1
|
Nicotra R, Lutz C, Messal HA, Jonkers J. Rat Models of Hormone Receptor-Positive Breast Cancer. J Mammary Gland Biol Neoplasia 2024; 29:12. [PMID: 38913216 PMCID: PMC11196369 DOI: 10.1007/s10911-024-09566-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 06/07/2024] [Indexed: 06/25/2024] Open
Abstract
Hormone receptor-positive (HR+) breast cancer (BC) is the most common type of breast cancer among women worldwide, accounting for 70-80% of all invasive cases. Patients with HR+ BC are commonly treated with endocrine therapy, but intrinsic or acquired resistance is a frequent problem, making HR+ BC a focal point of intense research. Despite this, the malignancy still lacks adequate in vitro and in vivo models for the study of its initiation and progression as well as response and resistance to endocrine therapy. No mouse models that fully mimic the human disease are available, however rat mammary tumor models pose a promising alternative to overcome this limitation. Compared to mice, rats are more similar to humans in terms of mammary gland architecture, ductal origin of neoplastic lesions and hormone dependency status. Moreover, rats can develop spontaneous or induced mammary tumors that resemble human HR+ BC. To date, six different types of rat models of HR+ BC have been established. These include the spontaneous, carcinogen-induced, transplantation, hormone-induced, radiation-induced and genetically engineered rat mammary tumor models. Each model has distinct advantages, disadvantages and utility for studying HR+ BC. This review provides a comprehensive overview of all published models to date.
Collapse
Affiliation(s)
- Raquel Nicotra
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, Netherlands
- Oncode Institute, Amsterdam, Netherlands
| | - Catrin Lutz
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, Netherlands.
- Oncode Institute, Amsterdam, Netherlands.
| | - Hendrik A Messal
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, Netherlands.
- Oncode Institute, Amsterdam, Netherlands.
| | - Jos Jonkers
- Division of Molecular Pathology, The Netherlands Cancer Institute, Amsterdam, Netherlands.
- Oncode Institute, Amsterdam, Netherlands.
| |
Collapse
|
2
|
Bartlett JM, Xu K, Wong J, Pond G, Zhang Y, Spears M, Salunga R, Mallon E, Taylor KJ, Hasenburg A, Markopoulos C, Dirix L, van de Velde CJ, Rea D, Schnabel CA, Treuner K, Bayani J. Validation of the Prognostic Performance of Breast Cancer Index in Hormone Receptor-Positive Postmenopausal Breast Cancer Patients in the TEAM Trial. Clin Cancer Res 2024; 30:1509-1517. [PMID: 38345755 PMCID: PMC11016895 DOI: 10.1158/1078-0432.ccr-23-2436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/01/2023] [Accepted: 02/08/2024] [Indexed: 04/16/2024]
Abstract
PURPOSE Patients with early-stage hormone receptor-positive (HR+) breast cancer face a prolonged risk of recurrence even after adjuvant endocrine therapy. The Breast Cancer Index (BCI) is significantly prognostic for overall (0-10 years) and late (5-10 years) distant recurrence (DR) risk in N0 and N1 patients. Here, BCI prognostic performance was evaluated in HR+ postmenopausal women from the Tamoxifen and Exemestane Adjuvant Multinational (TEAM) trial. EXPERIMENTAL DESIGN 3,544 patients were included in the analysis (N = 1,519 N0, N = 2,025 N+). BCI risk groups were calculated using pre-specified cutoff points. Kaplan-Meier analyses and log-rank tests were used to assess the prognostic significance of BCI risk groups based on DR. Hazard ratios (HR) and confidence intervals (CI) were calculated using Cox models with and without clinical covariates. RESULTS For overall 10-year DR, BCI was significantly prognostic in Ni0 (N = 1,196) and N1 (N = 1,234) patients who did not receive prior chemotherapy (P < 0.001). In patients who were DR-free for 5 years, 10-year late DR rates for low- and high-risk groups were 5.4% and 9.3% (N0 cohort, N = 1,285) and 4.8% and 12.2% (N1 cohort, N = 1,625) with multivariate HRs of 2.25 (95% CI, 1.30-3.88; P = 0.004) and 2.67 (95% CI, 1.53-4.63; P < 0.001), respectively. Late DR performance was substantially improved using previously optimized cutoff points, identifying BCI low-risk groups with even lower 10-year late DR rates of 3.8% and 2.7% in N0 and N1 patients, respectively. CONCLUSIONS The TEAM trial represents the largest prognostic validation study for BCI to date and provides a more representative assessment of late DR risk to guide individualized treatment decision-making for HR+ patients with early-stage breast cancer.
Collapse
Affiliation(s)
- John M.S. Bartlett
- Cancer Research UK Scotland Centre, University of Edinburgh, Edinburgh, United Kingdom
| | - Keying Xu
- Diagnostic Development, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Jenna Wong
- Biotheranostics, A Hologic Company, San Diego, California
| | - Gregory Pond
- Department of Biostatistics, McMaster University, Hamilton, Ontario
| | - Yi Zhang
- Biotheranostics, A Hologic Company, San Diego, California
| | - Melanie Spears
- Diagnostic Development, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario
| | | | | | - Karen J. Taylor
- Cancer Research UK Scotland Centre, University of Edinburgh, Edinburgh, United Kingdom
| | - Annette Hasenburg
- Department of Gynecology and Obstetrics, University Center Mainz, Mainz, Germany
| | | | - Luc Dirix
- St. Augustinus Hospital, Antwerp, Belgium
| | | | - Daniel Rea
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, United Kingdom
| | | | - Kai Treuner
- Biotheranostics, A Hologic Company, San Diego, California
| | - Jane Bayani
- Diagnostic Development, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario
| |
Collapse
|
3
|
Lin RK, Su CM, Lin SY, Thi Anh Thu L, Liew PL, Chen JY, Tzeng HE, Liu YR, Chang TH, Lee CY, Hung CS. Hypermethylation of TMEM240 predicts poor hormone therapy response and disease progression in breast cancer. Mol Med 2022; 28:67. [PMID: 35715741 PMCID: PMC9204905 DOI: 10.1186/s10020-022-00474-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/13/2022] [Indexed: 12/16/2022] Open
Abstract
Background Approximately 25% of patients with early-stage breast cancer experience cancer progression throughout the disease course. Alterations in TMEM240 in breast cancer were identified and investigated to monitor treatment response and disease progression. Methods Circulating methylated TMEM240 in the plasma of breast cancer patients was used to monitor treatment response and disease progression. The Cancer Genome Atlas (TCGA) data in Western countries and Illumina methylation arrays in Taiwanese breast cancer patients were used to identify novel hypermethylated CpG sites and genes related to poor hormone therapy response. Quantitative methylation-specific PCR (QMSP), real-time reverse transcription PCR, and immunohistochemical analyses were performed to measure DNA methylation and mRNA and protein expression levels in 394 samples from Taiwanese and Korean breast cancer patients. TMEM240 gene manipulation, viability, migration assays, RNA-seq, and MetaCore were performed to determine its biological functions and relationship to hormone drug treatment response in breast cancer cells. Results Aberrant methylated TMEM240 was identified in breast cancer patients with poor hormone therapy response using genome-wide methylation analysis in the Taiwan and TCGA breast cancer cohorts. A cell model showed that TMEM240, which is localized to the cell membrane and cytoplasm, represses breast cancer cell proliferation and migration and regulates the expression levels of enzymes involved in estrone and estradiol metabolism. TMEM240 protein expression was observed in normal breast tissues but was not detected in 88.2% (67/76) of breast tumors and in 90.0% (9/10) of metastatic tumors from breast cancer patients. QMSP revealed that in 54.5% (55/101) of Taiwanese breast cancer patients, the methylation level of TMEM240 was at least twofold higher in tumor tissues than in matched normal breast tissues. Patients with hypermethylation of TMEM240 had poor 10-year overall survival (p = 0.003) and poor treatment response, especially hormone therapy response (p < 0.001). Circulating methylated TMEM240 dramatically and gradually decreased and then diminished in patients without disease progression, whereas it returned and its levels in plasma rose again in patients with disease progression. Prediction of disease progression based on circulating methylated TMEM240 was found to have 87.5% sensitivity, 93.1% specificity, and 90.2% accuracy. Conclusions Hypermethylation of TMEM240 is a potential biomarker for treatment response and disease progression monitoring in breast cancer. Supplementary Information The online version contains supplementary material available at 10.1186/s10020-022-00474-9.
Collapse
Affiliation(s)
- Ruo-Kai Lin
- Program in Drug Discovery and Development Industry, Program in Clinical Drug Development of Herbal Medicine, Master Program in Clinical Genomics and Proteomics, College of Pharmacy, Graduate Institute of Pharmacognosy, Taipei Medical University, 250 Wu-Hsing Street, Taipei, Taiwan.,Clinical Trial Center, Taipei Medical University Hospital, 252 Wu-Hsing Street, Taipei, Taiwan
| | - Chih-Ming Su
- Division of General Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.,Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shih-Yun Lin
- Program in Drug Discovery and Development Industry, Program in Clinical Drug Development of Herbal Medicine, Master Program in Clinical Genomics and Proteomics, College of Pharmacy, Graduate Institute of Pharmacognosy, Taipei Medical University, 250 Wu-Hsing Street, Taipei, Taiwan
| | - Le Thi Anh Thu
- Quang Tri Medical College, Dien Bien Phu Str., Dong Luong District, Dong Ha City, Quang Tri, Vietnam
| | - Phui-Ly Liew
- Department of Pathology, Shuang Ho Hospital, Taipei Medical University, New Taipei, Taiwan.,Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jian-Yu Chen
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Huey-En Tzeng
- Division of Hematology and Oncology, Department of Medicine, Taipei Medical University Hospital, Taipei, Taiwan.,Department of Medical Research, Division of Hematology/Medical Oncology, Department of Medicine, Taichung Veterans General Hospital, Taichung City, Taiwan.,Program for Cancer Molecular Biology and Drug Discovery, and Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yun-Ru Liu
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei, Taiwan
| | - Tzu-Hao Chang
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Yang Lee
- Bioinformatics Center, Office of Data Science, Taipei Medical University, Taipei, Taiwan
| | - Chin-Sheng Hung
- Division of General Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan. .,Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan. .,Division of General Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei, Taiwan.
| |
Collapse
|
4
|
Wang C, Wei J, Huang L, Xu C. Expression of 34 βE12 may be an independent predictor of survival in breast cancer. J Int Med Res 2021; 49:300060520967774. [PMID: 34666529 PMCID: PMC8532230 DOI: 10.1177/0300060520967774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Objectives To investigate the relationship between high-molecular-weight cytokeratin (34βE12) and clinicopathological parameters (including HER-2, Ki67 and steroid receptors) in breast cancer to determine its usefulness as a prognostic marker. Methods In this retrospective study, the expression level 34βE12 was assessed in surgically resected breast cancer specimens by immunohistochemical staining. Data were correlated with the patients’ clinicopathological parameters. Results Of the 348 breast cancer tissue samples, 232 (67%) showed positive expression of 34βE12. There were statistically significant differences between the positive and negative 34βE12 expression groups in tumour size, lymph node involvement, oestrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor-2 (HER2) status. There were no differences between groups in age, tumour grade, or Ki67 status. In addition, patients who were positive for 34βE12 had significantly extended overall survival. In multivariate analysis, the expression level of 34βE12 was found to be a significant independent prognostic factor. Conclusion These results suggest that positive 34βE12 expression is associated with a favourable outcome in breast cancer and so may be a useful prognostic factor. Further studies are required to confirm these results.
Collapse
Affiliation(s)
- Chuchu Wang
- Department of Breast and Thyroid Surgery, the First Affiliated Hospital of Shaoxing University, Shaoxing University Medical College, Zhejiang, China
| | - Jiangguo Wei
- Department of Pathology, the First Affiliated Hospital of Shaoxing University, Zhejiang, China
| | - Liming Huang
- Department of Breast and Thyroid Surgery, the First Affiliated Hospital of Shaoxing University, Shaoxing University Medical College, Zhejiang, China.,Department of Medical Research Centre, the First Affiliated Hospital of Shaoxing University, Zhejiang, China
| | - Chaoyang Xu
- Department of Breast and Thyroid Surgery, the First Affiliated Hospital of Shaoxing University, Shaoxing University Medical College, Zhejiang, China.,Department of Medical Research Centre, the First Affiliated Hospital of Shaoxing University, Zhejiang, China
| |
Collapse
|
5
|
Association of PET-based estradiol-challenge test for breast cancer progesterone receptors with response to endocrine therapy. Nat Commun 2021; 12:733. [PMID: 33531464 PMCID: PMC7854611 DOI: 10.1038/s41467-020-20814-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 12/22/2020] [Indexed: 01/04/2023] Open
Abstract
Estrogen receptor (ER) testing of breast cancer imperfectly predicts response to endocrine therapy (ET). We hypothesize that a brief estradiol challenge will increase tumor progesterone receptor (PgR) levels only in tumors with functional ER. In this prospective, phase 2, single-center, single-arm trial (NCT02455453), we report the association of response to ET with change in tumor uptake of the progestin analog, 21-[18F]fluorofuranylnorprogesterone (FFNP), before and after a one-day estradiol challenge. In 43 postmenopausal women with advanced ER+ breast cancer, we show a post-challenge increase in tumor FFNP uptake only in 28 subjects with clinical benefit from ET (responders), but not in 15 without clinical benefit (nonresponders) (p < 0.0001), indicating 100% sensitivity and specificity. We further show significantly longer survival (p < 0.0001) in the responding subjects. Our results demonstrate that change in tumor FFNP uptake after estradiol challenge is highly predictive of response to ET in women with ER+ breast cancer. Clinical estrogen receptor (ER) testing for breast cancer is limited in predicting response to endocrine therapy (ET). In this phase 2 clinical trial, authors demonstrate that the responsiveness to ET can be predicted by use of PET/CT with 21-[18F]fluorofuranylnorprogesterone (FFNP) to detect the change in tumor progesterone receptor (PgR) levels after a one-day estradiol challenge.
Collapse
|
6
|
Aggarwal S, Vaid A, Ramesh A, Parikh PM, Purohit S, Avasthi B, Gupta S, Ranjan S, Kaushal V, Salim S, Singh R, Minhas S, Doval D. Practical consensus recommendations on management of HR + ve early breast cancer with specific reference to genomic profiling. South Asian J Cancer 2020; 7:96-101. [PMID: 29721472 PMCID: PMC5909304 DOI: 10.4103/sajc.sajc_110_18] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Breast cancer is a heterogeneous disease and patients are managed clinically based on ER, PR, HER2 expression, and key risk factors. The use of gene expression assays for early stage disease is already common practice. These tests have found a place in risk stratifying the heterogeneous group of stage I–II breast cancers for recurrence, for predicting chemotherapy response, and for predicting breast cancer-related mortality. Most guidelines for hormone receptor (HR)–positive early breast cancer recommend addition of adjuvant chemotherapy for most women, leading to overtreatment, which causes considerable morbidity and cost. Expert oncologist discussed about strategies of gene expression assays and aid in chemotherapy recommendations for treatment of HR + ve EBC and the expert group used data from published literature, practical experience and opinion of a large group of academic oncologists to arrive at this practical consensus recommendations for the benefit of community oncologists.
Collapse
Affiliation(s)
- S Aggarwal
- Department of Medical Oncology, Sir Ganga Ram Hospital, New Delhi, India
| | - A Vaid
- Department of Medical Oncology, Medanata Hospital, Gurugram, Haryana, India
| | - A Ramesh
- Department of Medical Oncology, HCG Cancer Center, Chennai, Tamil Nadu, India.,Department of Medical Oncology, SMH Curie Cancer Center, New Delhi, India
| | - Purvish M Parikh
- Department of Oncology, Shalby Cancer and Research Institute, Mumbai, Maharashtra, India
| | - S Purohit
- Department of Medical Oncology, Artemis Hospital, Gurugram, Haryana, India
| | - B Avasthi
- Department or Radiation Oncology, Fortis Hospital, New Delhi, India
| | - S Gupta
- Department of Medical Oncology, Sarvodaya Hospital, Faridabad, Haryana, India
| | - S Ranjan
- Department of Medicine, INHS Sanjivani, Kochi, Kerala, India
| | - V Kaushal
- Department of Radiation Oncology, RCC, Rohtak, Haryana, India
| | - S Salim
- Department of Oncology, Hakim Sanaullah Cancer Center, Baramulla, Jammu and Kashmir, India
| | - R Singh
- Department of Medical Oncology, Army Hospital R and R, New Delhi, India
| | - S Minhas
- Department of Medical Oncology, Medanata Hospital, Gurugram, Haryana, India
| | - D Doval
- Department of Medical Oncology, Rajiv Gandhi Cancer Institute, New Delhi, India
| |
Collapse
|
7
|
Tian Z, Tang J, Liao X, Yang Q, Wu Y, Wu G. Identification of a 9-gene prognostic signature for breast cancer. Cancer Med 2020; 9:9471-9484. [PMID: 33090721 PMCID: PMC7774725 DOI: 10.1002/cam4.3523] [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: 06/23/2020] [Revised: 08/28/2020] [Accepted: 09/18/2020] [Indexed: 12/24/2022] Open
Abstract
Breast cancer (BRCA) is the most common cancer among women and is the second leading cause of cancer death in women. In this study, we developed a 9‐gene prognostic signature to predict the prognosis of patients with BRCA. GSE20685, GSE42568, GSE20711, and GSE88770 were used as training sets. The Kaplan–Meier plot was constructed to assess survival differences and log‐rank test was performed to evaluate the statistical significance. The overall survival (OS) of patients in the low‐risk group was significantly higher than that in the high‐risk group. ROC analysis indicated that this 9‐gene signature shows good diagnostic efficiency both in OS and disease‐free survival (DFS). The 9‐gene signature was further validated through GSE16446, GSE7390, and TCGA‐BRCA datasets. We also established a nomogram that integrates clinicopathological features and 9‐gene signature. The analysis of the calibration plot showed that the nomogram has good prognostic performance. More convincingly, real‐time reverse transcription‐polymerase chain reaction (RT‐PCR) results indicated that the protective prognostic factors in BRCA patients were downregulated, whereas the dangerous prognostic factors were upregulated. The innovation of this article is not only constructing a prognostic gene signature, but also combining with clinical information to further establish a nomogram to better predict the survival probability of patients. It is worth mentioning that this signature also does not depend on other clinical factors or variables.
Collapse
Affiliation(s)
- Zelin Tian
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jianing Tang
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xing Liao
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qian Yang
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yumin Wu
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Gaosong Wu
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
8
|
Design, synthesis, and biological activity of a novel series of benzofuran derivatives against oestrogen receptor-dependent breast cancer cell lines. Bioorg Chem 2020; 95:103566. [PMID: 31935604 DOI: 10.1016/j.bioorg.2020.103566] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 12/14/2019] [Accepted: 01/02/2020] [Indexed: 01/17/2023]
Abstract
A docking study of a novel series of benzofuran derivatives with ERα was conducted. In this study, we report the synthesis of a novel series of benzofuran derivatives and evaluation of their anticancer activity in vitro against MCF-7 human breast cancer cells, as well as their potential toxicity to ER-independent MDA-MB-231 breast cancer cells, human renal epithelial HEK-293 cells, and human immortal keratinocytes (HaCaT cells) by using the MTT colorimetric assay. The screening results indicated that the target compounds exhibited anti-breast cancer activity. The target compound 2-benzoyl-3-methyl-6-[2-(morpholin-4-yl)ethoxy]benzofuran hydrochloride (4e) exhibited excellent activity against anti-oestrogen receptor-dependent breast cancer cells and low toxicity. The preliminary structure-activity relationships of the target benzofuran derivatives have been summarised. In conclusion, the novel benzofuran scaffold may be a promising lead for the development of potential oestrogen receptor inhibitors.
Collapse
|
9
|
Wu JR, Zhao Y, Zhou XP, Qin X. Estrogen receptor 1 and progesterone receptor are distinct biomarkers and prognostic factors in estrogen receptor-positive breast cancer: Evidence from a bioinformatic analysis. Biomed Pharmacother 2020; 121:109647. [DOI: 10.1016/j.biopha.2019.109647] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 11/01/2019] [Accepted: 11/01/2019] [Indexed: 12/12/2022] Open
|
10
|
Inda MA, Blok EJ, Kuppen PJK, Charehbili A, den Biezen-Timmermans EC, van Brussel A, Fruytier SE, Meershoek-Klein Kranenbarg E, Kloet S, van der Burg B, Martens JWM, Sims AH, Turnbull AK, Dixon JM, Verhaegh W, Kroep JR, van de Velde CJH, van de Stolpe A. Estrogen Receptor Pathway Activity Score to Predict Clinical Response or Resistance to Neoadjuvant Endocrine Therapy in Primary Breast Cancer. Mol Cancer Ther 2019; 19:680-689. [PMID: 31727690 DOI: 10.1158/1535-7163.mct-19-0318] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 08/08/2019] [Accepted: 11/08/2019] [Indexed: 11/16/2022]
Abstract
Endocrine therapy is important for management of patients with estrogen receptor (ER)-positive breast cancer; however, positive ER staining does not reliably predict therapy response. We assessed the potential to improve prediction of response to endocrine treatment of a novel test that quantifies functional ER pathway activity from mRNA levels of ER pathway-specific target genes. ER pathway activity was assessed on datasets from three neoadjuvant-treated ER-positive breast cancer patient cohorts: Edinburgh: 3-month letrozole, 55 pre-/2-week/posttreatment matched samples; TEAM IIa: 3- to 6-month exemestane, 49 pre-/28 posttreatment paired samples; and NEWEST: 16-week fulvestrant, 39 pretreatment samples. ER target gene mRNA levels were measured in fresh-frozen tissue (Edinburgh, NEWEST) with Affymetrix microarrays, and in formalin-fixed paraffin-embedded samples (TEAM IIa) with qRT-PCR. Approximately one third of ER-positive patients had a functionally inactive ER pathway activity score (ERPAS), which was associated with a nonresponding status. Quantitative ERPAS decreased significantly upon therapy (P < 0.001 Edinburgh and TEAM IIa). Responders had a higher pretreatment ERPAS and a larger 2-week decrease in activity (P = 0.02 Edinburgh). Progressive disease was associated with low baseline ERPAS (P = 0.03 TEAM IIa; P = 0.02 NEWEST), which did not decrease further during treatment (P = 0.003 TEAM IIa). In contrast, the staining-based ER Allred score was not significantly associated with therapy response (P = 0.2). The ERPAS identified a subgroup of ER-positive patients with a functionally inactive ER pathway associated with primary endocrine resistance. Results confirm the potential of measuring functional ER pathway activity to improve prediction of response and resistance to endocrine therapy.
Collapse
Affiliation(s)
| | - Erik J Blok
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands.,Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Peter J K Kuppen
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Ayoub Charehbili
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | - Sevgi E Fruytier
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Susan Kloet
- Leiden Genome Technology Center, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | - Andrew H Sims
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research UK Centre, MRC Institute of Genetics and Molecular Medicine, Edinburgh, United Kingdom
| | - Arran K Turnbull
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research UK Centre, MRC Institute of Genetics and Molecular Medicine, Edinburgh, United Kingdom.,Edinburgh Breast Unit, Western General Hospital, Edinburgh, United Kingdom
| | - J Michael Dixon
- Edinburgh Breast Unit, Western General Hospital, Edinburgh, United Kingdom
| | | | - Judith R Kroep
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | | | | |
Collapse
|
11
|
Li X, Wang M, Wang M, Yu X, Guo J, Sun T, Yao L, Zhang Q, Xu Y. Predictive and Prognostic Roles of Pathological Indicators for Patients with Breast Cancer on Neoadjuvant Chemotherapy. J Breast Cancer 2019; 22:497-521. [PMID: 31897326 PMCID: PMC6933033 DOI: 10.4048/jbc.2019.22.e49] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 10/11/2019] [Indexed: 02/06/2023] Open
Abstract
Currently, neoadjuvant chemotherapy is a standard therapeutic strategy for breast cancer, as it can provide timely and individualized chemo-sensitivity information and is beneficial for custom-designing subsequent treatment strategies. To accurately select candidates for neoadjuvant chemotherapy, the association between various immunohistochemical biomarkers of primary disease and tumor response to neoadjuvant chemotherapy has been investigated, and results have shown that certain pathological indicators evaluated after neoadjuvant chemotherapy are associated with long-term prognosis. The Food and Drug Administration (FDA) has recommended that complete pathological response can be used as a surrogate endpoint for neoadjuvant chemotherapy, which is related to better prognosis. Considering that residual tumor persists in the majority of patients after neoadjuvant chemotherapy, the value of various pathological indicators of residual disease in predicting the long-term outcomes is being extensively investigated. This review summarizes and compares various predictive and prognostic indicators for patients who have received neoadjuvant chemotherapy, and analyzes their efficacy in different breast cancer subtypes.
Collapse
Affiliation(s)
- Xinyan Li
- Department of Breast Surgery, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Mozhi Wang
- Department of Breast Surgery, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Mengshen Wang
- Department of Breast Surgery, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Xueting Yu
- Department of Breast Surgery, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Jingyi Guo
- Department of Breast Surgery, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Tie Sun
- Department of Breast Surgery, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Litong Yao
- Department of Breast Surgery, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Qiang Zhang
- Department of Breast Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, China
| | - Yingying Xu
- Department of Breast Surgery, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
12
|
Fiche M, Scabia V, Aouad P, Battista L, Treboux A, Stravodimou A, Zaman K, Dormoy V, Ayyanan A, Sflomos G, Brisken C. Intraductal patient-derived xenografts of estrogen receptor α-positive breast cancer recapitulate the histopathological spectrum and metastatic potential of human lesions. J Pathol 2018; 247:287-292. [PMID: 30430577 PMCID: PMC6590246 DOI: 10.1002/path.5200] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 11/05/2018] [Accepted: 11/08/2018] [Indexed: 01/24/2023]
Abstract
Estrogen receptor α‐positive (ER‐positive) or ‘luminal’ breast cancers were notoriously difficult to establish as patient‐derived xenografts (PDXs). We and others recently demonstrated that the microenvironment is critical for ER‐positive tumor cells; when grafted as single cells into milk ducts of NOD Scid gamma females, >90% of ER‐positive tumors can be established as xenografts and recapitulate many features of the human disease in vivo. This intraductal approach holds promise for personalized medicine, yet human and murine stroma are organized differently and this and other species specificities may limit the value of this model. Here, we analyzed 21 ER‐positive intraductal PDXs histopathologically. We found that intraductal PDXs vary in extent and define four histopathological patterns: flat, lobular, in situ and invasive, which occur in pure and combined forms. The intraductal PDXs replicate earlier stages of tumor development than their clinical counterparts. Micrometastases are already detected when lesions appear in situ. Tumor extent, histopathological patterns and micrometastatic load correlate with biological properties of their tumors of origin. Our findings add evidence to the validity of the intraductal model for in vivo studies of ER‐positive breast cancer and raise the intriguing possibility that tumor cell dissemination may occur earlier than currently thought. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Maryse Fiche
- International Cancer Prevention Institute, Epalinges, Switzerland
| | - Valentina Scabia
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Patrick Aouad
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Laura Battista
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Assia Treboux
- Centre Hospitalier Universitaire Vaudois, University Hospital of Lausanne, Lausanne, Switzerland
| | - Athina Stravodimou
- Centre Hospitalier Universitaire Vaudois, University Hospital of Lausanne, Lausanne, Switzerland
| | - Khalil Zaman
- Centre Hospitalier Universitaire Vaudois, University Hospital of Lausanne, Lausanne, Switzerland
| | -
- Réseau Lausannois du Sein (RLS), Lausanne, Switzerland
| | - Valerian Dormoy
- INSERM UMR_S1250, Université de Reims Champagne-Ardenne (URCA), Reims, France
| | - Ayyakkannu Ayyanan
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - George Sflomos
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Cathrin Brisken
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| |
Collapse
|
13
|
Martínez-Pérez C, Turnbull AK, Dixon JM. The evolving role of receptors as predictive biomarkers for metastatic breast cancer. Expert Rev Anticancer Ther 2018; 19:121-138. [PMID: 30501540 DOI: 10.1080/14737140.2019.1552138] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION In breast cancer, estrogen receptor (ER) and human epidermal growth factor receptor 2 (HER2) are essential biomarkers to predict response to endocrine and anti-HER2 therapies, respectively. In metastatic breast cancer, the use of these receptors and targeted therapies present additional challenges: temporal heterogeneity, together with limited sampling methodologies, hinders receptor status assessment, and the constant evolution of the disease invariably leads to resistance to treatment. Areas covered: This review summarizes the genomic abnormalities in ER and HER2, such as mutations, amplifications, translocations, and alternative splicing, emerging as novel biomarkers that provide an insight into underlying mechanisms of resistance and hold potential predictive value to inform treatment selection. We also describe how liquid biopsies for sampling of circulating markers and ultrasensitive detection technologies have emerged which complement ongoing efforts for biomarker discovery and analysis. Expert commentary: While evidence suggests that genomic aberrations in ER and HER2 could contribute to meeting the pressing need for better predictive biomarkers, efforts need to be made to standardize assessment methods and better understand the resistance mechanisms these markers denote. Taking advantage of emerging technologies, research in upcoming years should include prospective trials incorporating these predictors into the study design to validate their potential clinical value.
Collapse
Affiliation(s)
- Carlos Martínez-Pérez
- a Breast Cancer Now Edinburgh Team, Institute of Genetics and Molecular Medicine , University of Edinburgh, Western General Hospital , Edinburgh , UK
| | - Arran K Turnbull
- a Breast Cancer Now Edinburgh Team, Institute of Genetics and Molecular Medicine , University of Edinburgh, Western General Hospital , Edinburgh , UK
| | - J Michael Dixon
- a Breast Cancer Now Edinburgh Team, Institute of Genetics and Molecular Medicine , University of Edinburgh, Western General Hospital , Edinburgh , UK.,b Edinburgh Breast Unit , Western General Hospital , Edinburgh , UK
| |
Collapse
|
14
|
Liang X, Briaux A, Becette V, Benoist C, Boulai A, Chemlali W, Schnitzler A, Baulande S, Rivera S, Mouret-Reynier MA, Bouvet LV, De La Motte Rouge T, Lemonnier J, Lerebours F, Callens C. Molecular profiling of hormone receptor-positive, HER2-negative breast cancers from patients treated with neoadjuvant endocrine therapy in the CARMINA 02 trial (UCBG-0609). J Hematol Oncol 2018; 11:124. [PMID: 30305115 PMCID: PMC6180434 DOI: 10.1186/s13045-018-0670-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 09/26/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Postmenopausal women with large, hormone receptor (HR)-positive/HER2-negative and low-proliferative breast cancer derived a benefit from neoadjuvant endocrine therapy (NET) in the CARMINA02 trial. This study was designed to correlate gene expression and mutation profiles with both response to NET and prognosis. METHODS Gene expression profiling using RNA sequencing was performed in 86 pre-NET and post-NET tumor samples. Targeted next-generation sequencing of 91 candidate breast cancer-associated genes was performed on DNA samples from 89 patients. Molecular data were correlated with radiological response and relapse-free survival. RESULTS The transcriptional profile of tumors to NET in responders involved immune-associated genes enriched in activated Th1 pathway, which remained unchanged in non-responders. Immune response was confirmed by analysis of tumor-infiltrating lymphocytes (TILs). The percentage of TILs was significantly increased post-NET compared to pre-NET samples in responders (p = 0.0071), but not in non-responders (p = 0.0938). Gene expression revealed that lipid metabolism was the main molecular function related to prognosis, while PPARγ is the most important upstream regulator gene. The most frequently mutated genes were PIK3CA (48.3%), CDH1 (20.2%), PTEN (15.7%), TP53 (10.1%), LAMA2 (10.1%), BRCA2 (9.0%), MAP3K1 (7.9%), ALK (6.7%), INPP4B (6.7%), NCOR1 (6.7%), and NF1 (5.6%). Cell cycle and apoptosis pathway and PIK3CA/AKT/mTOR pathway were altered significantly more frequently in non-responders than in responders (p = 0.0017 and p = 0.0094, respectively). The average number of mutations per sample was significantly higher in endocrine-resistant tumors (2.88 vs. 1.64, p = 0.03), but no difference was observed in terms of prognosis. ESR1 hotspot mutations were detected in 3.4% of treatment-naive tumors. CONCLUSIONS The Th1-related immune system and lipid metabolism appear to play key roles in the response to endocrine therapy and prognosis in HR-positive/HER2-negative breast cancer. Deleterious somatic mutations in the cell cycle and apoptosis pathway and PIK3CA/AKT/mTOR pathway may be relevant for clinical management. TRIAL REGISTRATION This trial is registered with ClinicalTrials.gov ( NCT00629616 ) on March 6, 2008, retrospectively registered.
Collapse
Affiliation(s)
- Xu Liang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Breast Oncology, Peking University Cancer Hospital & Institute, Beijing, China.,Pharmacogenomic Unit, Department of Genetics, Curie Institute, PSL Research University, Paris, France
| | - Adrien Briaux
- Pharmacogenomic Unit, Department of Genetics, Curie Institute, PSL Research University, Paris, France
| | - Véronique Becette
- Department of Biopathology, Curie Institute, René Huguenin Hospital, Saint-Cloud, France
| | - Camille Benoist
- Pharmacogenomic Unit, Department of Genetics, Curie Institute, PSL Research University, Paris, France
| | - Anais Boulai
- Pharmacogenomic Unit, Department of Genetics, Curie Institute, PSL Research University, Paris, France
| | - Walid Chemlali
- Pharmacogenomic Unit, Department of Genetics, Curie Institute, PSL Research University, Paris, France
| | - Anne Schnitzler
- Pharmacogenomic Unit, Department of Genetics, Curie Institute, PSL Research University, Paris, France
| | - Sylvain Baulande
- Institut Curie Genomics of Excellence (ICGex) Platform, Curie Institute, PSL Research University, Paris, France
| | - Sofia Rivera
- Department of Radiotherapy, Gustave Roussy, Villejuif, France
| | | | | | | | | | - Florence Lerebours
- Department of Medical Oncology, Curie Institute, René Huguenin Hospital, Saint-Cloud, France
| | - Céline Callens
- Pharmacogenomic Unit, Department of Genetics, Curie Institute, PSL Research University, Paris, France.
| |
Collapse
|
15
|
Sato N, Masuda N, Morimoto T, Ueno T, Kanbayashi C, Kaneko K, Yasojima H, Saji S, Sasano H, Morita S, Ohno S, Toi M. Neoadjuvant endocrine therapy with exemestane followed by response-guided combination therapy with low-dose cyclophosphamide in postmenopausal patients with estrogen receptor-positive breast cancer: A multicenter, open-label, phase II study. Cancer Med 2018; 7:3044-3056. [PMID: 29905023 PMCID: PMC6051169 DOI: 10.1002/cam4.1600] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 05/08/2018] [Accepted: 05/12/2018] [Indexed: 01/28/2023] Open
Abstract
Patients with estrogen receptor (ER)‐positive breast cancer are less likely to achieve a pathological complete response (pCR) with neoadjuvant chemotherapy. Neoadjuvant endocrine therapy may be more appropriate than neoadjuvant chemotherapy in these hormone‐sensitive patients. Most patients with ER‐positive breast cancer are postmenopausal, and therefore, generally older and less able to tolerate chemotherapy. We aimed to investigate the efficacy and safety of tailored neoadjuvant endocrine and chemoendocrine therapy for postmenopausal breast cancer patients. Untreated patients with primary invasive ER‐positive, HER2‐negative, stage I‐IIIA breast cancer, and Ki67 index ≤30% were enrolled. Patients received exemestane 25 mg/d for 12 weeks. Based on clinical response and change in Ki67 index, assessed at 8‐12 weeks, patients with complete response (CR), partial response (PR) with Ki67 index ≤5% after treatment, or stable disease (SD) with Ki67 index ≤5% before and after treatment were defined as responders. For the subsequent 24 weeks, responders continued exemestane monotherapy (group A), and nonresponders received exemestane 25 mg/d plus cyclophosphamide 50 mg/d (group B). The primary endpoint was clinical response at weeks 24 and 36. A total of 59 patients (median age, 69 years) started initial exemestane monotherapy. After exclusion of three patients who discontinued during this period, 56 remained enrolled to receive subsequent treatment. Clinical response rates (CR and PR) and 95% CI at weeks 24 and 36 were 85% (12/14; 57.2%‐98.2%) and 71% (10/14; 41.9%‐91.6%), respectively, in group A; and 54% (23/42; 38.7%‐70.2%) and 71% (30/42; 55.4%‐84.3%), respectively, in group B. At week 36, no significant difference was found in median Ki67 index between the groups (3.5% and 4.0%). There were no treatment‐related deaths. We found that clinical response comparable to that of responders was achieved in nonresponders after addition of cyclophosphamide to the initial endocrine therapy.
Collapse
Affiliation(s)
- Nobuaki Sato
- Department of Breast Oncology, Niigata Cancer Center Hospital, Niigata, Japan
| | - Norikazu Masuda
- Department of Surgery, Breast Oncology, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Takashi Morimoto
- Department of Breast Surgery, Yao Municipal Hospital, Osaka, Japan
| | - Takayuki Ueno
- Breast Surgical Oncology, Breast Oncology Center, Cancer Institute Hospital, Tokyo, Japan
| | - Chizuko Kanbayashi
- Department of Breast Oncology, Niigata Cancer Center Hospital, Niigata, Japan
| | - Koji Kaneko
- Department of Breast Oncology, Niigata Cancer Center Hospital, Niigata, Japan
| | - Hiroyuki Yasojima
- Department of Surgery, Breast Oncology, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Shigehira Saji
- Department of Medical Oncology, Fukushima Medical University, Fukushima, Japan
| | | | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shinji Ohno
- Breast Oncology Center, Cancer Institute Hospital, Tokyo, Japan
| | - Masakazu Toi
- Department of Surgery (Breast Surgery), Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
16
|
Brantley KD, Kjærsgaard A, Cronin-Fenton D, Yacoub R, Nielsen AS, Lauridsen KL, Hamilton-Dutoit S, Lash TL. Stanniocalcin Expression as a Predictor of Late Breast Cancer Recurrence. Cancer Epidemiol Biomarkers Prev 2018; 27:653-659. [PMID: 29593009 DOI: 10.1158/1055-9965.epi-17-0905] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 12/12/2017] [Accepted: 03/20/2018] [Indexed: 11/16/2022] Open
Abstract
Background: Expression of human paracrine hormones stanniocalcin 1 (STC1) and stanniocalcin 2 (STC2) may potentiate late breast cancer recurrence. We tested the hypothesis that expression of STC1 and STC2 in primary breast tumors is more strongly associated with late versus early recurrences.Methods: A total of 541 estrogen receptor-positive, tamoxifen-treated (ER+/TAM+) and 300 ER-negative, tamoxifen-untreated (ER-/TAM-) breast cancer patients who experienced recurrence within 10 years of primary diagnosis and matched recurrence-free controls were selected from a cohort of 11,251 Danish breast cancer patients diagnosed with stage I, II, or III breast cancer during 1985 to 2001. The association between IHC expression of STC1 and STC2 in primary breast tumor tissue microarrays and breast cancer recurrence was evaluated within median time to recurrence quintiles.Results: The association between STC1 expression, dichotomized as positive or negative, and recurrence was strongly positive for the final time quintile (6-10 years postdiagnosis) in the ER+/TAM+ group [aOR = 2.70; 95% confidence interval (CI): 1.22-5.98]. Regression of the log ORs relating dichotomous STC1 and STC2 expression to recurrence by median time to recurrence (year) resulted in a relatively large positive effect estimate for STC1 (β = 0.16; 95% CI, -0.03-0.36) and a near-null positive effect estimate for STC2 (β = 0.04; 95% CI, -0.14-0.21).Conclusions: Our results suggest a stronger association between primary tumor STC1 expression and late recurrence, as opposed to early recurrence, although no clear trend was apparent.Impact: STC1 expression in the primary tumor may potentiate late recurrences, suggesting dormancy pathways that merit further investigation. Cancer Epidemiol Biomarkers Prev; 27(6); 653-9. ©2018 AACR.
Collapse
Affiliation(s)
- Kristen D Brantley
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia.
| | - Anders Kjærsgaard
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Rami Yacoub
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia
| | - Anja S Nielsen
- Institute of Pathology, Aarhus University Hospital, Aarhus, Denmark
| | | | | | - Timothy L Lash
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia.,Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark.,Winship Cancer Institute, Atlanta, Georgia
| |
Collapse
|
17
|
Sellers ZP, Bujko K, Schneider G, Kucia M, Ratajczak MZ. Novel evidence that pituitary sex hormones regulate migration, adhesion, and proliferation of embryonic stem cells and teratocarcinoma cells. Oncol Rep 2017; 39:851-859. [PMID: 29207191 PMCID: PMC5783624 DOI: 10.3892/or.2017.6108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 11/07/2017] [Indexed: 12/22/2022] Open
Abstract
The pituitary sex hormones (SexHs): follicle-stimulating hormone (FSH), luteinizing hormone (LH), and prolactin (PRL) regulate several functions crucial for reproduction, including oogenesis, spermatogenesis, and lactation. An important source of prolactin-like hormones, known as lactogens, is the placenta, and lactogens bind to the PRL receptor (PRLR) with high affinity and thereby mimic the actions of PRL. Recently, it has been demonstrated that pituitary SexHs were involved in metastatic lung cancer, certain sarcomas, and leukemia. In the present study we aimed to investigate whether FSH, LH, and PRL were able to stimulate stem cells involved in early development. To address this issue we employed a murine embryonic stem cell line (ES-D3) as well as two teratocarcinoma cell lines, P19 (murine) and NTera2 (human). We determined that all these cells expressed SexH receptors at the mRNA and protein levels and that stimulation of these receptors induced phosphorylation of p42/44 MAPK, p38 MAPK, and AKT. Moreover, ES-D3, P19, and NTera2 cells responded with increased migration and adhesion to physiological concentrations of pituitary SexHs. In view of these findings we proposed that maternal-derived pituitary SexHs regulate the biology of stem cells involved in early development.
Collapse
Affiliation(s)
- Zachariah Payne Sellers
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Kamila Bujko
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Gabriela Schneider
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| | - Magdalena Kucia
- Department of Regenerative Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Mariusz Z Ratajczak
- Stem Cell Institute at James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
18
|
Analytical validation of a 12-gene molecular test for the prediction of distant recurrence in breast cancer. Future Sci OA 2017; 3:FSO221. [PMID: 28884016 PMCID: PMC5583651 DOI: 10.4155/fsoa-2017-0051] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 05/18/2017] [Indexed: 11/30/2022] Open
Abstract
Aim: To validate the analytical performance of a 12-gene molecular assay that predicts distant recurrence for early-stage ER+/HER2- invasive breast cancer as run within a central reference laboratory. Materials & methods: Formalin-fixed paraffin-embedded breast resections were evaluated by quantitative reverse transcription polymerase chain reaction for the expression of eight target genes, three housekeeper genes and one control gene to assess for DNA contamination. Results: The assay results were highly correlated with a validated reference laboratory. The assay had a broad linear range for input RNA, with similar amplicon efficiencies for target and housekeeper genes. The assay test was highly reproducible, with comparable inter- and intrabatch precision to the reference laboratory. Conclusion: These studies demonstrate that the 12-gene molecular assay is highly robust and accurate. Several tests have been developed to aid in breast cancer treatment decisions by assessing the likelihood that an individual’s breast cancer will recur. One such test is a 12-gene molecular assay. This test provides a clinical score that combines molecular information from genes within the tumor with clinical information to determine whether a breast cancer is at high or low risk of recurrence. The data presented here show that this test is very reproducible and robust, making it appropriate and reliable for clinical use.
Collapse
|
19
|
Reinert T, Saad ED, Barrios CH, Bines J. Clinical Implications of ESR1 Mutations in Hormone Receptor-Positive Advanced Breast Cancer. Front Oncol 2017; 7:26. [PMID: 28361033 PMCID: PMC5350138 DOI: 10.3389/fonc.2017.00026] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 02/14/2017] [Indexed: 12/21/2022] Open
Abstract
Hormone receptor-positive breast cancer is the most frequent breast cancer subtype. Endocrine therapy (ET) targeting the estrogen receptor (ER) pathway represents the main initial therapeutic approach. The major strategies include estrogen deprivation and the use of selective estrogen modulators or degraders, which show efficacy in the management of metastatic and early-stage disease. However, clinical resistance associated with progression of disease remains a significant therapeutic challenge. Mutations of the ESR1 gene, which encodes the ER, have been increasingly recognized as an important mechanism of ET resistance, with a prevalence that ranges from 11 to 39%. The majority of these mutations are located within the ligand-binding domain and result in an estrogen-independent constitutive activation of the ER and, therefore, resistance to estrogen deprivation therapy such as aromatase inhibition. ESR1 mutations, most often detected from liquid biopsies, have been consistently associated with a worse outcome and are being currently evaluated as a potential biomarker to guide therapeutic decisions. At the same time, targeted therapy directed to ESR1-mutated clones is an appealing concept with preclinical and clinical work in progress.
Collapse
Affiliation(s)
- Tomas Reinert
- Hospital de Câncer Mãe de Deus, Universidade Federal do Rio Grande do Sul , Porto Alegre , Brazil
| | | | | | - José Bines
- Instituto Nacional de Câncer , Rio de Janeiro , Brazil
| |
Collapse
|
20
|
Ma CX, Gao F, Luo J, Northfelt DW, Goetz M, Forero A, Hoog J, Naughton M, Ademuyiwa F, Suresh R, Anderson KS, Margenthaler J, Aft R, Hobday T, Moynihan T, Gillanders W, Cyr A, Eberlein TJ, Hieken T, Krontiras H, Guo Z, Lee MV, Spies NC, Skidmore ZL, Griffith OL, Griffith M, Thomas S, Bumb C, Vij K, Bartlett CH, Koehler M, Al-Kateb H, Sanati S, Ellis MJ. NeoPalAna: Neoadjuvant Palbociclib, a Cyclin-Dependent Kinase 4/6 Inhibitor, and Anastrozole for Clinical Stage 2 or 3 Estrogen Receptor-Positive Breast Cancer. Clin Cancer Res 2017; 23:4055-4065. [PMID: 28270497 DOI: 10.1158/1078-0432.ccr-16-3206] [Citation(s) in RCA: 249] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 12/19/2016] [Accepted: 03/01/2017] [Indexed: 01/15/2023]
Abstract
Purpose: Cyclin-dependent kinase (CDK) 4/6 drives cell proliferation in estrogen receptor-positive (ER+) breast cancer. This single-arm phase II neoadjuvant trial (NeoPalAna) assessed the antiproliferative activity of the CDK4/6 inhibitor palbociclib in primary breast cancer as a prelude to adjuvant studies.Experimental Design: Eligible patients with clinical stage II/III ER+/HER2- breast cancer received anastrozole 1 mg daily for 4 weeks (cycle 0; with goserelin if premenopausal), followed by adding palbociclib (125 mg daily on days 1-21) on cycle 1 day 1 (C1D1) for four 28-day cycles unless C1D15 Ki67 > 10%, in which case patients went off study due to inadequate response. Anastrozole was continued until surgery, which occurred 3 to 5 weeks after palbociclib exposure. Later patients received additional 10 to 12 days of palbociclib (Cycle 5) immediately before surgery. Serial biopsies at baseline, C1D1, C1D15, and surgery were analyzed for Ki67, gene expression, and mutation profiles. The primary endpoint was complete cell cycle arrest (CCCA: central Ki67 ≤ 2.7%).Results: Fifty patients enrolled. The CCCA rate was significantly higher after adding palbociclib to anastrozole (C1D15 87% vs. C1D1 26%, P < 0.001). Palbociclib enhanced cell-cycle control over anastrozole monotherapy regardless of luminal subtype (A vs. B) and PIK3CA status with activity observed across a broad range of clinicopathologic and mutation profiles. Ki67 recovery at surgery following palbociclib washout was suppressed by cycle 5 palbociclib. Resistance was associated with nonluminal subtypes and persistent E2F-target gene expression.Conclusions: Palbociclib is an active antiproliferative agent for early-stage breast cancer resistant to anastrozole; however, prolonged administration may be necessary to maintain its effect. Clin Cancer Res; 23(15); 4055-65. ©2017 AACR.
Collapse
Affiliation(s)
- Cynthia X Ma
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri.
| | - Feng Gao
- Division of Public Health Science, Siteman Cancer Center Biostatistics Core, Washington University School of Medicine, St. Louis, Missouri
| | - Jingqin Luo
- Division of Public Health Science, Siteman Cancer Center Biostatistics Core, Washington University School of Medicine, St. Louis, Missouri
| | - Donald W Northfelt
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, Arizona
| | - Matthew Goetz
- Department of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Andres Forero
- Department of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jeremy Hoog
- 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
| | - Foluso Ademuyiwa
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Rama Suresh
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Karen S Anderson
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, Arizona
| | - 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
| | - Timothy Hobday
- Department of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Timothy Moynihan
- Department of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - William Gillanders
- Section of Endocrine and Oncologic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Amy Cyr
- Section of Endocrine and Oncologic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Timothy J Eberlein
- Section of Endocrine and Oncologic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Tina Hieken
- Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | - Helen Krontiras
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Zhanfang Guo
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Michelle V Lee
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Nicholas C Spies
- 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
| | - Obi L Griffith
- Division of Oncology, Department of Internal 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
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri.,Department of Genetics, Washington University School of Medicine, St. Louis, Missouri
| | - Shana Thomas
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Caroline Bumb
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Kiran Vij
- Division of Oncology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri
| | | | | | - Hussam Al-Kateb
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Souzan Sanati
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Matthew J Ellis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
21
|
Association of DW/DCE-MRI features with prognostic factors in breast cancer. Int J Biol Markers 2017; 32:e118-e125. [PMID: 27646773 DOI: 10.5301/jbm.5000230] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2016] [Indexed: 01/03/2023]
Abstract
BACKGROUND Through analyzing apparent diffusion coefficient (ADC) values and morphological evaluations, this research aimed to study how magnetic resonance imaging (MRI)-based breast lesion characteristics can enhance the diagnosis and prognosis of breast cancer. METHODS: A total of 118 breast lesions, including 50 benign and 68 malignant lesions, from 106 patients were analyzed. All lesions were measured with both diffusion-weighted (DW) and dynamic contrast-enhanced (DCE) MRI. The average ADC of breast lesions was analyzed at b values of 600, 800 and 1,000 s/mm2. Lesion margins, lesion enhancement patterns, and dynamic curves were also investigated. The relations between MRI-based features and molecular prognostic factors were evaluated using Spearman's rank correlation analysis. RESULTS: A b value of 800 s/mm2 was used to distinguish malignant from benign breast lesions, with an ADC cutoff value of 1.365 × 10-3 mm2/s. The average ADC value between invasive ductal carcinoma (IDC) and ductal carcinoma in situ (DCIS) was significantly different. Malignant lesions were more likely to have spiculated margins, heterogeneous enhancement and washout curves. On the other hand, DCIS was more likely to have spiculated margins, heterogeneous/rim enhancement and plateau/washout dynamic curves. A significant negative correlation was found between progesterone receptor (PR) status and dynamic imaging (p = 0.027), while a significant positive correlation was found between Ki-67 status and lesion enhancement (p = 0.045). CONCLUSIONS: Both ADC values and MRI morphological assessment could be used to distinguish malignant breast lesions from benign ones.
Collapse
|
22
|
Oza A, Ma CX. New Insights in Estrogen Receptor (ER) Biology and Implications for Treatment. CURRENT BREAST CANCER REPORTS 2017. [DOI: 10.1007/s12609-017-0231-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
23
|
Extended adjuvant endocrine therapy in hormone-receptor-positive early breast cancer. Curr Opin Oncol 2016; 28:455-460. [DOI: 10.1097/cco.0000000000000323] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
24
|
Identifying biomarkers to select patients with early breast cancer suitable for extended adjuvant endocrine therapy. Curr Opin Oncol 2016; 28:461-468. [PMID: 27606693 DOI: 10.1097/cco.0000000000000324] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW In this review, we summarize recent and current biomarkers and assays that are being considered in the selection of suitable patients with estrogen receptor-positive early breast cancer for extended (years 5-10) adjuvant endocrine therapy (AET). RECENT FINDINGS Women with estrogen receptor-positive early-stage breast cancer (65% of cases) continue to have late risk for distant recurrence extending beyond 5 years from surgery. Recent large trials have consistently demonstrated improvement for prolonging endocrine therapy. However, endocrine therapy can cause women bothersome side effects and can negatively impact quality of life. Determining which patients remain at risk for disease recurrence and predicting which of these patients would derive the most benefit from the addition of extended AET are key issues faced by patients and oncologists today. A number of predictive molecular assays have been developed and are being considered as tools to be used in guiding the implementation of adjuvant systemic therapy. SUMMARY The future holds much promise and as more information and understanding is acquired, treatment regimens will increasingly incorporate clinically validated biomarker assays in the decision-making process that will be of great benefit to these patients. Proving clinical utility, though, will ultimately decide their implementation.
Collapse
|