologic complete response and a 17-gene signature predictive of prognosis were identified. Patients were categorised into low-risk (n=45) and high-risk groups (n=33) (HR 70.0, P=0.004). Conclusions: This study yielded preliminary data on the expression of specific genes predicting pathologic complete response and disease-free survival in a cohort of chemonaïve breast cancer patients. Further validation may distinguish those who would benefit most from perioperative chemotherapy as well as those needing further intervention.
Collapse
|
30
|
Christgen M, Steinemann D, Kühnle E, Länger F, Gluz O, Harbeck N, Kreipe H. Lobular breast cancer: Clinical, molecular and morphological characteristics. Pathol Res Pract 2016; 212:583-97. [DOI: 10.1016/j.prp.2016.05.002] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 04/11/2016] [Accepted: 05/04/2016] [Indexed: 01/20/2023]
|
31
|
Sivalingam VN, Kitson S, McVey R, Roberts C, Pemberton P, Gilmour K, Ali S, Renehan AG, Kitchener HC, Crosbie EJ. Measuring the biological effect of presurgical metformin treatment in endometrial cancer. Br J Cancer 2016; 114:281-9. [PMID: 26794276 PMCID: PMC4742583 DOI: 10.1038/bjc.2015.453] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Revised: 11/26/2015] [Accepted: 12/02/2015] [Indexed: 12/25/2022] Open
Abstract
Background: Preclinical studies in endometrial cancer (EC) show that metformin reduces cellular proliferation by PI3K-AKT-mTOR inhibition. We tested the hypothesis that short-term presurgical metformin reduces cellular proliferation in atypical endometrial hyperplasia (AEH) and endometrioid EC, and assessed the feasibility of using phosphorylated PI3K-AKT-mTOR proteins as tissue end points. Methods: Women with AEH or EC received metformin 850 mg twice a day or no drug in the presurgical window between diagnosis and hysterectomy. Before and after the window, tissue samples were obtained; serum markers of insulin resistance (e.g. homeostasis model of assessment of insulin resistance index) were determined; and anthropometrics measured (e.g. BMI). Cell proliferation (Ki-67) and PI3K-AKT-mTOR phosphostatus were assessed by immunohistochemistry and scored blinded to treatment. Results: Twenty-eight metformin-treated and 12 untreated patients, well matched for age and BMI, completed the study. Metformin treatment (median 20 days, range 7–34) was associated with a 17.2% reduction in tumour Ki-67 (95% CI −27.4, −7.0, P=0.002), in a dose-dependent manner. Tumour PI3K-AKT-mTOR protein phosphostatus varied but the effects were not significant after adjusting for changes in controls. Conclusions: Short-term metformin was associated with reduced Ki-67 expression in EC. Changes in tumour PI3K-AKT-mTOR protein phosphostatus were seen in both groups. Future studies should address the variability attributed to different sampling techniques including devascularisation of the uterus at hysterectomy.
Collapse
Affiliation(s)
- V N Sivalingam
- Gynaecological Oncology Research Group, Institute of Cancer Sciences, University of Manchester, St Mary's Hospital, Oxford Road, Manchester M13 9WL, UK
| | - S Kitson
- Gynaecological Oncology Research Group, Institute of Cancer Sciences, University of Manchester, St Mary's Hospital, Oxford Road, Manchester M13 9WL, UK
| | - R McVey
- Department of Histopathology, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - C Roberts
- Centre for Biostatistics, Institute of Population Health, University of Manchester, Manchester, UK
| | - P Pemberton
- Clinical Biochemistry Department, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - K Gilmour
- Obstetrics and Gynaecology Department, Tameside General Hospital, Tameside, UK
| | - S Ali
- Obstetrics and Gynaecology Department, The Royal Oldham Hospital, Pennine Acute Hospitals NHS Trust, Oldham, UK
| | - A G Renehan
- Cancer Studies and Surgery Research Group, Institute of Cancer Sciences, University of Manchester, Manchester, UK
| | - H C Kitchener
- Gynaecological Oncology Research Group, Institute of Cancer Sciences, University of Manchester, St Mary's Hospital, Oxford Road, Manchester M13 9WL, UK
| | - E J Crosbie
- Gynaecological Oncology Research Group, Institute of Cancer Sciences, University of Manchester, St Mary's Hospital, Oxford Road, Manchester M13 9WL, UK
| |
Collapse
|
32
|
Ma CX, Sanchez C, Gao F, Crowder R, Naughton M, Pluard T, Creekmore A, Guo Z, Hoog J, Lockhart AC, Doyle A, Erlichman C, Ellis MJ. A Phase I Study of the AKT Inhibitor MK-2206 in Combination with Hormonal Therapy in Postmenopausal Women with Estrogen Receptor-Positive Metastatic Breast Cancer. Clin Cancer Res 2016; 22:2650-8. [PMID: 26783290 DOI: 10.1158/1078-0432.ccr-15-2160] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 01/02/2016] [Indexed: 01/28/2023]
Abstract
PURPOSE PI3K/AKT pathway activation is an important endocrine resistance mechanism in estrogen receptor-positive (ER(+)) breast cancer. After promising preclinical modeling of MK-2206, an allosteric pan-AKT inhibitor, with either estrogen deprivation or fulvestrant, we conducted a phase I trial in patients with metastatic ER(+)HER2(-) breast cancer to determine the recommended phase II treatment dose (RPTD) of MK-2206 when combined with either anastrozole, fulvestrant, or anastrozole/fulvestrant. EXPERIMENTAL DESIGN ER(+) breast cancer cell lines were exposed in vitro to MK-2206 plus estrogen deprivation with or without fulvestrant and monitored for apoptosis. A standard 3+3 design was employed to first determine the maximum tolerated dose (MTD) of MK-2206 plus anastrozole based on cycle 1 toxicity. Each cycle was 28 days. The RPTD was determined on the basis of toxicities observed at MTD level during the first 3 cycles. Subsequent patients received MK-2206, at the RPTD determined above, plus fulvestrant or anastrozole/fulvestrant to define RPTD for these additional regimens. RESULTS MK-2206 induced apoptosis in parental ER(+) but not in long-term estrogen-deprived cell lines, for which fulvestrant was required for apoptosis induction. Thirty-one patients enrolled. The RPTD was defined as MK-2206 150 mg orally weekly with prednisone prophylaxis for each combination. Grade 3 rash was dose limiting. 42% (95% CI, 23%-63%) patients derived clinical benefit without progression within 6 months. Response was not associated with tumor PIK3CA mutation. CONCLUSIONS MK-2206 plus endocrine treatments were tolerable. MK-2206 in combination with anastrozole is being further evaluated in a phase II neoadjuvant trial for newly diagnosed ER(+)HER2(-) breast cancer. Clin Cancer Res; 22(11); 2650-8. ©2016 AACRSee related commentary by Jansen et al., p. 2599.
Collapse
Affiliation(s)
- Cynthia X Ma
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri.
| | - Cesar Sanchez
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Feng Gao
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Robert Crowder
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Michael Naughton
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Timothy Pluard
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Allison Creekmore
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Zhanfang Guo
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Jeremy Hoog
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - A Craig Lockhart
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Austin Doyle
- Cancer Therapy Evaluation Program, NCI, Bethesda, MD
| | | | - Matthew J Ellis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
33
|
Barroso-Sousa R, Silva DDAFR, Alessi JVM, Mano MS. Neoadjuvant endocrine therapy in breast cancer: current role and future perspectives. Ecancermedicalscience 2016; 10:609. [PMID: 26823678 PMCID: PMC4720494 DOI: 10.3332/ecancer.2016.609] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Indexed: 12/18/2022] Open
Abstract
Luminal breast cancer, as defined by oestrogen and/or progesterone expression by immunohistochemistry, accounts for up to 75% of all breast cancers. In this population, endocrine therapy is likely to account for most of the gains obtained with the administration of adjuvant systemic treatment. The role of adjuvant chemotherapy in these patients remains debatable since it is known that only a small fraction of patients will derive meaningful benefit from this treatment whilst the majority will be exposed to significant and unnecessary chemotherapy-related toxicities, in particular the elderly and frail. Therefore, neoadjuvant endocrine therapy (NET) becomes an attractive option for selected patients with hormonal-receptor positive locally advanced breast cancer. In this review, we discuss the current role of NET and future perspectives in the field.
Collapse
Affiliation(s)
- Romualdo Barroso-Sousa
- Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina, Universidade de São Paulo, Av Dr Arnaldo, 251, 5o andar, 01246-000 São Paulo, SP, Brazil
| | - Danilo D A Fonseca Reis Silva
- Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina, Universidade de São Paulo, Av Dr Arnaldo, 251, 5o andar, 01246-000 São Paulo, SP, Brazil
| | - Joao Victor Machado Alessi
- Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina, Universidade de São Paulo, Av Dr Arnaldo, 251, 5o andar, 01246-000 São Paulo, SP, Brazil
| | - Max Senna Mano
- Instituto do Câncer do Estado de São Paulo (ICESP), Faculdade de Medicina, Universidade de São Paulo, Av Dr Arnaldo, 251, 5o andar, 01246-000 São Paulo, SP, Brazil
| |
Collapse
|
34
|
Yamashita H. Tumor biology in estrogen receptor-positive, human epidermal growth factor receptor type 2-negative breast cancer: Mind the menopausal status. World J Clin Oncol 2015; 6:220-224. [PMID: 26677435 PMCID: PMC4675907 DOI: 10.5306/wjco.v6.i6.220] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 08/25/2015] [Accepted: 10/27/2015] [Indexed: 02/06/2023] Open
Abstract
Breast cancer is not one disease, but can be categorized into four major molecular subtypes according to hormone receptor [estrogen receptor (ER) and progesterone receptor (PgR)] and human epidermal growth factor receptor type 2 (HER2) expression status. Ki67 labeling index and/or multigene assays are used to classify ER-positive, HER2-negative breast cancer into luminal A and luminal B (HER2-negative) subtypes. To date, most studies analyzing predictive or prognostic factors in ER-positive breast cancer have been performed in postmenopausal women, mainly using patients and samples in adjuvant aromatase inhibitor trials. In contrast, even the clinical roles of PgR and Ki67 have been little analyzed so far in premenopausal women. PgR is one of the estrogen-responsive genes, and it has been reported that plasma estradiol levels are related to expression levels of estrogen-responsive genes including PGR in ER-positive breast cancer. In this article, biological differences, especially differences in expression of PgR and Ki67 in ER-positive breast cancer between pre- and postmenopausal women are discussed. Clinical roles of PgR and Ki67 in ER-positive breast cancer differ between pre- and postmenopausal women. We suggest that the mechanisms of development and estrogen-dependent growth of ER-positive breast cancer might differ according to menopausal status.
Collapse
|
35
|
Quenel-Tueux N, Debled M, Rudewicz J, MacGrogan G, Pulido M, Mauriac L, Dalenc F, Bachelot T, Lortal B, Breton-Callu C, Madranges N, de Lara CT, Fournier M, Bonnefoi H, Soueidan H, Nikolski M, Gros A, Daly C, Wood H, Rabbitts P, Iggo R. Clinical and genomic analysis of a randomised phase II study evaluating anastrozole and fulvestrant in postmenopausal patients treated for large operable or locally advanced hormone-receptor-positive breast cancer. Br J Cancer 2015; 113:585-94. [PMID: 26171933 PMCID: PMC4647692 DOI: 10.1038/bjc.2015.247] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 06/05/2015] [Accepted: 06/15/2015] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The aim of this study was to assess the efficacy of neoadjuvant anastrozole and fulvestrant treatment of large operable or locally advanced hormone-receptor-positive breast cancer not eligible for initial breast-conserving surgery, and to identify genomic changes occurring after treatment. METHODS One hundred and twenty post-menopausal patients were randomised to receive 1 mg anastrozole (61 patients) or 500 mg fulvestrant (59 patients) for 6 months. Genomic DNA copy number profiles were generated for a subgroup of 20 patients before and after treatment. RESULTS A total of 108 patients were evaluable for efficacy and 118 for toxicity. The objective response rate determined by clinical palpation was 58.9% (95% CI=45.0-71.9) in the anastrozole arm and 53.8% (95% CI=39.5-67.8) in the fulvestrant arm. The breast-conserving surgery rate was 58.9% (95% CI=45.0-71.9) in the anastrozole arm and 50.0% (95% CI=35.8-64.2) in the fulvestrant arm. Pathological responses >50% occurred in 24 patients (42.9%) in the anastrozole arm and 13 (25.0%) in the fulvestrant arm. The Ki-67 score fell after treatment but there was no significant difference between the reduction in the two arms (anastrozole 16.7% (95% CI=13.3-21.0) before, 3.2% (95% CI=1.9-5.5) after, n=43; fulvestrant 17.1% (95%CI=13.1-22.5) before, 3.2% (95% CI=1.8-5.7) after, n=38) or between the reduction in Ki-67 in clinical responders and non-responders. Genomic analysis appeared to show a reduction of clonal diversity following treatment with selection of some clones with simpler copy number profiles. CONCLUSIONS Both anastrozole and fulvestrant were effective and well-tolerated, enabling breast-conserving surgery in over 50% of patients. Clonal changes consistent with clonal selection by the treatment were seen in a subgroup of patients.
Collapse
Affiliation(s)
- Nathalie Quenel-Tueux
- Institut Bergonié Comprehensive Cancer Centre, 229 Cours de l'Argonne, F-33000 Bordeaux, France
| | - Marc Debled
- Institut Bergonié Comprehensive Cancer Centre, 229 Cours de l'Argonne, F-33000 Bordeaux, France
| | - Justine Rudewicz
- Institut Bergonié Comprehensive Cancer Centre, 229 Cours de l'Argonne, F-33000 Bordeaux, France
- INSERM U916, 229 Cours de l'Argonne, F-33000 Bordeaux, France
- University Bordeaux, 16 Avenue Léon Duguit, F-33608 Pessac, France
- Bordeaux Bioinformatics Centre, University Bordeaux, 146, rue Léo Saignat, F-33076 Bordeaux, France
- CNRS UMR5800, Bordeaux Computer Science Lab, 351 Cours de la Libération, F-33405 Talence, France
| | - Gaetan MacGrogan
- Institut Bergonié Comprehensive Cancer Centre, 229 Cours de l'Argonne, F-33000 Bordeaux, France
- INSERM U916, 229 Cours de l'Argonne, F-33000 Bordeaux, France
- University Bordeaux, 16 Avenue Léon Duguit, F-33608 Pessac, France
| | - Marina Pulido
- Inserm Clinical Investigation Centre CIC1401, Epidemiological Unit, 229 Cours de l'Argonne, Bordeaux 33076, France
- Clinical and Epidemiological Research Unit, Institut Bergonie, 229 Cours de l'Argonne, Bordeaux 33076, France
| | - Louis Mauriac
- Institut Bergonié Comprehensive Cancer Centre, 229 Cours de l'Argonne, F-33000 Bordeaux, France
| | - Florence Dalenc
- Institut Claudius Regaud, IUCT-Oncopole Toulouse, 1 Avenue Irène Joliot-Curie, F-31059 Toulouse, France
| | - Thomas Bachelot
- CLCC Lyon, 28 Promenade Léa et Napoléon Bullukian, F-69008 Lyon, France
| | - Barbara Lortal
- Institut Bergonié Comprehensive Cancer Centre, 229 Cours de l'Argonne, F-33000 Bordeaux, France
| | - Christelle Breton-Callu
- Institut Bergonié Comprehensive Cancer Centre, 229 Cours de l'Argonne, F-33000 Bordeaux, France
| | - Nicolas Madranges
- Institut Bergonié Comprehensive Cancer Centre, 229 Cours de l'Argonne, F-33000 Bordeaux, France
| | - Christine Tunon de Lara
- Institut Bergonié Comprehensive Cancer Centre, 229 Cours de l'Argonne, F-33000 Bordeaux, France
| | - Marion Fournier
- Institut Bergonié Comprehensive Cancer Centre, 229 Cours de l'Argonne, F-33000 Bordeaux, France
| | - Hervé Bonnefoi
- Institut Bergonié Comprehensive Cancer Centre, 229 Cours de l'Argonne, F-33000 Bordeaux, France
- INSERM U916, 229 Cours de l'Argonne, F-33000 Bordeaux, France
- University Bordeaux, 16 Avenue Léon Duguit, F-33608 Pessac, France
| | - Hayssam Soueidan
- Bordeaux Bioinformatics Centre, University Bordeaux, 146, rue Léo Saignat, F-33076 Bordeaux, France
| | - Macha Nikolski
- University Bordeaux, 16 Avenue Léon Duguit, F-33608 Pessac, France
- Bordeaux Bioinformatics Centre, University Bordeaux, 146, rue Léo Saignat, F-33076 Bordeaux, France
- CNRS UMR5800, Bordeaux Computer Science Lab, 351 Cours de la Libération, F-33405 Talence, France
| | - Audrey Gros
- Institut Bergonié Comprehensive Cancer Centre, 229 Cours de l'Argonne, F-33000 Bordeaux, France
- INSERM U916, 229 Cours de l'Argonne, F-33000 Bordeaux, France
- University Bordeaux, 16 Avenue Léon Duguit, F-33608 Pessac, France
| | - Catherine Daly
- Leeds Institute of Cancer and Pathology, University of Leeds, Beckett Street, Leeds LS9 7TF, UK
| | - Henry Wood
- Leeds Institute of Cancer and Pathology, University of Leeds, Beckett Street, Leeds LS9 7TF, UK
| | - Pamela Rabbitts
- Leeds Institute of Cancer and Pathology, University of Leeds, Beckett Street, Leeds LS9 7TF, UK
| | - Richard Iggo
- Institut Bergonié Comprehensive Cancer Centre, 229 Cours de l'Argonne, F-33000 Bordeaux, France
- INSERM U916, 229 Cours de l'Argonne, F-33000 Bordeaux, France
- University Bordeaux, 16 Avenue Léon Duguit, F-33608 Pessac, France
| |
Collapse
|
36
|
Christgen M, von Ahsen S, Christgen H, Länger F, Kreipe H. The region-of-interest size impacts on Ki67 quantification by computer-assisted image analysis in breast cancer. Hum Pathol 2015. [PMID: 26206765 DOI: 10.1016/j.humpath.2015.05.016] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Therapeutic decision-making in breast cancer depends on histopathologic biomarkers and is influenced by the Ki67 proliferation index. Computer-assisted image analysis (CAIA) promises to improve Ki67 quantification. Several commercial applications have been developed for semiautomated CAIA-based Ki67 quantification, many of which rely on measurements in user-defined regions of interest (ROIs). Because of intratumoral proliferative heterogeneity, definition of the ROI is an important step in the analytical procedure. This study explores the ROI size impacts on Ki67 quantification. Whole-slide sections of 100 breast cancers were immunostained with the anti-Ki67 antibody 30-9 and were analyzed on the iScan Coreo digital pathology platform using a Food and Drug Administration-cleared Ki67 quantification software version v5.3 (Virtuoso; Ventana, Tucson, TX). For each case, the Ki67 labeling index (LI) was determined in multiple ROIs of gradually increasing size centered around a high-proliferation area. The spatial Ki67 decline was modeled with nonlinear regression. Depending on the ROI size, the median Ki67 LI varied between 55% and 15%. The proportion of tumors classified as Ki67 low according to the St Gallen 2013/2015 cutoff increased from 2% to 56%, as the ROI size increased from 50 to 10,000 cells captured. The interrater reliability of conventional Ki67 assessment versus CAIA-based Ki67 quantification was also dependent on the ROI size and varied between slight and almost perfect agreement (Cohen κ = 0.06-0.85). In conclusion, the ROI size is a critically important parameter for semiautomated Ki67 quantification by CAIA. Ki67 LIs determined on platforms like iScan Coreo/Virtuoso require an ROI size adjustment, for which we offer a downloadable data transformation tool.
Collapse
Affiliation(s)
- Matthias Christgen
- Institute of Pathology, Hannover Medical School, 30625 Hannover, Germany.
| | - Sabrina von Ahsen
- Institute of Pathology, Hannover Medical School, 30625 Hannover, Germany
| | | | - Florian Länger
- Institute of Pathology, Hannover Medical School, 30625 Hannover, Germany
| | - Hans Kreipe
- Institute of Pathology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
37
|
Rusz O, Vörös A, Varga Z, Kelemen G, Uhercsák G, Nikolényi A, Ormándi K, Simonka Z, Kahán Z. One-Year Neoadjuvant Endocrine Therapy in Breast Cancer. Pathol Oncol Res 2015; 21:977-84. [PMID: 25753983 DOI: 10.1007/s12253-015-9911-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 02/16/2015] [Indexed: 12/14/2022]
Abstract
The evaluation of the effects of 1-year endocrine therapy (NET) was aimed at. A retrospective analysis of 42 cases with 46 stage II-III invasive, hormone receptor-positive, HER2-negative breast cancers was performed. One-year NET was planned with letrozole (n = 33, postmenopausal group), or with goserelin plus letrozole (n = 7) or with goserelin plus tamoxifen (n = 2) (premenopausal group). Surgery was performed in accordance with the initial stage and the response to therapy. With regard to the tumor remaining in the surgical specimen, risk groups were constructed: Group 1: stage 0, pathological complete regression (pCR); Group 2: stages IA-IIA; Group 3: stages ≥ IIB + cases with clinical progression. Due to local progression, NET was replaced by neoadjuvant chemotherapy in three patients (four tumors). In two postmenopausal patients, letrozole was replaced by tamoxifen because of the insufficient treatment effect. In 19/42 cases, breast-conserving surgery was performed. Within Group 1, there was no cancer in four cases, while only DCIS remained in 2 (pCR: 13 %); Groups 2 and 3 comprised 25 and 15 cases, respectively. The likeliness of a good response (Groups 1 and 2 vs. Group 3) to NET was increased by 7 % for every 1 % increase of the expression of ER (OR = 1.070; 95 % CI: 1.007-1.138, p = 0.029). Progression-free survival differed according to treatment response (p = 0.001). The post-therapy Ki67 value of ≤ 15 % had only a marginal effect on survival. No other associations were detected between the tumor characteristics and the therapeutic response or survival. Long-duration NET is effective and safe in cases of hormone-sensitive breast cancer.
Collapse
Affiliation(s)
- Orsolya Rusz
- Department of Oncotherapy, University of Szeged, Korányi fasor 12, 6720, Szeged, Hungary
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Elumalai P, Arunakaran J. Review on molecular and chemopreventive potential of nimbolide in cancer. Genomics Inform 2014; 12:156-64. [PMID: 25705153 PMCID: PMC4330249 DOI: 10.5808/gi.2014.12.4.156] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2014] [Revised: 11/09/2014] [Accepted: 11/09/2014] [Indexed: 12/16/2022] Open
Abstract
Cancer is the most dreaded disease in human and also major health problem worldwide. Despite its high occurrence, the exact molecular mechanisms of the development and progression are not fully understood. The existing cancer therapy based on allopathic medicine is expensive, exhibits side effects; and may also alter the normal functioning of genes. Thus, a non-toxic and effective mode of treatment is needed to control cancer development and progression. Some medicinal plants offer a safe, effective and affordable remedy to control the cancer progression. Nimbolide, a limnoid derived from the neem (Azadirachta indica) leaves and flowers of neem, is widely used in traditional medical practices for treating various human diseases. Nimbolide exhibits several pharmacological effects among which its anticancer activity is the most promising. The previous studies carried out over the decades have shown that nimbolide inhibits cell proliferation and metastasis of cancer cells. This review highlights the current knowledge on the molecular targets that contribute to the observed anticancer activity of nimbolide related to induction of apoptosis and cell cycle arrest; and inhibition of signaling pathways related to cancer progression.
Collapse
Affiliation(s)
- Perumal Elumalai
- Department of Endocrinology, Dr. ALM Post Graduate Institute of Basic Medical Sciences, University of Madras, Chennai 600113, India
| | - Jagadeesan Arunakaran
- Department of Endocrinology, Dr. ALM Post Graduate Institute of Basic Medical Sciences, University of Madras, Chennai 600113, India
| |
Collapse
|
39
|
Kilickap S, Kaya Y, Yucel B, Tuncer E, Babacan NA, Elagoz S. Higher Ki67 expression is associates with unfavorable prognostic factors and shorter survival in breast cancer. Asian Pac J Cancer Prev 2014; 15:1381-5. [PMID: 24606469 DOI: 10.7314/apjcp.2014.15.3.1381] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The prognostic value of the Ki67 expression level is yet unclear in breast cancer. The aim of this study was to investigate the association between Ki67 expression levels and prognostic factors such as grade, Her2 and hormone receptor expression status in breast cancers. MATERIALS AND METHODS Clinical and pathological features of the patients with breast cancer were retreived from the hospital records. RESULTS In this study, 163 patients with breast cancer were analyzed, with a mean age of 53.4±12.2 years. Median Ki67 positivity was 20% and Ki67-high tumors were significantly associated with high grade (p<0.001), lymphovascular invasion (p=0.001), estrogen receptor (ER) negativity (p=0.035), Her2 positivity (p=0.001), advanced stage (p<0.001) and lymph node positivity (p<0.003) . Lower Ki67 levels were significantly associated with longer median relapse-free and overall survival compared to those of higher Ki67 levels. CONCLUSIONS High Ki67 expression is associated with ER negativity, Her2 positivity, higher grade and axillary lymph node involvement in breast cancers. The level of Ki67 expression is a prognostic factor predicting relapse-free and overall survival in breast cancer patients.
Collapse
Affiliation(s)
- Saadettin Kilickap
- Department of Medical Oncology, Hacettepe University Institute of Cancer, Turkey E-mail :
| | | | | | | | | | | |
Collapse
|
40
|
Alistar A, Chou JW, Nagalla S, Black MA, D'Agostino R, Miller LD. Dual roles for immune metagenes in breast cancer prognosis and therapy prediction. Genome Med 2014; 6:80. [PMID: 25419236 PMCID: PMC4240891 DOI: 10.1186/s13073-014-0080-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 10/02/2014] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Neoadjuvant chemotherapy for breast cancer leads to considerable variability in clinical responses, with only 10 to 20% of cases achieving complete pathologic responses (pCR). Biological and clinical factors that determine the extent of pCR are incompletely understood. Mounting evidence indicates that the patient's immune system contributes to tumor regression and can be modulated by therapies. The cell types most frequently observed with this association are effector tumor infiltrating lymphocytes (TILs), such as cytotoxic T cells, natural killer cells and B cells. We and others have shown that the relative abundance of TILs in breast cancer can be quantified by intratumoral transcript levels of coordinately expressed, immune cell-specific genes. Through expression microarray analysis, we recently discovered three immune gene signatures, or metagenes, that appear to reflect the relative abundance of distinct tumor-infiltrating leukocyte populations. The B/P (B cell/plasma cell), T/NK (T cell/natural killer cell) and M/D (monocyte/dendritic cell) immune metagenes were significantly associated with distant metastasis-free survival of patients with highly proliferative cancer of the basal-like, HER2-enriched and luminal B intrinsic subtypes. METHODS Given the histopathological evidence that TIL abundance is predictive of neoadjuvant treatment efficacy, we evaluated the therapy-predictive potential of the prognostic immune metagenes. We hypothesized that pre-chemotherapy immune gene signatures would be significantly predictive of tumor response. In a multi-institutional, meta-cohort analysis of 701 breast cancer patients receiving neoadjuvant chemotherapy, gene expression profiles of tumor biopsies were investigated by logistic regression to determine the existence of therapy-predictive interactions between the immune metagenes, tumor proliferative capacity, and intrinsic subtypes. RESULTS By univariate analysis, the B/P, T/NK and M/D metagenes were all significantly and positively associated with favorable pathologic responses. In multivariate analyses, proliferative capacity and intrinsic subtype altered the significance of the immune metagenes in different ways, with the M/D and B/P metagenes achieving the greatest overall significance after adjustment for other variables. CONCLUSIONS Gene expression signatures of infiltrating immune cells carry both prognostic and therapy-predictive value that is impacted by tumor proliferative capacity and intrinsic subtype. Anti-tumor functions of plasma B cells and myeloid-derived antigen-presenting cells may explain more variability in pathologic response to neoadjuvant chemotherapy than previously recognized.
Collapse
Affiliation(s)
- Angela Alistar
- Department of Internal Medicine, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC 27157 USA ; The Comprehensive Cancer Center of Wake Forest University, Medical Center Blvd, Winston Salem, NC 27157 USA
| | - Jeff W Chou
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC 27157 USA
| | - Srikanth Nagalla
- Department of Medicine, Jefferson Medical College, 1015 Chestnut Street, Suite 1321, Philadelphia, PA 19107 USA
| | - Michael A Black
- Department of Biochemistry, Otago School of Medical Sciences, University of Otago, 710 Cumberland Street, Dunedin, 9054 New Zealand
| | - Ralph D'Agostino
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC 27157 USA ; The Comprehensive Cancer Center of Wake Forest University, Medical Center Blvd, Winston Salem, NC 27157 USA
| | - Lance D Miller
- Department of Cancer Biology, Wake Forest School of Medicine, Medical Center Blvd, Winston-Salem, NC 27157 USA ; The Comprehensive Cancer Center of Wake Forest University, Medical Center Blvd, Winston Salem, NC 27157 USA
| |
Collapse
|
41
|
Abstract
Around 70% of all breast cancers are estrogen receptor alpha positive and hence their development is highly dependent on estradiol. While the invention of endocrine therapies has revolusioned the treatment of the disease, resistance to therapy eventually occurs in a large number of patients. This paper seeks to illustrate and discuss the complexity and heterogeneity of the mechanisms which underlie resistance and the approaches proposed to combat them. It will also focus on the use and development of methods for predicting which patients are likely to develop resistance.
Collapse
|
42
|
Ki67 measured in metastatic tissue and prognosis in patients with advanced breast cancer. Breast Cancer Res Treat 2014; 147:407-14. [DOI: 10.1007/s10549-014-3096-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 08/06/2014] [Indexed: 11/28/2022]
|
43
|
The apoptotic pathways effect of fine particulate from cooking oil fumes in primary fetal alveolar type II epithelial cells. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2014; 761:35-43. [DOI: 10.1016/j.mrgentox.2014.01.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 12/19/2013] [Accepted: 01/07/2014] [Indexed: 12/18/2022]
|
44
|
Dixon JM. Prospects of neoadjuvant aromatase inhibitor therapy in breast cancer. Expert Rev Anticancer Ther 2014; 8:453-63. [DOI: 10.1586/14737140.8.3.453] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
45
|
Sagar S, Esau L, Holtermann K, Hikmawan T, Zhang G, Stingl U, Bajic VB, Kaur M. Induction of apoptosis in cancer cell lines by the Red Sea brine pool bacterial extracts. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 13:344. [PMID: 24305113 PMCID: PMC4235048 DOI: 10.1186/1472-6882-13-344] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 11/28/2013] [Indexed: 01/17/2023]
Abstract
BACKGROUND Marine microorganisms are considered to be an important source of bioactive molecules against various diseases and have great potential to increase the number of lead molecules in clinical trials. Progress in novel microbial culturing techniques as well as greater accessibility to unique oceanic habitats has placed the marine environment as a new frontier in the field of natural product drug discovery. METHODS A total of 24 microbial extracts from deep-sea brine pools in the Red Sea have been evaluated for their anticancer potential against three human cancer cell lines. Downstream analysis of these six most potent extracts was done using various biological assays, such as Caspase-3/7 activity, mitochondrial membrane potential (MMP), PARP-1 cleavage and expression of γH2Ax, Caspase-8 and -9 using western blotting. RESULTS In general, most of the microbial extracts were found to be cytotoxic against one or more cancer cell lines with cell line specific activities. Out of the 13 most active microbial extracts, six extracts were able to induce significantly higher apoptosis (>70%) in cancer cells. Mechanism level studies revealed that extracts from Chromohalobacter salexigens (P3-86A and P3-86B(2)) followed the sequence of events of apoptotic pathway involving MMP disruption, caspase-3/7 activity, caspase-8 cleavage, PARP-1 cleavage and Phosphatidylserine (PS) exposure, whereas another Chromohalobacter salexigens extract (K30) induced caspase-9 mediated apoptosis. The extracts from Halomonas meridiana (P3-37B), Chromohalobacter israelensis (K18) and Idiomarina loihiensis (P3-37C) were unable to induce any change in MMP in HeLa cancer cells, and thus suggested mitochondria-independent apoptosis induction. However, further detection of a PARP-1 cleavage product, and the observed changes in caspase-8 and -9 suggested the involvement of caspase-mediated apoptotic pathways. CONCLUSION Altogether, the study offers novel findings regarding the anticancer potential of several halophilic bacterial species inhabiting the Red Sea (at the depth of 1500-2500 m), which constitute valuable candidates for further isolation and characterization of bioactive molecules.
Collapse
|
46
|
Haroon S, Hashmi AA, Khurshid A, Kanpurwala MA, Mujtuba S, Malik B, Faridi N. Ki67 Index in Breast Cancer: Correlation with Other Prognostic Markers and Potential in Pakistani Patients. Asian Pac J Cancer Prev 2013; 14:4353-8. [DOI: 10.7314/apjcp.2013.14.7.4353] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
47
|
Patani N, Martin LA, Dowsett M. Biomarkers for the clinical management of breast cancer: international perspective. Int J Cancer 2013; 133:1-13. [PMID: 23280579 DOI: 10.1002/ijc.27997] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2012] [Accepted: 12/07/2012] [Indexed: 12/14/2022]
Abstract
The higher incidence of breast cancer in developed countries has been tempered by reductions in mortality, largely attributable to mammographic screening programmes and advances in adjuvant therapy. Optimal systemic management requires consideration of clinical, pathological and biological parameters. Oestrogen receptor alpha (ERα), progesterone receptor (PgR) and human epidermal growth factor receptor 2 (HER2) are established biomarkers evaluated at diagnosis, which identify cardinal subtypes of breast cancer. Their prognostic and predictive utility effectively guides systemic treatment with endocrine, anti-HER2 and chemotherapy. Hence, accurate and reliable determination remains of paramount importance. However, the goals of personalized medicine and targeted therapies demand further information regarding residual risk and potential benefit of additional treatments in specific circumstances. The need for biomarkers which are fit for purpose, and the demands placed upon them, is therefore expected to increase. Technological advances, in particular high-throughput global gene expression profiling, have generated multi-gene signatures providing further prognostic and predictive information. The rational integration of routinely evaluated clinico-pathological parameters with key indicators of biological activity, such as proliferation markers, also provides a ready opportunity to improve the information available to guide systemic therapy decisions. The additional value of such information and its proper place in patient management is currently under evaluation in prospective clinical trials. Expanding the utility of biomarkers to lower resource settings requires an emphasis on cost effectiveness, quality assurance and possible international variations in tumor biology; the potential for improved clinical outcomes should be justified against logistical and economic considerations.
Collapse
Affiliation(s)
- Neill Patani
- The Breakthrough Breast Cancer Research Center, The Institute of Cancer Research, London, United Kingdom
| | | | | |
Collapse
|
48
|
Treatment Response to Preoperative Anthracycline-Based Chemotherapy in Locally Advanced Breast Cancer: The Relevance of Proliferation and Apoptosis Rates. Pathol Oncol Res 2013; 19:577-88. [DOI: 10.1007/s12253-013-9621-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 02/28/2013] [Indexed: 12/11/2022]
|
49
|
PIK3CA genotype and a PIK3CA mutation-related gene signature and response to everolimus and letrozole in estrogen receptor positive breast cancer. PLoS One 2013; 8:e53292. [PMID: 23301057 PMCID: PMC3534682 DOI: 10.1371/journal.pone.0053292] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Accepted: 11/27/2012] [Indexed: 11/19/2022] Open
Abstract
The phosphatidylinositol 3′ kinase (PI3K) pathway is commonly activated in breast cancer and aberrations such as PI3K mutations are common. Recent exciting clinical trial results in advanced estrogen receptor-positive (ER) breast cancer support mTOR activation is a major means of estrogen-independent tumor growth. Hence the means to identify a responsive breast cancer population that would most benefit from these compounds in the adjuvant or earlier stage setting is of high interest. Here we study PIK3CA genotype as well as a previously reported PI3K/mTOR-pathway gene signature (PIK3CA-GS) and their ability to estimate the level of PI3K pathway activation in two clinical trials of newly diagnosed ER-positive breast cancer patients- a total of 81 patients- one of which was randomized between letrozole and placebo vs letrozole and everolimus. The main objectives were to correlate the baseline PIK3CA genotype and GS with the relative change from baseline to day 15 in Ki67 (which has been shown to be prognostic in breast cancer) and phosphorylated S6 (S240) immunohistochemistry (a substrate of mTOR). In the randomized dataset, the PIK3CA-GS could identify those patients with the largest relative decreases in Ki67 to letrozole/everolimus (R = −0.43, p = 0.008) compared with letrozole/placebo (R = 0.07, p = 0.58; interaction test p = 0.02). In a second dataset of pre-surgical everolimus alone, the PIK3CA-GS was not significantly correlated with relative change in Ki67 (R = −0.11, p = 0.37) but with relative change in phosphorlyated S6 (S240) (R = −0.46, p = 0.028). PIK3CA genotype was not significantly associated with any endpoint in either datasets. Our results suggest that the PIK3CA-GS has potential to identify those ER-positive BCs who may benefit from the addition of everolimus to letrozole. Further evaluation of the PIK3CA-GS as a predictive biomarker is warranted as it may facilitate better selection of responsive patient populations for mTOR inhibition in combination with letrozole.
Collapse
|
50
|
Shimizu D, Ishikawa T, Tanabe M, Sasaki T, Ichikawa Y, Chishima T, Endo I. Preoperative endocrine therapy with goserelin acetate and tamoxifen in hormone receptor-positive premenopausal breast cancer patients. Breast Cancer 2012. [PMID: 23184499 DOI: 10.1007/s12282-012-0429-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND The use of preoperative endocrine therapy for breast cancer has increased during the last decade. Although several studies have reported favorable response rates in postmenopausal women, its effectiveness in premenopausal women remains unknown. This study therefore aimed to evaluate the potential benefits of preoperative endocrine therapy in premenopausal women. METHODS Fifty-three patients with estrogen receptor (ER)-positive, human epidermal growth factor receptor 2 (HER2)-negative invasive breast cancer were included in this study. Preoperative endocrine therapy with goserelin acetate and tamoxifen was administered for 3 months. Clinical evaluations were performed by ultrasonography before and after endocrine therapy. Pathological evaluations were performed using core biopsy and surgical specimens. Immunohistochemical evaluations of ER, progesterone receptor (PgR), HER2, and Ki-67 were performed before and after endocrine therapy. RESULTS Partial response (PR) was observed in 23 % (12/53) and progressive disease (PD) in 2 % (2/53) of patients. Significant suppression of Ki-67 was observed following endocrine therapy in 90 % (47/52) of patients (P < 0.0001). Significant downregulation of PgR was observed after endocrine therapy (P = 0.0002), which tended to be correlated with clinical response (P = 0.058). CONCLUSIONS Three months of preoperative endocrine therapy with goserelin acetate and tamoxifen was safe and effective in premenopausal patients with invasive breast cancer, with a 23 % PR rate. Changes in PgR and Ki-67 expression might be promising markers for endocrine responsiveness.
Collapse
Affiliation(s)
- Daisuke Shimizu
- Department of Breast and Thyroid Surgery, Yokohama City University Medical Center, 4-57 Urafunecho, Minami-ku, Yokohama, Kanagawa, 232-0024, Japan,
| | | | | | | | | | | | | |
Collapse
|