1
|
Mahrhofer M, Reichert R, Siegwart LC, Russe E, Schoeller T, Wechselberger G, Weitgasser L. Risk of perioperative hormonal breast cancer therapy for microvascular flap complications in breast reconstruction. J Plast Reconstr Aesthet Surg 2023; 85:143-148. [PMID: 37487260 DOI: 10.1016/j.bjps.2023.07.004] [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: 07/17/2022] [Revised: 06/11/2023] [Accepted: 07/02/2023] [Indexed: 07/26/2023]
Abstract
BACKGROUND Hormone therapy with selective estrogen modulators (tamoxifen) and aromatase inhibitors is commonly used in the treatment of breast cancer. While the increased risk for thromboembolic events has been known since their early application, the potential risk in microsurgical breast reconstruction is still debated. This study aimed to evaluate the risk for microvascular flap complications in patients with perioperative hormone therapy. METHODS All patients who underwent microsurgical breast reconstruction with a deep inferior epigastric perforator (DIEP) or transverse myocutaneous gracilis flap at our institution between March 2010 and November 2020 were retrospectively identified in our records. Patients were grouped according to the type and use of perioperative hormone therapy. Flap-related thromboembolic events, flap loss, and revision procedures were compared and analyzed between groups. Risk factors associated with postoperative microsurgical complications were determined. RESULTS A total of 560 patients (656 flaps) were included in our analysis. One hundred ninety-eight patients (224 flaps) received perioperative hormone therapy (35.4%) and 50 (8.9%) postoperative microsurgical events occurred. Tamoxifen and aromatase inhibitors were not associated with postoperative microsurgical events (p = 0.254), full flap loss (p = 0.702), or partial flap loss (p = 0.916). Patients receiving DIEP flaps had a higher risk for postoperative microsurgical complications (OR 2.36, p = 0.004) and partial flap loss (OR 14.66, p < 0.001). A BMI > 30 was associated with an increased risk for partial flap loss (OR 4.2; p < 0.001) CONCLUSION: This article presents one of the largest single-center datasets for the risks of hormone therapy in microsurgical breast reconstruction. Our results show that perioperative hormone therapy does not increase the risk for microsurgical complications. The findings of our study do challenge the common practice of discontinued hormone therapy before microsurgical breast reconstruction.
Collapse
Affiliation(s)
- Maximilian Mahrhofer
- Department of Plastic and Reconstructive Surgery, Marienhospital Stuttgart, Teaching Hospital of the Eberhard Karls University Tuebingen, Germany.
| | - Raphael Reichert
- Department of Plastic and Reconstructive Surgery, Marienhospital Stuttgart, Teaching Hospital of the Eberhard Karls University Tuebingen, Germany
| | - Laura Cosima Siegwart
- Department of Hand, Plastic and Reconstructive Surgery, Microsurgery, Burn Center, BG Trauma Center Ludwigshafen, Hand and Plastic Surgery, University of Heidelberg, Ludwigshafen, Germany
| | - Elisabeth Russe
- Department of Plastic and Reconstructive Surgery, Hospital of the Brothers of St. John of God, Paracelsus Medical University Salzburg, Austria
| | - Thomas Schoeller
- Department of Plastic and Reconstructive Surgery, Marienhospital Stuttgart, Teaching Hospital of the Eberhard Karls University Tuebingen, Germany
| | - Gottfried Wechselberger
- Department of Plastic and Reconstructive Surgery, Hospital of the Brothers of St. John of God, Paracelsus Medical University Salzburg, Austria
| | - Laurenz Weitgasser
- Department of Plastic and Reconstructive Surgery, Marienhospital Stuttgart, Teaching Hospital of the Eberhard Karls University Tuebingen, Germany
| |
Collapse
|
2
|
Phoenix JT, Budreika A, Kostlan RJ, Hwang JH, Fanning SW, Kregel S. Editorial: Hormone resistance in cancer. Front Endocrinol (Lausanne) 2023; 14:1272932. [PMID: 37693345 PMCID: PMC10484586 DOI: 10.3389/fendo.2023.1272932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 08/17/2023] [Indexed: 09/12/2023] Open
Affiliation(s)
- John T. Phoenix
- Department of Cancer Biology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
- Integrated Program in Biomedical Science, Biochemistry, Molecular and Cancer Biology, Loyola University Chicago, Maywood, IL, United States
| | - Audris Budreika
- Department of Cancer Biology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
| | - Raymond J. Kostlan
- Department of Cancer Biology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
- Integrated Program in Biomedical Science, Biochemistry, Molecular and Cancer Biology, Loyola University Chicago, Maywood, IL, United States
| | - Justin H. Hwang
- Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Sean W. Fanning
- Department of Cancer Biology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
| | - Steven Kregel
- Department of Cancer Biology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
| |
Collapse
|
3
|
Aquaporin 5 (AQP5) expression in breast cancer and its clinicopathological characteristics. PLoS One 2023; 18:e0270752. [PMID: 36706090 PMCID: PMC9882752 DOI: 10.1371/journal.pone.0270752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 06/16/2022] [Indexed: 01/28/2023] Open
Abstract
The role of aquaporin water channels (AQPs) has become an area of great interest in human carcinogenesis. In this report, we have demonstrated the expression of AQP5 in breast cancer by analyzing 591 tissue samples with 7-year follow-ups. By immunochemistry analysis, AQP5 overexpression was observed in 36% (212/591 cases). Then, we have focused on the clinicopathologic variables among cancer tissue samples with strong AQP5 expression (3+ expression, 60/591 cases). The strong AQP5 expression was positively correlated with tumor grade in BCs (p<0.001) and was more frequent in ER/PR-negative BCs than positive ones (14.9% vs. 3.3% and 13.1% vs. 4.8%, respectively, both p<0.001), while Her2/neu-positive status was positively correlated with strong expression of AQP5 (p = 0.005). Of note, breast cancer patients with positive AQP expression (212/591 cases) showed a less favorable breast cancer specific survival rate over 7 years of follow and we further conclude that AQP5 expression is an independent molecular marker associated with worse clinical outcomes. By fluorescence in situ hybridization (FISH), we have identified evidence of gene amplification in 3 of 30 readable breast cancer and further conclude that, in breast cancer, at least some part of AQP5 overexpression is associated with an aberration in the genome level.
Collapse
|
4
|
Cheng X, Zhao JX, Dong F, Cao XC. ARID1A Mutation in Metastatic Breast Cancer: A Potential Therapeutic Target. Front Oncol 2021; 11:759577. [PMID: 34804958 PMCID: PMC8599951 DOI: 10.3389/fonc.2021.759577] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/15/2021] [Indexed: 12/05/2022] Open
Abstract
Distant metastasis is the principal cause of mortality for breast cancer patients. Targeting specific mutations that have been acquired during the evolution process of advanced breast cancer is a potential means of enhancing the clinical efficacy of treatment strategies. In metastatic breast cancer, ARID1A is the most prevalent mutation of the SWI/SNF complex, which regulates DNA repair, recombination, and gene transcription. The low expression of ARID1A is associated with poor disease-free survival and overall survival of patients with luminal A or HER2-rich breast cancer. In addition, ARID1A plays a prominent role in maintaining luminal characteristics and has an advantage for identifying responses to treatment, including endocrine therapies, HDAC inhibitors and CDK4/6 inhibitors. The therapeutic vulnerabilities initiated by ARID1A alterations encourage us to explore new approaches to cope with ARID1A mutant-related drug resistance or metastasis. In this review, we describe the mutation profiles of ARID1A in metastatic breast cancer and the structure and function of ARID1A and the SWI/SNF complex as well as discuss the potential mechanisms of ARID1A-mediated endocrine resistance and therapeutic potential.
Collapse
Affiliation(s)
- Xuan Cheng
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Jian-Xiong Zhao
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Feng Dong
- Department of Neurosurgery, Tianjin Medical University General Hospital and Laboratory of Neuro-Oncology, Tianjin Neurological Institute, Tianjin, China
- State Key Laboratory of Experimental Hematology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, Department of Cell Biology, Tianjin Medical University, Tianjin, China
| | - Xu-Chen Cao
- The First Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| |
Collapse
|
5
|
Roarty K, Echeverria GV. Laboratory Models for Investigating Breast Cancer Therapy Resistance and Metastasis. Front Oncol 2021; 11:645698. [PMID: 33777805 PMCID: PMC7988094 DOI: 10.3389/fonc.2021.645698] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 01/28/2021] [Indexed: 01/16/2023] Open
Abstract
While numerous therapies are highly efficacious in early-stage breast cancers and in particular subsets of breast cancers, therapeutic resistance and metastasis unfortunately arise in many patients. In many cases, tumors that are resistant to standard of care therapies, as well as tumors that have metastasized, are treatable but incurable with existing clinical strategies. Both therapy resistance and metastasis are multi-step processes during which tumor cells must overcome diverse environmental and selective hurdles. Mechanisms by which tumor cells achieve this are numerous and include acquisition of invasive and migratory capabilities, cell-intrinsic genetic and/or epigenetic adaptations, clonal selection, immune evasion, interactions with stromal cells, entering a state of dormancy or senescence, and maintaining self-renewal capacity. To overcome therapy resistance and metastasis in breast cancer, the ability to effectively model each of these mechanisms in the laboratory is essential. Herein we review historic and the current state-of-the-art laboratory model systems and experimental approaches used to investigate breast cancer metastasis and resistance to standard of care therapeutics. While each model system has inherent limitations, they have provided invaluable insights, many of which have translated into regimens undergoing clinical evaluation. We will discuss the limitations and advantages of a variety of model systems that have been used to investigate breast cancer metastasis and therapy resistance and outline potential strategies to improve experimental modeling to further our knowledge of these processes, which will be crucial for the continued development of effective breast cancer treatments.
Collapse
Affiliation(s)
- Kevin Roarty
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, United States
| | - Gloria V Echeverria
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, United States.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, United States.,Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, United States.,Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
6
|
Ross DS. Breast. Genomic Med 2020. [DOI: 10.1007/978-3-030-22922-1_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
7
|
The ER-alpha mutation Y537S confers Tamoxifen-resistance via enhanced mitochondrial metabolism, glycolysis and Rho-GDI/PTEN signaling: Implicating TIGAR in somatic resistance to endocrine therapy. Aging (Albany NY) 2019; 10:4000-4023. [PMID: 30573703 PMCID: PMC6326696 DOI: 10.18632/aging.101690] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 11/29/2018] [Indexed: 01/01/2023]
Abstract
Naturally-occurring somatic mutations in the estrogen receptor gene (ESR1) have been previously implicated in the clinical development of resistance to hormonal therapies, such as Tamoxifen. For example, the somatic mutation Y537S has been specifically associated with acquired endocrine resistance. Briefly, we recombinantly-transduced MCF7 cells with a lentiviral vector encoding ESR1 (Y537S). As a first step, we confirmed that MCF7-Y537S cells are indeed functionally resistant to Tamoxifen, as compared with vector alone controls. Importantly, further phenotypic characterization of Y537S cells revealed that they show increased resistance to Tamoxifen-induced apoptosis, allowing them to form mammospheres with higher efficiency, in the presence of Tamoxifen. Similarly, Y537S cells had elevated basal levels of ALDH activity, a marker of “stemness”, which was also Tamoxifen-resistant. Metabolic flux analysis of Y537S cells revealed a hyper-metabolic phenotype, with significantly increased mitochondrial respiration and high ATP production, as well as enhanced aerobic glycolysis. Finally, to understand which molecular signaling pathways that may be hyper-activated in Y537S cells, we performed unbiased label-free proteomics analysis. Our results indicate that TIGAR over-expression and the Rho-GDI/PTEN signaling pathway appear to be selectively activated by the Y537S mutation. Remarkably, this profile is nearly identical in MCF7-TAMR cells; these cells were independently-generated in vitro, suggesting a highly conserved mechanism underlying Tamoxifen-resistance. Importantly, we show that the Y537S mutation is specifically associated with the over-expression of a number of protein markers of poor clinical outcome (COL6A3, ERBB2, STAT3, AFP, TFF1, CDK4 and CD44). In summary, we have uncovered a novel metabolic mechanism leading to endocrine resistance, which may have important clinical implications for improving patient outcomes.
Collapse
|
8
|
Hartmaier RJ, Trabucco SE, Priedigkeit N, Chung JH, Parachoniak CA, Vanden Borre P, Morley S, Rosenzweig M, Gay LM, Goldberg ME, Suh J, Ali SM, Ross J, Leyland-Jones B, Young B, Williams C, Park B, Tsai M, Haley B, Peguero J, Callahan RD, Sachelarie I, Cho J, Atkinson JM, Bahreini A, Nagle AM, Puhalla SL, Watters RJ, Erdogan-Yildirim Z, Cao L, Oesterreich S, Mathew A, Lucas PC, Davidson NE, Brufsky AM, Frampton GM, Stephens PJ, Chmielecki J, Lee AV. Recurrent hyperactive ESR1 fusion proteins in endocrine therapy-resistant breast cancer. Ann Oncol 2019; 29:872-880. [PMID: 29360925 PMCID: PMC5913625 DOI: 10.1093/annonc/mdy025] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background Estrogen receptor-positive (ER-positive) metastatic breast cancer is often intractable due to endocrine therapy resistance. Although ESR1 promoter switching events have been associated with endocrine-therapy resistance, recurrent ESR1 fusion proteins have yet to be identified in advanced breast cancer. Patients and methods To identify genomic structural rearrangements (REs) including gene fusions in acquired resistance, we undertook a multimodal sequencing effort in three breast cancer patient cohorts: (i) mate-pair and/or RNAseq in 6 patient-matched primary-metastatic tumors and 51 metastases, (ii) high coverage (>500×) comprehensive genomic profiling of 287-395 cancer-related genes across 9542 solid tumors (5216 from metastatic disease), and (iii) ultra-high coverage (>5000×) genomic profiling of 62 cancer-related genes in 254 ctDNA samples. In addition to traditional gene fusion detection methods (i.e. discordant reads, split reads), ESR1 REs were detected from targeted sequencing data by applying a novel algorithm (copyshift) that identifies major copy number shifts at rearrangement hotspots. Results We identify 88 ESR1 REs across 83 unique patients with direct confirmation of 9 ESR1 fusion proteins (including 2 via immunoblot). ESR1 REs are highly enriched in ER-positive, metastatic disease and co-occur with known ESR1 missense alterations, suggestive of polyclonal resistance. Importantly, all fusions result from a breakpoint in or near ESR1 intron 6 and therefore lack an intact ligand binding domain (LBD). In vitro characterization of three fusions reveals ligand-independence and hyperactivity dependent upon the 3' partner gene. Our lower-bound estimate of ESR1 fusions is at least 1% of metastatic solid breast cancers, the prevalence in ctDNA is at least 10× enriched. We postulate this enrichment may represent secondary resistance to more aggressive endocrine therapies applied to patients with ESR1 LBD missense alterations. Conclusions Collectively, these data indicate that N-terminal ESR1 fusions involving exons 6-7 are a recurrent driver of endocrine therapy resistance and are impervious to ER-targeted therapies.
Collapse
Affiliation(s)
- R J Hartmaier
- Foundation Medicine Inc., Cambridge; Department of Pharmacology and Chemical Biolog, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, USA; Women's Cancer Research Center, Magee-Women's Research Institute, Pittsburgh, USA.
| | | | - N Priedigkeit
- Department of Pharmacology and Chemical Biolog, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, USA; Women's Cancer Research Center, Magee-Women's Research Institute, Pittsburgh, USA
| | | | | | | | - S Morley
- Foundation Medicine Inc., Cambridge
| | | | - L M Gay
- Foundation Medicine Inc., Cambridge
| | | | - J Suh
- Foundation Medicine Inc., Cambridge
| | - S M Ali
- Foundation Medicine Inc., Cambridge
| | - J Ross
- Foundation Medicine Inc., Cambridge
| | - B Leyland-Jones
- Department of Molecular and Experimental Medicine, Avera Cancer Institute, Sioux Falls, USA
| | - B Young
- Department of Molecular and Experimental Medicine, Avera Cancer Institute, Sioux Falls, USA
| | - C Williams
- Department of Molecular and Experimental Medicine, Avera Cancer Institute, Sioux Falls, USA
| | - B Park
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, USA
| | - M Tsai
- Minnesota Oncology, Minneapolis, USA
| | - B Haley
- UT Southwestern Medical Center, Dallas, USA
| | - J Peguero
- Oncology Consultants Research Department, Houston, USA
| | | | | | - J Cho
- New Bern Cancer Care, New Bern, USA
| | - J M Atkinson
- Women's Cancer Research Center, Magee-Women's Research Institute, Pittsburgh, USA
| | - A Bahreini
- Women's Cancer Research Center, Magee-Women's Research Institute, Pittsburgh, USA; Department of Human Genetics, University of Pittsburgh, Pittsburgh, USA; Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - A M Nagle
- Department of Pharmacology and Chemical Biolog, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, USA; Women's Cancer Research Center, Magee-Women's Research Institute, Pittsburgh, USA
| | - S L Puhalla
- Women's Cancer Research Center, Magee-Women's Research Institute, Pittsburgh, USA; Foundation Medicine Inc., Cambridge; Department of Molecular and Experimental Medicine, Avera Cancer Institute, Sioux Falls, USA
| | - R J Watters
- Department of Pharmacology and Chemical Biolog, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, USA; Women's Cancer Research Center, Magee-Women's Research Institute, Pittsburgh, USA; Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, USA
| | - Z Erdogan-Yildirim
- Women's Cancer Research Center, Magee-Women's Research Institute, Pittsburgh, USA; Department of Human Genetics, University of Pittsburgh, Pittsburgh, USA
| | - L Cao
- Women's Cancer Research Center, Magee-Women's Research Institute, Pittsburgh, USA; Central South University Xiangya School of Medicine, China
| | - S Oesterreich
- Department of Pharmacology and Chemical Biolog, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, USA; Women's Cancer Research Center, Magee-Women's Research Institute, Pittsburgh, USA
| | - A Mathew
- Department of Medicine, University of Pittsburgh, Pittsburgh, USA
| | - P C Lucas
- Department of Pathology, University of Pittsburgh, Pittsburgh, USA
| | - N E Davidson
- Foundation Medicine Inc., Cambridge; Department of Molecular and Experimental Medicine, Avera Cancer Institute, Sioux Falls, USA
| | - A M Brufsky
- Foundation Medicine Inc., Cambridge; Department of Molecular and Experimental Medicine, Avera Cancer Institute, Sioux Falls, USA
| | | | | | | | - A V Lee
- Department of Pharmacology and Chemical Biolog, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, USA; Women's Cancer Research Center, Magee-Women's Research Institute, Pittsburgh, USA
| |
Collapse
|
9
|
Piscuoglio S, Ng CKY, Weigelt B, Chandarlapaty S, Reis-Filho JS. ESR1 and endocrine therapy resistance: more than just mutations. Ann Oncol 2019. [PMID: 29522117 DOI: 10.1093/annonc/mdy081] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- S Piscuoglio
- Institute of Pathology and Medical Genetics, University Hospital Basel, Basel, Switzerland
| | - C K Y Ng
- Institute of Pathology and Medical Genetics, University Hospital Basel, Basel, Switzerland; Department of Biomedicine, University of Basel, Basel, Switzerland
| | - B Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - S Chandarlapaty
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, USA
| | - J S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA.
| |
Collapse
|
10
|
Rani A, Stebbing J, Giamas G, Murphy J. Endocrine Resistance in Hormone Receptor Positive Breast Cancer-From Mechanism to Therapy. Front Endocrinol (Lausanne) 2019; 10:245. [PMID: 31178825 PMCID: PMC6543000 DOI: 10.3389/fendo.2019.00245] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 03/28/2019] [Indexed: 12/24/2022] Open
Abstract
The importance and role of the estrogen receptor (ER) pathway has been well-documented in both breast cancer (BC) development and progression. The treatment of choice in women with metastatic breast cancer (MBC) is classically divided into a variety of endocrine therapies, 3 of the most common being: selective estrogen receptor modulators (SERM), aromatase inhibitors (AI) and selective estrogen receptor down-regulators (SERD). In a proportion of patients, resistance develops to endocrine therapy due to a sophisticated and at times redundant interference, at the molecular level between the ER and growth factor. The progression to endocrine resistance is considered to be a gradual, step-wise process. Several mechanisms have been proposed but thus far none of them can be defined as the complete explanation behind the phenomenon of endocrine resistance. Although multiple cellular, molecular and immune mechanisms have been and are being extensively studied, their individual roles are often poorly understood. In this review, we summarize current progress in our understanding of ER biology and the molecular mechanisms that predispose and determine endocrine resistance in breast cancer patients.
Collapse
Affiliation(s)
- Aradhana Rani
- School of Life Sciences, University of Westminster, London, United Kingdom
- *Correspondence: Aradhana Rani
| | - Justin Stebbing
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Georgios Giamas
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | - John Murphy
- School of Life Sciences, University of Westminster, London, United Kingdom
| |
Collapse
|
11
|
Immunohistochemical analysis of estrogen receptor in breast cancer with ESR1 mutations detected by hybrid capture-based next-generation sequencing. Mod Pathol 2019; 32:81-87. [PMID: 30158597 PMCID: PMC6309269 DOI: 10.1038/s41379-018-0116-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 07/18/2018] [Accepted: 07/21/2018] [Indexed: 02/07/2023]
Abstract
Estrogen receptor-α (ER-α), encoded by ESR1, is detected by immunohistochemistry in approximately 70% of invasive breast cancers and serves as a strong predictive biomarker. ESR1-activating mutations in the ligand-binding domain have been reported in up to 35-40% of ER-positive metastatic breast cancers and are associated with endocrine therapy resistance and disease progression. At present, it is unclear whether ESR1 mutations alter the immunohistochemical detection of ER performed in routine clinical practice. In this study, ESR1 mutations in breast cancer were identified utilizing Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT), a Food and Drug Administration-approved hybridization capture-based next-generation sequencing assay. Five hundred and eighty-six breast cancers from patients with locally advanced or metastatic disease were analyzed using MSK-IMPACT in the study period. ESR1 somatic alterations were identified in 67 breast cancer samples from 66 patients. Immunohistochemical analysis of ER, progesterone receptor, and human epidermal growth factor receptor 2 was performed on the primary and treated breast cancers from these patients at the time of diagnosis. Twenty unique ESR1 mutations were identified involving the ligand-binding domain, all in breast cancer samples from patients previously treated with endocrine therapy. The most frequent mutations were D538G (n = 22), Y537S (n = 7), and E380Q (n = 7). All breast cancer samples with an ESR1 mutation were ER-positive by immunohistochemistry. Review of the ER immunohistochemistry in the paired untreated primary tumor and treated tumor from 34 patients showed no detectable change in the ER-positive immunohistochemical status (median percentage of invasive tumor cells with nuclear staining: untreated primary tumor 90%, treated tumor 95%). We conclude that ESR1 mutations do not appreciably diminish ER-positive staining by immunohistochemistry. In addition to standard biomarker testing by immunohistochemistry, the assessment of ESR1 mutations by molecular testing can help guide the clinical management of patients with ER-positive breast cancer in the setting of endocrine resistance and progression of disease.
Collapse
|
12
|
Timeless Is a Novel Estrogen Receptor Co-activator Involved in Multiple Signaling Pathways in MCF-7 Cells. J Mol Biol 2018; 430:1531-1543. [DOI: 10.1016/j.jmb.2018.03.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 03/06/2018] [Accepted: 03/06/2018] [Indexed: 01/08/2023]
|
13
|
Khajah MA, Mathew PM, Luqmani YA. Na+/K+ ATPase activity promotes invasion of endocrine resistant breast cancer cells. PLoS One 2018; 13:e0193779. [PMID: 29590154 PMCID: PMC5874017 DOI: 10.1371/journal.pone.0193779] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 02/17/2018] [Indexed: 01/27/2023] Open
Abstract
Background The Na+/K+-ATPase (NKP) is an important ion transporter also involved in signal transduction. Its expression profile is altered in various tumours including that of the breast. We studied the effect of inhibiting NKP activity in non-tumorigenic breast cell line and in estrogen receptor positive and negative breast cancer cells. Methods Expression and localization of NKP and downstream signaling molecules were determined by RT-PCR, western blotting and immunofluorescence. Cell proliferation, apoptosis and cell cycle stage were determined using MTT, annexin V and flow cytometry. Cell motility and invasion were determined using wound healing and matrigel assays. Total matrix metalloproteinase (MMP) was determined by a fluorescence-based assay. Results NKP was mainly localized on the cell membrane. Its baseline expression and activity were enhanced in breast cancer compared to the non-tumorigenic breast cell line. Ouabain and 3,4,5,6-tetrahydroxyxanthone (TTX) treatment significantly inhibited NKP activity, which significantly reduced cell proliferation, motility, invasion and pH-induced membrane blebbing. EGF stimulation induced internalization of NKP from the cell membrane to the cytoplasm. Ouabain inhibited EGF-induced phosphorylation of Rac/cdc42, profillin, ERK1/2 and P70S6K. Conclusions The NKP may offer a novel therapeutic target in breast cancer patients who have developed metastasis, aiming to improve therapeutic outcomes and enhance survival rate.
Collapse
|
14
|
Tsuboi K, Nagatomo T, Gohno T, Higuchi T, Sasaki S, Fujiki N, Kurosumi M, Takei H, Yamaguchi Y, Niwa T, Hayashi SI. Single CpG site methylation controls estrogen receptor gene transcription and correlates with hormone therapy resistance. J Steroid Biochem Mol Biol 2017; 171:209-217. [PMID: 28412323 DOI: 10.1016/j.jsbmb.2017.04.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Revised: 03/14/2017] [Accepted: 04/06/2017] [Indexed: 12/19/2022]
Abstract
Hormone therapy is the most effective treatment for patients with estrogen receptor α-positive breast cancers. However, although resistance occurs during treatment in some cases and often reflects changed estrogen receptor α status, the relationship between changes in estrogen receptor α expression and resistance to therapy are poorly understood. In this study, we identified a mechanism for altered estrogen receptor α expression during disease progression and acquired hormone therapy resistance in aromatase inhibitor-resistant breast cancer cell lines. Subsequently, we investigated promoter switching and DNA methylation status of the estrogen receptor α promoter, and found marked changes of methylation at a single CpG site (CpG4) in resistant cells. In addition, luciferase reporter assays showed reduced transcriptional activity from this methylated CpG site. This CpG region was also completely conserved among species, suggesting that it acts as a methylation-sensitive Ets-2 transcription factor binding site, as confirmed using chromatin immunoprecipitation assays. In estrogen receptor α-positive tumors, CpG4 methylation levels were inversely correlated with estrogen receptor α expression status, suggesting that single CpG site plays an important role in the regulation of estrogen receptor α transcription.
Collapse
Affiliation(s)
- Kouki Tsuboi
- Department of Molecular and Functional Dynamics, Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, 980-8575, Japan
| | - Takamasa Nagatomo
- Department of Molecular and Functional Dynamics, Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, 980-8575, Japan
| | - Tatsuyuki Gohno
- Department of Molecular and Functional Dynamics, Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, 980-8575, Japan
| | - Toru Higuchi
- Department of Molecular and Functional Dynamics, Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, 980-8575, Japan
| | - Shunta Sasaki
- Department of Molecular and Functional Dynamics, Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, 980-8575, Japan
| | - Natsu Fujiki
- Department of Molecular and Functional Dynamics, Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, 980-8575, Japan
| | - Masafumi Kurosumi
- Department of Pathology, Saitama Cancer Center, Ina-machi, Saitama, 362-0806, Japan
| | - Hiroyuki Takei
- Division of Breast Surgery, Saitama Cancer Center, Ina-machi, Saitama, 362-0806, Japan
| | - Yuri Yamaguchi
- Resarch Institute for Clinical Oncology, Saitama Cancer Center, Ina-machi, Saitama, 362-0806, Japan
| | - Toshifumi Niwa
- Department of Molecular and Functional Dynamics, Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, 980-8575, Japan
| | - Shin-Ichi Hayashi
- Department of Molecular and Functional Dynamics, Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, 980-8575, Japan; Center for Regulatory Epigenome and Diseases, Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, 980-8575, Japan.
| |
Collapse
|
15
|
Bettaieb A, Paul C, Plenchette S, Shan J, Chouchane L, Ghiringhelli F. Precision medicine in breast cancer: reality or utopia? J Transl Med 2017. [PMID: 28623955 PMCID: PMC5474301 DOI: 10.1186/s12967-017-1239-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Many cancers, including breast cancer, have demonstrated prognosis and support advantages thanks to the discovery of targeted therapies. The advent of these new approaches marked the rise of precision medicine, which leads to improve the diagnosis, prognosis and treatment of cancer. Precision medicine takes into account the molecular and biological specificities of the patient and their tumors that will influence the treatment determined by physicians. This new era of medicine is accessible through molecular genetics platforms, the development of high-speed sequencers and means of analysis of these data. Despite the spectacular results in the treatment of cancers including breast cancer, described in this review, not all patients however can benefit from this new strategy. This seems to be related to the many genetic mutations, which may be different from one patient to another or within the same patient. It comes to give new impetus to the research—both from a technological and biological point of view—to make the hope of precision medicine accessible to all.
Collapse
Affiliation(s)
- Ali Bettaieb
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000, Paris, France. .,LIIC, EA7269, Université de Bourgogne Franche Comté, 21000, Dijon, France. .,Immunology and Immunotherapy of Cancer Laboratory, EA7269, Université de Bourgogne, EPHE 7 Bd Jeanne d'Arc, 21079, Dijon, France.
| | - Catherine Paul
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000, Paris, France.,LIIC, EA7269, Université de Bourgogne Franche Comté, 21000, Dijon, France
| | - Stéphanie Plenchette
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, 75000, Paris, France.,LIIC, EA7269, Université de Bourgogne Franche Comté, 21000, Dijon, France
| | - Jingxuan Shan
- Laboratory of Genetic Medicine and Immunology, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - Lotfi Chouchane
- Laboratory of Genetic Medicine and Immunology, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, Qatar
| | - François Ghiringhelli
- Département d'Oncologie Médicale, Centre Georges-François-Leclerc, 21000, Dijon, France.,Plateforme de Transfert en Biologie Cancérologique, Centre Georges-François-Leclerc, 21000, Dijon, France.,UMR 1231 Inserm-Université de Bourgogne Franche Comté, UFR des Sciences de Santé, 21000, Dijon, France.,Université de Bourgogne, 21000, Dijon, France
| |
Collapse
|
16
|
Khajah MA, Mathew PM, Luqmani YA. Inhibitors of PI3K/ERK1/2/p38 MAPK Show Preferential Activity Against Endocrine-Resistant Breast Cancer Cells. Oncol Res 2017; 25:1283-1295. [PMID: 28276317 PMCID: PMC7841054 DOI: 10.3727/096504017x14883245308282] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Current mainstream pharmacological options for the treatment of endocrine-resistant breast cancer have limitations in terms of their side effect profile and lack of discrimination between normal and cancer cells. In the current study, we assessed the responses of normal breast epithelial cells MCF10A, estrogen receptor-positive (ER+) MCF-7, and ER-silenced pII breast cancer cells to inhibitors (either individually or in combination) of downstream signaling molecules. The expression/activity of ERK1/2, p38 MAPK, and Akt was determined by Western blotting. Cell proliferation, motility, and invasion were determined using MTT, wound healing, and Matrigel assays, respectively. Morphological changes in response to variation in external pH were assessed by light microscopy. Our results demonstrated that the inhibitors of ERK1/2 (PD0325901), p38 MAPK (SB203580), and PI3K (LY294002) preferentially reduce breast cancer cell proliferation. In pII cells, they also reduced motility, invasion, and bleb formation induced by alkaline conditions. Combination treatment with lower concentrations of inhibitors was significantly more effective than single agents and was more effective against the cancer cell lines than the normal MCF10A. In contrast, the commonly used cytotoxic agent paclitaxel did not sufficiently discriminate between the MCF10A and the cancer cells. We concluded that combination therapy using ERK1/2 inhibitor and either p38 MAPK or PI3K inhibitor may provide a greater therapeutic benefit in treating breast cancer by specifically targeting cancer cells with lower doses of each drug than needed individually, potentially reducing unwanted side effects.
Collapse
|
17
|
Fanning SW, Mayne CG, Dharmarajan V, Carlson KE, Martin TA, Novick SJ, Toy W, Green B, Panchamukhi S, Katzenellenbogen BS, Tajkhorshid E, Griffin PR, Shen Y, Chandarlapaty S, Katzenellenbogen JA, Greene GL. Estrogen receptor alpha somatic mutations Y537S and D538G confer breast cancer endocrine resistance by stabilizing the activating function-2 binding conformation. eLife 2016; 5:12792. [PMID: 26836308 PMCID: PMC4821807 DOI: 10.7554/elife.12792] [Citation(s) in RCA: 216] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 01/31/2016] [Indexed: 12/15/2022] Open
Abstract
Somatic mutations in the estrogen receptor alpha (ERα) gene (ESR1), especially Y537S and D538G, have been linked to acquired resistance to endocrine therapies. Cell-based studies demonstrated that these mutants confer ERα constitutive activity and antiestrogen resistance and suggest that ligand-binding domain dysfunction leads to endocrine therapy resistance. Here, we integrate biophysical and structural biology data to reveal how these mutations lead to a constitutively active and antiestrogen-resistant ERα. We show that these mutant ERs recruit coactivator in the absence of hormone while their affinities for estrogen agonist (estradiol) and antagonist (4-hydroxytamoxifen) are reduced. Further, they confer antiestrogen resistance by altering the conformational dynamics of the loop connecting Helix 11 and Helix 12 in the ligand-binding domain of ERα, which leads to a stabilized agonist state and an altered antagonist state that resists inhibition. Around one in every eight women will be diagnosed with breast cancer in their lifetime. Hormone-based therapies – also referred to antiestrogen drugs – target a protein called estrogen receptor alpha and are effective treatments for the majority of these cancers. Unfortunately, about half of patients will develop recurrent breast cancers even though the cancer continues to produce the target of the drugs. The estrogen receptor alpha drives breast cancer in a number of ways, many of which require the receptor to be activated by binding to the hormone estrogen. When estrogen binds it causes the receptor to change shape to expose a surface where other proteins called coactivators can bind. Once a coactivator is bound, the estrogen receptor is active and signals the cancer cell to grow, divide, invade local tissues, and spread to new sites in the body. Antiestrogen drugs competitively block the binding of estrogen to the receptor and cause the receptor to take on a different shape that inhibits the binding of the coactivator. However, recent studies identified mutations at specific sites in the gene that encodes estrogen receptor alpha in a large subset of patients with breast cancers that have spread. These mutations make the receptor resistant to antiestrogen drugs, and two mutations (called Y537S and D538G) account for approximately 70% of cases. However, it was not clear how these mutations altered the activity of estrogen receptor alpha at the molecular level. Fanning, Mayne, Dharmarajan et al. now show these two most common mutations allow estrogen receptor alpha to bind to the coactivator in the absence of hormone. This unfortunately also reduces the effectiveness of one of the mostly widely administered antiestrogen therapies – a drug called tamoxifen. However, Fanning, Mayne, Dharmarajan et al. also show that the newer and more potent antiestrogens that are currently under examination in clinical trials should be highly effective at treating the cancers with the mutated versions of estrogen receptor alpha. Applying the knowledge gained from these new findings toward the development of new antiestrogens could help reverse the impact of these common mutations. If successful, these new drugs will provide life-saving treatments for many breast cancer patients.
Collapse
Affiliation(s)
- Sean W Fanning
- Ben May Department for Cancer Research, University of Chicago, Chicago, United States
| | - Christopher G Mayne
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, United States.,Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, United States.,Department of Biochemistry, Center for Biophysics and Computational Biology, University of Illinois at Urbana-Champaign, Urbana, United States
| | | | - Kathryn E Carlson
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, United States
| | - Teresa A Martin
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, United States
| | - Scott J Novick
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, United States
| | - Weiyi Toy
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, United States
| | - Bradley Green
- Ben May Department for Cancer Research, University of Chicago, Chicago, United States
| | - Srinivas Panchamukhi
- Ben May Department for Cancer Research, University of Chicago, Chicago, United States
| | - Benita S Katzenellenbogen
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, United States
| | - Emad Tajkhorshid
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, United States.,Department of Biochemistry, Center for Biophysics and Computational Biology, University of Illinois at Urbana-Champaign, Urbana, United States
| | - Patrick R Griffin
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, United States
| | - Yang Shen
- Department of Electrical and Computer Engineering, TEES-AgriLife Center for Bioinformatics and Genomic Systems Engineering, Texas A&M University, College Station, United States
| | - Sarat Chandarlapaty
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, United States
| | | | - Geoffrey L Greene
- Ben May Department for Cancer Research, University of Chicago, Chicago, United States
| |
Collapse
|
18
|
Endocrine therapy resistance in breast cancer: current status, possible mechanisms and overcoming strategies. Future Med Chem 2015; 7:1511-9. [PMID: 26306654 DOI: 10.4155/fmc.15.93] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Endocrine therapy has become one of most effective forms of targeted adjuvant therapy for hormone-sensitive breast cancer and may be given after surgery or radiotherapy, and also prior, or subsequent to chemotherapy. Current commonly used drugs for adjuvant endocrine therapy can be divided into following three classes: selective estrogen receptor modulators, aromatase inhibitors and selective estrogen receptor downregulators. Tumor cells can develop resistance to endocrine therapy, a major obstacle limiting the success of breast cancer treatment. The complicated crosstalk, both genomic and nongenomic, between estrogen receptors and growth factors was considered to be a crucial factor contributing to endocrine resistance. However, resistance to this therapy is thought to be a progressive, step-wise process, and the underlying mechanism remains unclear. In this review, we summarize the possible biological and molecular mechanisms that underlie endocrine resistance, and discuss some novel strategies to overcoming these issues.
Collapse
|
19
|
Wang T, Liu JH, Zhang J, Wang L, Chen C, Dai PG. A multiplex allele-specific real-time PCR assay for screening of ESR1 mutations in metastatic breast cancer. Exp Mol Pathol 2015; 98:152-7. [PMID: 25746663 DOI: 10.1016/j.yexmp.2015.03.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2014] [Revised: 11/10/2014] [Accepted: 03/02/2015] [Indexed: 11/26/2022]
Abstract
BACKGROUND Acquired resistance to endocrine-based therapies occurs in virtually all estrogen receptor-α (ERα, encoded by ESR1) positive breast cancer patients. The underlying molecular mechanism is attributed to the activating mutations in ESR1. These mutations provide an exciting opportunity for the development of new antagonists that specifically inhibit the mutant proteins. Therefore, accurate detection of ESR1 mutations is of critical importance in clinical practice. MATERIALS AND METHODS We carried out a single tube, multiplex allele-specific real-time PCR assay for the detection of four ESR1 mutations (Y537S, Y537C, Y537N, and D538G). RESULTS The assay was found to be highly specific and sensitive. With this assay, as low as 1% mutant DNA template in wild type DNA could be detected. Fifteen DNA samples were prepared from archived formalin-fixed paraffin-embedded metastatic breast cancer biopsies. They were further screened with this assay, and three samples were identified as ESR1 mutant. The results were validated with pyrosequencing and complete concordance was observed between the two assays. CONCLUSION The multiplex allele-specific real-time PCR assay provides a rapid and reliable diagnostic tool for accurate detection of ESR1 mutations. This procedure may be used in the clinical treatment of breast cancer.
Collapse
Affiliation(s)
- Ting Wang
- The National Engineering Research Center for Miniaturized Detection Systems, School of Life Sciences, Northwest University, Xi'an, PR China
| | - Jin-Hui Liu
- The National Engineering Research Center for Miniaturized Detection Systems, School of Life Sciences, Northwest University, Xi'an, PR China
| | - Jie Zhang
- The National Engineering Research Center for Miniaturized Detection Systems, School of Life Sciences, Northwest University, Xi'an, PR China
| | - Le Wang
- The National Engineering Research Center for Miniaturized Detection Systems, School of Life Sciences, Northwest University, Xi'an, PR China; The Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, PR China
| | - Chao Chen
- The National Engineering Research Center for Miniaturized Detection Systems, School of Life Sciences, Northwest University, Xi'an, PR China
| | - Peng-Gao Dai
- The National Engineering Research Center for Miniaturized Detection Systems, School of Life Sciences, Northwest University, Xi'an, PR China.
| |
Collapse
|
20
|
Hayashi SI, Kimura M. Mechanisms of hormonal therapy resistance in breast cancer. Int J Clin Oncol 2015; 20:262-7. [PMID: 25652907 DOI: 10.1007/s10147-015-0788-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 01/19/2015] [Indexed: 02/07/2023]
Abstract
Whilst estrogen receptor (ER)-positive breast cancers are preferentially treated with hormone therapy, approximately one-third of them relapse. The mechanisms of refractoriness have been investigated by numerous studies but have not been fully clarified. Hormonal therapy resistance, particularly aromatase inhibitor (AI) resistance, may be related to the acquisition of alternative intracellular ER signaling. We have been investing the mechanisms using cancer specimens and cell lines by monitoring the transcription activity of ERs. AI refractory specimens showed diverse ER activity in the adenovirus estrogen receptor element-green fluorescent protein (ERE-GFP) assay and varied sensitivity to anti-estrogens, indicating the existence of multiple resistant mechanisms. We established six different types of cell lines mimicking AI resistance from ERE-GFP-introduced ER-positive cell lines. They revealed that multiple and alternative ER activating pathways were involved in the resistance, such as phosphorylation-dependent or androgen metabolite-dependent mechanisms. The response to fulvestrant and mammalian target of rapamycin inhibitor also varied among individual resistant cell lines. These results indicate that further subclassification of ER-positive breast cancer is extremely important to decide the therapeutic management of not only hormonal therapy but also new molecular target therapy.
Collapse
Affiliation(s)
- Shin-ichi Hayashi
- Department of Molecular and Functional Dynamics, and Center for Regulatory Epigenomics and Diseases, Tohoku University Graduate School of Medicine, Aoba-ku, Sendai, Japan,
| | | |
Collapse
|
21
|
Fujiki N, Konno H, Kaneko Y, Gohno T, Hanamura T, Imami K, Ishihama Y, Nakanishi K, Niwa T, Seino Y, Yamaguchi Y, Hayashi SI. Estrogen response element-GFP (ERE-GFP) introduced MCF-7 cells demonstrated the coexistence of multiple estrogen-deprivation resistant mechanisms. J Steroid Biochem Mol Biol 2014; 139:61-72. [PMID: 24128438 DOI: 10.1016/j.jsbmb.2013.08.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 08/17/2013] [Accepted: 08/20/2013] [Indexed: 10/26/2022]
Abstract
The acquisition of estrogen-deprivation resistance and estrogen receptor (ER) signal-independence in ER-positive breast cancer is one of the crucial steps in advancing the aggressiveness of breast cancer; however, this has not yet been elucidated in detail. To address this issue, we established several estrogen-deprivation-resistant (EDR) breast cancer cell lines from our unique MCF-7 cells, which had been stably transfected with an ERE-GFP reporter plasmid. Three cell lines with high ER activity and another 3 cell lines with no ER activity were established from cell cloning by monitoring GFP expression in living cells. The former three ERE-GFP-positive EDR cell lines showed the overexpression of ER and high expression of several ER-target genes. Further analysis of intracellular signaling factors revealed a marked change in the phosphorylation status of ERα on Ser167 and Akt on Thr308 by similar mechanisms reported previously; however, we could not find any changes in MAP-kinase factors. Comprehensive phospho-proteomic analysis also indicated the possible contribution of the Akt pathway to the phosphorylation of ERα. On the other hand, constitutive activation of c-Jun N-terminal kinase (JNK) was observed in ERE-GFP-negative EDR cells, and the growth of these cells was inhibited by a JNK inhibitor. An IGF1R-specific inhibitor diminished the phosphorylation of JNK, which suggested that a novel signaling pathway, IGF1R-JNK, may be important for the proliferation of ER-independent MCF-7 cells. These results indicate that ER-positive breast cancer cells can acquire resistance by more than two mechanisms at a time, which suggests that multiple mechanisms may occur simultaneously. This finding also implies that breast cancers with different resistance mechanisms can concomitantly occur and mingle in an individual patient, and may be a cause of the recurrence of cancer.
Collapse
Affiliation(s)
- Natsu Fujiki
- Department of Molecular and Functional Dynamics, Tohoku University Graduate School of Medicine, Aoba-ku, Sendai 980-8575, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Anti-estrogens and selective estrogen-receptor modulators. Mol Oncol 2013. [DOI: 10.1017/cbo9781139046947.084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
23
|
ESR1 ligand-binding domain mutations in hormone-resistant breast cancer. Nat Genet 2013; 45:1439-45. [PMID: 24185512 PMCID: PMC3903423 DOI: 10.1038/ng.2822] [Citation(s) in RCA: 924] [Impact Index Per Article: 77.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 10/10/2013] [Indexed: 02/07/2023]
Abstract
Seventy percent of breast cancers express estrogen receptor (ER), and most of these are sensitive to ER inhibition. However, many such tumors for unknown reasons become refractory to inhibition of estrogen action in the metastatic setting. We conducted a comprehensive genetic analysis of two independent cohorts of metastatic ER-positive breast tumors and identified mutations in ESR1 affecting the ligand-binding domain (LBD) in 14 of 80 cases. These included highly recurrent mutations encoding p.Tyr537Ser, p.Tyr537Asn and p.Asp538Gly alterations. Molecular dynamics simulations suggest that the structures of the Tyr537Ser and Asp538Gly mutants involve hydrogen bonding of the mutant amino acids with Asp351, thus favoring the agonist conformation of the receptor. Consistent with this model, mutant receptors drive ER-dependent transcription and proliferation in the absence of hormone and reduce the efficacy of ER antagonists. These data implicate LBD-mutant forms of ER in mediating clinical resistance to hormonal therapy and suggest that more potent ER antagonists may be of substantial therapeutic benefit.
Collapse
|
24
|
Fu X, Osborne CK, Schiff R. Biology and therapeutic potential of PI3K signaling in ER+/HER2-negative breast cancer. Breast 2013; 22 Suppl 2:S12-8. [PMID: 24011769 DOI: 10.1016/j.breast.2013.08.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
PI3K is a central node mediating growth factor receptor signaling. With its downstream effectors such as AKT and mTOR, and its crosstalk with the RAS/RAF/MEK/MAPK pathway, it plays a vital role in cancer cell proliferation, metabolism, and survival. Recent breast cancer (BC) molecular portraits delineate PI3K as the most frequently altered pathway, with recurrent PIK3CA mutations mostly found in the luminal subtypes of BC. The transcriptomic and proteomic signatures of PI3K pathway activation associate with reduced estrogen receptor α (ER) levels and activity, and with the luminal B subtype of BC that has a relatively poor outcome. However, oncogenic transforming PIK3CA mutations have been shown to predict a better outcome in ER+/HER2-negative BC treated with endocrine therapy. In this review, we summarize the recent findings in the cause-and-effect of PI3K pathway aberration and endocrine sensitivity, especially the crosstalk with the ER pathway. Potential therapeutic approaches based on these findings are also discussed.
Collapse
Affiliation(s)
- Xiaoyong Fu
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA; Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | |
Collapse
|
25
|
Ogas T, Kondratyuk TP, Pezzuto JM. Resveratrol analogs: promising chemopreventive agents. Ann N Y Acad Sci 2013; 1290:21-9. [DOI: 10.1111/nyas.12196] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Talysa Ogas
- Daniel K. Inouye College of Pharmacy; University of Hawaii at Hilo; Hilo Hawaii
| | | | - John M. Pezzuto
- Daniel K. Inouye College of Pharmacy; University of Hawaii at Hilo; Hilo Hawaii
| |
Collapse
|
26
|
Adjuvant Hormonal Therapy in Postmenopausal Women with Breast Cancer: Physician's Choices. Int J Breast Cancer 2013; 2012:849592. [PMID: 23304528 PMCID: PMC3523410 DOI: 10.1155/2012/849592] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2012] [Accepted: 11/20/2012] [Indexed: 11/17/2022] Open
Abstract
The choice of adjuvant hormonal therapy in postmenopausal women with hormone receptor positive breast cancer has remained a matter of controversy and debate. The variety of agents is available, with each claiming to be superior. This clinical survey was undertaken to get an impression of the physician's first choice of therapy in an attempt to find out what questions still need to be answered in the making of “standard of care.” A web-based clinical survey was sent to the cancer physicians around the world, and 182 physicians responded to the survey. Most were medical oncologists in a tertiary care hospital. 36.3% preferred Anastrozole, 35.2% Tamoxifen, and 22.2% Letrozole as their first choice. Data support (67.8%) and safety concerns (30%) were given as the main reasons for the choice, 63.7% switched their therapy, and 24% had to switch because of side effects. 73.6% used 5 years of adjuvant hormonal therapy, 6.6% for 7 years, and 4.4% for 10 years. 61.5% follow their patients 3 times monthly, and 73.2% used laboratory and radiological assessment at each followup. Conclusion. Physicians show disagreement over the choice and duration of hormonal therapy in this patient population. Clinical trials leading to firm recommendations to set standards from which patients benefit the most are needed.
Collapse
|
27
|
Mechanisms of resistance to endocrine therapy in breast cancer: focus on signaling pathways, miRNAs and genetically based resistance. Int J Mol Sci 2012; 14:108-45. [PMID: 23344024 PMCID: PMC3565254 DOI: 10.3390/ijms14010108] [Citation(s) in RCA: 192] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 12/10/2012] [Accepted: 12/12/2012] [Indexed: 12/14/2022] Open
Abstract
Breast cancer is the most frequent malignancy diagnosed in women. Approximately 70% of breast tumors express the estrogen receptor (ER). Tamoxifen and aromatase inhibitors (AIs) are the most common and effective therapies for patients with ERα-positive breast cancer. Alone or combined with chemotherapy, tamoxifen significantly reduces disease progression and is associated with more favorable impact on survival in patients. Unfortunately, endocrine resistance occurs, either de novo or acquired during the course of the treatment. The mechanisms that contribute to hormonal resistance include loss or modification in the ERα expression, regulation of signal transduction pathways, altered expression of specific microRNAs, balance of co-regulatory proteins, and genetic polymorphisms involved in tamoxifen metabolic activity. Because of the clinical consequences of endocrine resistance, new treatment strategies are arising to make the cells sensitive to tamoxifen. Here, we will review the current knowledge on mechanisms of endocrine resistance in breast cancer cells. In addition, we will discuss novel therapeutic strategies to overcome such resistance. Undoubtedly, circumventing endocrine resistance should help to improve therapy for the benefit of breast cancer patients.
Collapse
|
28
|
Cirillo F, Nassa G, Tarallo R, Stellato C, De Filippo MR, Ambrosino C, Baumann M, Nyman TA, Weisz A. Molecular mechanisms of selective estrogen receptor modulator activity in human breast cancer cells: identification of novel nuclear cofactors of antiestrogen-ERα complexes by interaction proteomics. J Proteome Res 2012; 12:421-31. [PMID: 23170835 DOI: 10.1021/pr300753u] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Estrogen receptor alpha (ERα) is a ligand-activated transcription factor that controls key cellular pathways via protein-protein interactions involving multiple components of transcriptional coregulator and signal transduction complexes. Natural and synthetic ERα ligands are classified as agonists (17β-estradiol/E(2)), selective estrogen receptor modulators (SERMs: Tamoxifen/Tam and Raloxifene/Ral), and pure antagonists (ICI 182,780-Fulvestrant/ICI), according to the response they elicit in hormone-responsive cells. Crystallographic analyses reveal ligand-dependent ERα conformations, characterized by specific surface docking sites for functional protein-protein interactions, whose identification is needed to understand antiestrogen effects on estrogen target tissues, in particular breast cancer (BC). Tandem affinity purification (TAP) coupled to mass spectrometry was applied here to map nuclear ERα interactomes dependent upon different classes of ligands in hormone-responsive BC cells. Comparative analyses of agonist (E(2))- vs antagonist (Tam, Ral or ICI)-bound ERα interacting proteins reveal significant differences among ER ligands that relate with their biological activity, identifying novel functional partners of antiestrogen-ERα complexes in human BC cell nuclei. In particular, the E(2)-dependent nuclear ERα interactome is different and more complex than those elicited by Tam, Ral, or ICI, which, in turn, are significantly divergent from each other, a result that provides clues to explain the pharmacological specificities of these compounds.
Collapse
Affiliation(s)
- Francesca Cirillo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine and Surgery, University of Salerno, Baronissi, Salerno, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Khajah MA, Al Saleh S, Mathew PM, Luqmani YA. Differential effect of growth factors on invasion and proliferation of endocrine resistant breast cancer cells. PLoS One 2012; 7:e41847. [PMID: 22860018 PMCID: PMC3408495 DOI: 10.1371/journal.pone.0041847] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Accepted: 06/29/2012] [Indexed: 01/27/2023] Open
Abstract
We have established several breast cancer cell lines that exhibit a permanent ER-depleted phenotype, induced by shRNA transfection of MCF-7 cells, which afford a useful model for studying acquired endocrine resistance. Previously we showed that MDA-231 as well as ER-silenced cells could invade through simulated extracellular matrix components. However, the contribution of individual serum components responsible for cell invasion was not determined. In the present study, an under-agarose gel assay was used to quantitatively assess the invasive movement of two ER-silenced cell lines (pII and YS2.5) in comparison to the parental MCF-7, the ER negative MDA-231, and normal HBL100 cells, as well as a line that was ER-shRNA transfected but failed to exhibit ER down-regulation (YS1.2). We also examined the effect of the growth factors EGF, IGF-1, TGFβ, PDGFC and RANTES on pII cell invasion and proliferation. All breast cancer cell lines which had reduced ER expression exhibited a serum-dependent invasive ability related to the degree of induced ER loss. TGFβ treatment inhibited pII cell proliferation and enhanced their invasive ability but at a relatively high dose. IGF-1 and EGF enhanced pII cell proliferation, with the latter playing the major role in promoting cell invasion. PDGFC did not affect either process although it is highly expressed in pII cells. Differential effects were observed on activation of Akt and ERK1/2 suggesting their involvement as intracellular mediators of EGF induced invasion, in part through the regulation of matrix metalloproteinase activity. Targeting EGF receptor tyrosine kinase activity by erlotinib resulted in significant inhibition of both pII cell proliferation and directional invasion towards EGF suggesting that this drug has potential therapeutic usefulness for preventing spread of particularly endocrine resistant breast cancer.
Collapse
|
30
|
Optimization of the aromatase inhibitory activities of pyridylthiazole analogues of resveratrol. Bioorg Med Chem 2012; 20:2427-34. [DOI: 10.1016/j.bmc.2012.01.047] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2011] [Revised: 01/18/2012] [Accepted: 01/26/2012] [Indexed: 01/03/2023]
|
31
|
Hemachandra LP, Madhubhani P, Chandrasena R, Esala P, Chen SN, Main M, Lankin DC, Scism RA, Dietz BM, Pauli GF, Thatcher GRJ, Bolton JL. Hops (Humulus lupulus) inhibits oxidative estrogen metabolism and estrogen-induced malignant transformation in human mammary epithelial cells (MCF-10A). Cancer Prev Res (Phila) 2011; 5:73-81. [PMID: 21997247 DOI: 10.1158/1940-6207.capr-11-0348] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Long-term exposure to estrogens including those in traditional hormone replacement therapy (HRT) increases the risk of developing hormone-dependent cancers. As a result, women are turning to over-the-counter (OTC) botanical dietary supplements, such as black cohosh (Cimicifuga racemosa) and hops (Humulus lupulus), as natural alternatives to HRT. The two major mechanisms which likely contribute to estrogen and/or HRT cancer risk are: the estrogen receptor-mediated hormonal pathway; and the chemical carcinogenesis pathway involving formation of estrogen quinones that damage DNA and proteins, hence initiating and promoting carcinogenesis. Because, OTC botanical HRT alternatives are in widespread use, they may have the potential for chemopreventive effects on estrogen carcinogenic pathways in vivo. Therefore, the effect of OTC botanicals on estrogen-induced malignant transformation of MCF-10A cells was studied. Cytochrome P450 catalyzed hydroxylation of estradiol at the 4-position leads to an o-quinone believed to act as the proximal carcinogen. Liquid chromatography/tandem mass spectrometry analysis of estradiol metabolites showed that 4-hydroxylation was inhibited by hops, whereas black cohosh was without effect. Estrogen-induced expression of CYP450 1B1 and CYP450 1A1 was attenuated by the hops extract. Two phenolic constituents of hops (xanthohumol, XH; 8-prenylnaringenin, 8-PN) were tested: 8-PN was a potent inhibitor, whereas XH had no effect. Finally, estrogen-induced malignant transformation of MCF-10A cells was observed to be significantly inhibited by hops (5 μg/mL) and 8-PN (50 nmol/L). These data suggest that hops extracts possess cancer chemopreventive activity through attenuation of estrogen metabolism mediated by 8-PN.
Collapse
Affiliation(s)
- L P Hemachandra
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois at Chicago, 833 S. Wood Street, Chicago, IL 60612-7231, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Mayhoub AS, Marler L, Kondratyuk TP, Park EJ, Pezzuto JM, Cushman M. Optimizing thiadiazole analogues of resveratrol versus three chemopreventive targets. Bioorg Med Chem 2011; 20:510-20. [PMID: 22115839 DOI: 10.1016/j.bmc.2011.09.031] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2011] [Revised: 09/13/2011] [Accepted: 09/19/2011] [Indexed: 12/20/2022]
Abstract
Chemoprevention is an approach to decrease cancer morbidity and mortality through inhibition of carcinogenesis and prevention of disease progression. Although the trans stilbene derivative resveratrol has chemopreventive properties, its action is compromised by weak non-specific effects on many biological targets. Replacement of the stilbene ethylenic bridge of resveratrol with a 1,2,4-thiadiazole heterocycle and modification of the substituents on the two aromatic rings afforded potential chemopreventive agents with enhanced potencies and selectivities when evaluated as inhibitors of aromatase and NF-κB and inducers of quinone reductase 1 (QR1).
Collapse
Affiliation(s)
- Abdelrahman S Mayhoub
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy and the Purdue Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | | | | | | | | | | |
Collapse
|
33
|
Buzdar AU. Role of biologic therapy and chemotherapy in hormone receptor- and HER2-positive breast cancer. Ann Oncol 2009; 20:993-9. [PMID: 19150946 DOI: 10.1093/annonc/mdn739] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND To review the efficacy of chemotherapy and human epidermal growth factor receptor 2 (HER2)-targeted therapy when used in addition to hormonal therapy for the optimal management of estrogen receptor-positive (ER+)/human epidermal growth factor receptor 2-positive (HER2+) breast cancer. DESIGN Literature published from January 2003 to March 2008 was reviewed to assess the use of chemotherapy and biologic therapy in addition to hormonal agents. RESULTS Aromatase inhibitors (AIs) demonstrated greater effectiveness in the adjuvant setting than tamoxifen for the management of ER+ and HER2+ breast cancer. Evidence of cross talk between HER2- and ER-signaling pathways suggests that combined treatment with HER2 blockade and hormonal therapy may offer clinical advantages beyond those provided by hormonal therapy alone in ER+/HER2+ disease. Combined therapy with trastuzumab plus an aromatase AI significantly improves progression-free survival, response rates, and clinical benefits when compared with AI monotherapy in postmenopausal women. Several large studies demonstrated that trastuzumab significantly improves disease-free and overall survival when given in combination with, or following, chemotherapy, regardless of hormone receptor status. CONCLUSIONS HER2-targeted therapy maybe combined with AIs for the treatment of ER+/HER2+ metastatic breast cancer in postmenopausal women. HER2-targeted therapy in combination with AIs for treatment of ER+/HER2+ early breast cancer needs to be prospectively evaluated.
Collapse
Affiliation(s)
- A U Buzdar
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
34
|
|
35
|
Zilli M, Grassadonia A, Tinari N, Di Giacobbe A, Gildetti S, Giampietro J, Natoli C, Iacobelli S. Molecular mechanisms of endocrine resistance and their implication in the therapy of breast cancer. Biochim Biophys Acta Rev Cancer 2008; 1795:62-81. [PMID: 18804516 DOI: 10.1016/j.bbcan.2008.08.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2008] [Revised: 08/11/2008] [Accepted: 08/14/2008] [Indexed: 01/04/2023]
Abstract
The use of endocrine agents is a safe and effective treatment in the management of hormone-sensitive breast cancer. Unfortunately, sooner or later, tumor cells develop resistance to endocrine manipulation making useless this approach. During the last decade, new molecules and intracellular signaling pathways involved in endocrine resistance have been identified. Several studies have documented that estrogen receptor signaling may maintain a pivotal role in the tumor growth despite the failure of a previous hormonal treatment. In this review we will discuss the general principles for optimizing the choice of endocrine therapy based on an understanding of the molecular mechanisms responsible for resistance to the different anti-hormonal agents.
Collapse
Affiliation(s)
- Marinella Zilli
- Department of Oncology and Neurosciences, University G D'Annunzio Medical School, Via dei Vestini, Chieti, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Musgrove EA, Sergio CM, Loi S, Inman CK, Anderson LR, Alles MC, Pinese M, Caldon CE, Schütte J, Gardiner-Garden M, Ormandy CJ, McArthur G, Butt AJ, Sutherland RL. Identification of functional networks of estrogen- and c-Myc-responsive genes and their relationship to response to tamoxifen therapy in breast cancer. PLoS One 2008; 3:e2987. [PMID: 18714337 PMCID: PMC2496892 DOI: 10.1371/journal.pone.0002987] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2008] [Accepted: 07/29/2008] [Indexed: 11/30/2022] Open
Abstract
Background Estrogen is a pivotal regulator of cell proliferation in the normal breast and breast cancer. Endocrine therapies targeting the estrogen receptor are effective in breast cancer, but their success is limited by intrinsic and acquired resistance. Methodology/Principal Findings With the goal of gaining mechanistic insights into estrogen action and endocrine resistance, we classified estrogen-regulated genes by function, and determined the relationship between functionally-related genesets and the response to tamoxifen in breast cancer patients. Estrogen-responsive genes were identified by transcript profiling of MCF-7 breast cancer cells. Pathway analysis based on functional annotation of these estrogen-regulated genes identified gene signatures with known or predicted roles in cell cycle control, cell growth (i.e. ribosome biogenesis and protein synthesis), cell death/survival signaling and transcriptional regulation. Since inducible expression of c-Myc in antiestrogen-arrested cells can recapitulate many of the effects of estrogen on molecular endpoints related to cell cycle progression, the estrogen-regulated genes that were also targets of c-Myc were identified using cells inducibly expressing c-Myc. Selected genes classified as estrogen and c-Myc targets displayed similar levels of regulation by estrogen and c-Myc and were not estrogen-regulated in the presence of siMyc. Genes regulated by c-Myc accounted for 50% of all acutely estrogen-regulated genes but comprised 85% (110/129 genes) in the cell growth signature. siRNA-mediated inhibition of c-Myc induction impaired estrogen regulation of ribosome biogenesis and protein synthesis, consistent with the prediction that estrogen regulates cell growth principally via c-Myc. The ‘cell cycle’, ‘cell growth’ and ‘cell death’ gene signatures each identified patients with an attenuated response in a cohort of 246 tamoxifen-treated patients. In multivariate analysis the cell death signature was predictive independent of the cell cycle and cell growth signatures. Conclusions/Significance These functionally-based gene signatures can stratify patients treated with tamoxifen into groups with differing outcome, and potentially identify distinct mechanisms of tamoxifen resistance.
Collapse
Affiliation(s)
- Elizabeth A Musgrove
- Cancer Research Program, Garvan Institute of Medical Research, Sydney, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Huang CS, Kuo SH, Lien HC, Yang SY, You SL, Shen CY, Lin CH, Lu YS, Chang KJ. The CYP19 TTTA repeat polymorphism is related to the prognosis of premenopausal stage I-II and operable stage III breast cancers. Oncologist 2008; 13:751-760. [PMID: 18614591 DOI: 10.1634/theoncologist.2007-0246] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
PURPOSE Given the critical role of the CYP19 gene, encoding aromatase, in estrogen synthesis and the association of the estrogen level with its TTTA repeat polymorphism, the potential influence of this polymorphism on breast cancer survival, and hence management, deserves further study. METHODS Genotyping for the CYP19 TTTA repeat polymorphism was performed on 482 stage I-II and operable stage III Taiwanese breast cancer patients. Patients with more than seven TTTA repeats in either allele of CYP19 were defined as having the long allele. We correlated clinical variables and CYP19 genotypic polymorphism with outcome. RESULTS In hormone receptor (HR)-positive breast cancers, premenopausal patients with the long allele of the CYP19 polymorphism had a significantly higher overall survival (OS) rate (8-year, 89% versus 68%; p= .003) than those without it. This difference was further demonstrated by a multivariate analysis (OS hazard ratio, 1.53; p= .041). In postmenopausal women or patients with HR-negative breast cancer, there was no significant difference in OS between patients with or without the long allele. In premenopausal women with HR-positive cancers, adequate intensity adjuvant chemotherapy did not achieve a greater OS rate than suboptimal chemotherapy in patients with the long allele, but it resulted in a significantly higher OS rate (p= .011) than suboptimal chemotherapy in women without the long allele. CONCLUSIONS The CYP19 TTTA repeat polymorphism is associated with survival in premenopausal women, but not in postmenopausal women, with HR-positive breast cancers. Premenopausal women with the long allele have a greater survival rate and may not gain benefit from adjuvant chemotherapy.
Collapse
Affiliation(s)
- Chiun-Sheng Huang
- Department of Surgery, National Taiwan University Hospital, No. 7, Chung-Shan South Road, Taipei, Taiwan.
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Fjøsne HE, Jacobsen AB, Lundgren S. Adjuvant cyclic Tamoxifen and Megestrol acetate treatment in postmenopausal breast cancer patients – Longterm follow-up. Eur J Surg Oncol 2008; 34:6-12. [PMID: 17881183 DOI: 10.1016/j.ejso.2007.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2007] [Accepted: 07/02/2007] [Indexed: 10/22/2022] Open
Abstract
AIMS To evaluate possible differences in effect on recurrence-free survival (RFS) and overall survival (OS) in node positive postmenopausal breast cancer patients receiving Tamoxifen or cyclic Tam and Megestrol acetate as adjuvant treatment. METHODS Between 1989 and 1994, 489 patients with pT(1-2)pN+ hormone receptor positive or unknown tumours were included in a randomized national multicenter study to receive either Tam alone or cyclic Tam (8 weeks) and MA (8 weeks) for 2 years. Final follow-up was completed as of June 2002. Time from start of treatment to first recurrence and novel primary breast tumour, overall survival and cancer specific survival were estimated. RESULTS No differences in RFS, OS or cancer specific survival were observed between the two treatment groups. CONCLUSIONS Adjuvant treatment used as standard Tam alone, compared to Tam and MA, as employed in this group of patients gave similar outcomes. Side effects that led to cessation of study medication were observed in both arms.
Collapse
Affiliation(s)
- H E Fjøsne
- Breast and Endocrine Unit, Department of Surgery, St. Olavs University Hospital, N-7006 Trondheim, Norway.
| | | | | |
Collapse
|
39
|
Gligorov J, Azria D, Namer M, Khayat D, Spano JP. Novel therapeutic strategies combining antihormonal and biological targeted therapies in breast cancer: focus on clinical trials and perspectives. Crit Rev Oncol Hematol 2007; 64:115-28. [PMID: 17702596 DOI: 10.1016/j.critrevonc.2007.06.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2006] [Revised: 06/25/2007] [Accepted: 06/26/2007] [Indexed: 01/06/2023] Open
Abstract
Several models have been proposed to explain the mechanisms of endocrine resistance including aberrant growth-signaling pathways, and have led to the rational design of studies combining hormonotherapy with signal transduction inhibitors (STI) in advanced breast cancer. This article reviews the current status of these clinical trials. Preliminary results from the randomized controlled trials are rather disappointing. The mTOR inhibitor temsirolimus and the farnesyl transferase inhibitor tipifarnib combined with letrozole did not show any benefit compared to letrozole alone. As neoadjuvant therapy, gefinitib did not enhance the response rate induced by anastrozole. Interesting results were obtained with exemestane combined to celecoxib but should be further explored with adequate cardiac monitoring. Trastuzumab combined with anastrozole was more effective than anastrozole in terms of response rate and progression-free survival but not survival. Several controlled trials as first- or second-line therapy have started recently and over the next few years we should learn whether this approach will provide significant gains in efficacy.
Collapse
Affiliation(s)
- Joseph Gligorov
- Medical Oncology Department, CancerEst, Tenon Hospital, Paris, France
| | | | | | | | | |
Collapse
|
40
|
Abstract
Antagonizing estrogen by inhibition of aromatase has become a mainstay of adjuvant endocrine therapy in women with hormone receptor positive (ER+) breast cancer. Recent trials have shown an incremental gain for the AIs over tamoxifen when given as an up-front alternative to tamoxifen, but additionally added benefit is achieved by giving them in sequence with tamoxifen after either an early switch (2-3 years) or as a late switch (5 years). The true clinical implications of accelerated bone resorption from AIs is becoming better understood and its management defined. AI minimally effect quality of life. The chronic relapsing nature of ER+ breast cancer implies long term therapy will be of benefit in selected patients. Outstanding issues under investigation include optimal duration of endocrine therapy, optimal sequence, optimal agents and whether combining anti-estrogens will yield advantage. The role of AIs is also under investigation in premenopausal women in combination with ovarian function suppression. Identifying prognostic and predictive factors of endocrine therapy is important as is the identification and overcoming of resistance mechanisms. Both tumor and host signatures are being pursued to this end. Optimizing, expanding and extending endocrine therapy is likely to add further to patient outcome.
Collapse
Affiliation(s)
- Paul Goss
- Massachusetts General Hospital Cancer Center, Harvard Medical School, LRH 302, 55 Fruit Street, Boston, MA 02114, USA
| | | |
Collapse
|
41
|
Livi L, Saieva C, Paiar F, Simontacchi G, Galardi A, De Luca Cardillo C, Mangoni M, Paoletti L, Ponticelli P, Biti GP. Use of tamoxifen in pT1a–pT1b, pN0 breast cancer. Eur J Surg Oncol 2007; 33:271-5. [PMID: 16831531 DOI: 10.1016/j.ejso.2006.05.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2006] [Accepted: 05/31/2006] [Indexed: 11/28/2022] Open
Abstract
PURPOSE To evaluate if in low-risk breast cancer patients (pT1a-pT1b, pN0) tamoxifen can reduce local recurrence and improve survival. METHODS Retrospectively 700 patients were analyzed. All patients were treated from 1980 to 2003 with conservative surgery plus radiotherapy at the University of Florence. No patients were treated with adjuvant chemotherapy. Tamoxifen was prescribed in 359 patients (51.3%). The crude probability of survival (or local recurrence) was estimated by using Kaplan-Meier method, and survival (or local recurrence) comparisons were carried out using Cox proportional hazard regression models. RESULTS The univariate analysis for specific survival showed that only histological type and local recurrence were significant prognostic factors (log rank test: p=0.02 and p<0.0001, respectively). The Cox regression model by stepwise selection confirmed lobular histological type (p=0.008; HR: 3.83, 95% CI: 1.31-11.21) and local recurrence (p<0.001; HR: 9.05, 95% CI: 3.05-26.82) as independent prognostic factors for disease specific survival. For local disease free survival, multivariate analysis did not show any significant parameters. CONCLUSION In our series tamoxifen did not seem to improve disease specific survival and local disease specific survival. The number of events in terms of death for cancer or in terms of local recurrence is too small in this group of patients. However, according to our results we suggest not to prescribe tamoxifen in patients affected by pT1a-pT1b, pN0 breast cancer.
Collapse
Affiliation(s)
- L Livi
- Department of Radiotherapy-Oncology, University of Florence, Viale Morgagni No. 85, 50134 Florence, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Aromatase inhibitor-associated arthralgia syndrome. Breast 2007; 16:223-34. [PMID: 17368903 DOI: 10.1016/j.breast.2007.01.011] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2006] [Revised: 01/30/2007] [Accepted: 01/31/2007] [Indexed: 10/23/2022] Open
Abstract
Aromatase inhibitors (AIs) are widely used as an adjuvant endocrine treatment in postmenopausal women with early-stage breast cancer. Clinical trials have assessed 5 years of AI therapy, either as an alternative to tamoxifen for primary adjuvant therapy of breast cancer, or after 5 years of adjuvant tamoxifen. Treatment of 2-3 years' duration after 2-3 years of tamoxifen has also been studied. AI therapy brings side effects related to estrogen deprivation, and this side effect profile differs in clinically relevant ways from that seen with tamoxifen. In particular, the selective estrogen receptor modulatory effects of tamoxifen contribute to menopausal symptoms, vaginal discharge, and the rare but worrisome risks of thromboembolism and uterine carcinoma. By contrast, the low levels of estrogen achieved with aromatase inhibition contribute to menopausal symptoms, vaginal dryness and sexual dysfunction, and accelerated bone demineralization with risk of osteoporosis and osteoporotic fracture. Clinical experience also suggests that AI therapy is associated with a novel musculoskeletal side effect consisting of an arthralgia syndrome. The actual incidence of AI-associated arthralgias or musculoskeletal symptoms is not known, though such symptoms are quite prevalent and appear more commonly with AI use than with tamoxifen. Arthralgias can be a reason for discontinuation of AI treatment. The possible mechanisms of AI-associated arthralgia are unclear. Estrogen deficiency causes bone loss, which in turn contributes to arthralgia. Less well-studied functions of estrogen include regulating immune cells and cytokines involved in bone remodeling, and modulating pain sensitivity at the level of the central nervous system. Arthralgia and arthritis have seldom been rigorously differentiated in clinical trials of AIs. Assessment of inflammatory and rheumatologic markers, as well as detailed evaluation of patient symptoms using appropriate quality-of-life instruments, may be warranted in order to understand both the symptoms and the etiology of the arthralgia syndrome. Treatment options for arthralgia (primarily non-steroidal anti-inflammatory drugs) are currently inadequate, but areas of active research include high-dose vitamin D and new-targeted therapies to inhibit bone loss.
Collapse
|
43
|
Takei H, Suemasu K, Inoue K, Saito T, Okubo K, Koh J, Sato K, Tsuda H, Kurosumi M, Tabei T. Multicenter phase II trial of neoadjuvant exemestane for postmenopausal patients with hormone receptor-positive, operable breast cancer: Saitama Breast Cancer Clinical Study Group (SBCCSG-03). Breast Cancer Res Treat 2007; 107:87-94. [PMID: 18043897 DOI: 10.1007/s10549-007-9529-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2007] [Accepted: 01/19/2007] [Indexed: 01/03/2023]
Abstract
This multicenter phase II trial evaluated the efficacy and tolerability of 4 months of neoadjuvant exemestane in 44 postmenopausal patients with estrogen receptor (ER)-positive and/or progesterone receptor-positive, stage II to IIIB breast cancer measuring >or=3 cm. Pathological response was assessed by a central review board using response criteria proposed by the Japanese Breast Cancer Society. Clinical response [complete or partial response (PR)] was assessed by caliper, mammography, or ultrasound. Rates of breast-conserving surgery (BCS) and adverse events were also evaluated. A pathological response was observed in 13 (43%) of 30 patients who underwent surgery at 4 months. Fourteen patients were excluded from the pathological analysis: eight continued exemestane because of PR or stable disease (SD) at 4 months, three underwent chemotherapy because of progressive disease, and three underwent surgery within 2 months because of adverse events. A clinical response was seen in 27 (66%) of 41 evaluable patients. BCS was performed in 27 (90%) of 30 patients who underwent surgery at 4 months. Of the ten patients eligible for mastectomy at baseline, six chose to continue exemestane treatment without surgery because of a PR or SD at 4 months. Adverse events, most of which were grade 1, occurred in <or=10% of patients. These results suggest that neoadjuvant exemestane treatment is effective and well tolerated in postmenopausal women with ER-positive breast cancer. Further studies are required to determine the optimal duration of neoadjuvant treatment and to identify response criteria that can more accurately predict long-term outcomes.
Collapse
Affiliation(s)
- Hiroyuki Takei
- Division of Breast Surgery, Saitama Cancer Center, Kita-Adachi, Saitama, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Ginés Rubió J, Fernández Cortés F, Rifá Ferrer J. Inhibidores de la aromatasa en el tratamiento adyuvante de pacientes postmenopáusicas con cáncer de mama. Metanálisis. FARMACIA HOSPITALARIA 2007; 31:5-16. [PMID: 17439308 DOI: 10.1016/s1130-6343(07)75705-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
OBJECTIVE To evaluate the efficacy and safety of aromatase inhibitors (AIs) as adjuvant hormonal therapy for postmenopausal patients with hormone receptor positive breast cancer compared with tamoxifen therapy, or as a subsequential treatment to this therapy. METHOD We carried out a bibliographical search using the Medline database and papers presented at the American Society of Clinical Oncology and the San Antonio Breast Cancer Symposium conferences. The efficacy parameters evaluated were disease-free survival (DFS) and overall survival (OS), measured after the follow-up period for each of the tests analysed. The data selected were subjected to a stratified meta-analysis according to the different strategies used to introduce the AIs. The Mantel-Haenszel odds ratio (MH OR; at 95% CI) was calculated for each of the parameters evaluated. The principal side effects with respect to toxicity, described in the various comparative clinical trials, have been listed in a table. RESULTS Compared to tamoxifen, AIs have been shown to increase DFS both as a first-line therapy (MH OR = 0.83; 95% CI: 0.76-0.92) and when sequentially administered for 2-3 years following 2-3 years of tamoxifen treatment (MH OR = 0.65; 95% CI: 0.57-0.75). Similarly, their use after 5 years of tamoxifen therapy also produces an increase in DFS (MH OR = 0.63; 95% CI: 0.51-0.77). As for OS, a statistically significant difference is obtained only when the AI is administered following 2-3 years of tamoxifen treatment (MH OR = 0.77; 95% CI: 0.64-0.94). In comparison with tamoxifen, AIs reduce the incidence of thromboembolic and gynaecologic events, although they increase bone toxicity. CONCLUSIONS The clinical studies evaluated show the consistent benefits of AIs at different adjuvant treatment stages; however, we have been unable to establish the optimum moment for their introduction due to the absence of direct comparisons between the different strategies. We now need to focus on the selection of patient sub-groups which could benefit from their use as a first-line therapy, the long-term toxicity of AIs, and their capacity to increase OS, regardless of the strategy followed, after a longer monitoring period. In light of the evidence available, bearing in mind certain limitations, we propose criteria for the use of AIs in daily clinical practice.
Collapse
Affiliation(s)
- J Ginés Rubió
- Servicio de Farmacia, Hospital Son Dureta, Palma de Mallorca.
| | | | | |
Collapse
|
45
|
Ariazi EA, Lewis-Wambi JS, Gill SD, Pyle JR, Ariazi JL, Kim HR, Sharma CG, Cordera F, Shupp HA, Li T, Jordan VC. Emerging principles for the development of resistance to antihormonal therapy: implications for the clinical utility of fulvestrant. J Steroid Biochem Mol Biol 2006; 102:128-38. [PMID: 17085047 PMCID: PMC1810511 DOI: 10.1016/j.jsbmb.2006.09.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
We seek to evaluate the clinical consequences of resistance to antihormonal therapy by studying analogous animal xenograft models. Two approaches were taken: (1) MCF-7 tumors were serially transplanted into selective estrogen receptor modulator (SERM)-treated immunocompromised mice to mimic 5 years of SERM treatment. The studies in vivo were designed to replicate the development of acquired resistance to SERMs over years of clinical exposure. (2) MCF-7 cells were cultured long-term under SERM-treated or estrogen withdrawn conditions (to mimic aromatase inhibitors), and then injected into mice to generate endocrine-resistant xenografts. These tumor models have allowed us to define Phase I and Phase II antihormonal resistance according to their responses to E(2) and fulvestrant. Phase I SERM-resistant tumors were growth stimulated in response to estradiol (E(2)), but paradoxically, Phase II SERM and estrogen withdrawn-resistant tumors were growth inhibited by E(2). Fulvestrant did not support growth of Phases I and II SERM-resistant tumors, but did allow growth of Phase II estrogen withdrawn-resistant tumors. Importantly, fulvestrant plus E(2) in Phase II antihormone-resistant tumors reversed the E(2)-induced inhibition and instead resulted in growth stimulation. These data have important clinical implications. Based on these and prior laboratory findings, we propose a clinical strategy for optimal third-line therapy: patients who have responded to and then failed at least two antihormonal treatments may respond favorably to short-term low-dose estrogen due to E(2)-induced apoptosis, followed by treatment with fulvestrant plus an aromatase inhibitor to maintain low tumor burden and avoid a negative interaction between physiologic E(2) and fulvestrant.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - V. Craig Jordan
- Corresponding author: V. Craig Jordan, OBE, PhD, DSc, Vice President and Research Director for Medical Sciences, Alfred G. Knudson Chair of Cancer Research, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111-2497, USA. Tel.: +1 215 728 7410; Fax: +1 215 728 7034. E-mail address: (V.C. Jordan)
| |
Collapse
|
46
|
Abstract
Tamoxifen, once the gold standard adjuvant endocrine therapy for early breast cancer, is being challenged by third-generation aromatase inhibitors (AIs) that have demonstrated improved disease-free survival in a variety of adjuvant settings for early breast cancer. Tamoxifen and AIs have different safety profiles, which should allow physicians to begin to individualize treatment based on a patient's comorbidities and risk factors. Because of its properties as a partial estrogen agonist, tamoxifen has a positive effect on serum lipids and may confer a cardioprotective benefit, as well as a beneficial effect on bone health. However, tamoxifen increases the risk for endometrial cancer and cerebrovascular/thromboembolic events. In comparison, the major side effect of AIs is increased bone loss, which may heighten the risk for osteoporotic fractures and bone pain. Because of their superior efficacy and manageable side effects, AIs are a cost-effective alternative to tamoxifen, and clinical guidelines now embrace AIs as appropriate adjuvant therapy for hormone-sensitive early breast cancer. The anticipated results of ongoing trials will provide further insights into the long-term safety and application of AI therapy in the adjuvant setting.
Collapse
|
47
|
Lyon DE, Roux G, Voll S. Hormonal breast cancer agents: implications for the primary care provider. JOURNAL OF THE AMERICAN ACADEMY OF NURSE PRACTITIONERS 2006; 18:518-23. [PMID: 17064329 DOI: 10.1111/j.1745-7599.2006.00168.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
PURPOSE Over 2 million breast cancer survivors are in the United States, making women with breast cancer the largest group of cancer survivors. The purpose of this article is to review the current knowledge regarding survivorship issues in women with early-stage, estrogen receptor-positive breast cancer, focusing on advances in hormonal therapies for reducing risk of recurrence. DATA SOURCES Published research studies, clinical treatment guidelines, and ongoing clinical trials. CONCLUSIONS Innovations in antiestrogenic and estrogen modulator therapies are an important aspect of ongoing care after primary breast cancer treatment. Primary care providers may play an important role in monitoring potential complications of antiestrogenic treatment. IMPLICATIONS FOR PRACTICE This article reviews the current state of the science in hormonal breast cancer agents for breast cancer survivors. With the high incidence and prevalence of breast cancer, primary care providers need to be aware of the potential short- and long-term health risks and benefits of hormonal therapies for breast cancer survivors.
Collapse
Affiliation(s)
- Debra E Lyon
- School of Nursing, Virginia Commonwealth University, Richmond, VA 23298, USA.
| | | | | |
Collapse
|
48
|
Braverman AS, Sawhney H, Tendler A, Patel N, Rao S, El-Tamer M, Kamenova B, Weedon J. Serum estradiol above the postmenopausal level after chemotherapy-induced amenorrhea in breast cancer patients. ACTA ACUST UNITED AC 2006. [DOI: 10.2217/14750708.3.5.609] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
49
|
|