1
|
Al-Kelabi H, Al-Duhaidahawi D, Al-Khafaji K, Al-Masoudi NA. New tamoxifen analogs for breast cancer therapy: synthesis, aromatase inhibition and in silico studies. J Biomol Struct Dyn 2023; 41:12798-12807. [PMID: 36762686 DOI: 10.1080/07391102.2023.2175375] [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: 08/05/2022] [Accepted: 01/07/2023] [Indexed: 02/11/2023]
Abstract
A new class of tamoxifen analogues, using McMurry reaction conditions, is described. The scheme involved the conversion of ketoprofen (6) into amide derivatives 7 and 8, by coupling with N1,N1-substituted propan-1,3-diamine derivatives in the presence DIC and HOB. Treatment of 7 and 8 with various ketones under McMurry reaction conditions afforded the tamoxifen analogues 9-16. All the analogues were screened in vitro for their aromatase inhibitory and antiproliferative activity against MCF-7 breast cancer cells. Compounds 10, 11 and 12 showed a potent activity against MCF-7 cell lines breast cancer with IC50 values of 0.070, 0.042 and 0.077 µM of selectivity index (SI) 3.0, 2.5 and 2.6, respectively. Further, 12 exhibited potent activity against estrogen receptor (14.7 ± 2.4 nM), while compound 10 was the most active analogues against aromatase with IC50 of (0.070 nM). Furthermore, all new compounds were docked into human placental aromatase enzyme and estrogen receptor and showed very good correlations with experimental IC50. Therefore, we can consider these designed compounds as starting scaffold to design an efficient drug against estrogen receptor and aromatase.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Hasan Al-Kelabi
- Pharmacy Department, AL-Najaf Health Directorate, Ministry of Health, Najaf, Iraq
| | - Dunya Al-Duhaidahawi
- Department of Pharmacognosy, College of Pharmacy, University of Kufa Al-Najaf, Kufa, Iraq
| | | | - Najim A Al-Masoudi
- Department of Chemistry, College of Science, University of Basrah, Basrah, Iraq
| |
Collapse
|
2
|
Wang Y, Jing F, Wang H. Role of Exemestane in the Treatment of Estrogen-Receptor-Positive Breast Cancer: A Narrative Review of Recent Evidence. Adv Ther 2022; 39:862-891. [PMID: 34989983 DOI: 10.1007/s12325-021-01924-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/14/2021] [Indexed: 11/01/2022]
Abstract
INTRODUCTION Breast cancer (BC) is the most common type of cancer diagnosed among women worldwide with an estimated 2.3 million new cases every year. Almost two-thirds of all patients with BC have estrogen receptor-positive (ER+) tumors. In this review, the clinical evidence of exemestane in different treatment settings in ER+ BC is presented and summarized. SEARCH STRATEGY A search strategy with the keywords "breast cancer [MeSH Terms]" AND "exemestane [Title/Abstract]" was devised and a search was performed in PubMed. RESULTS The efficacy of exemestane in different treatment settings has been established by numerous clinical studies. Exemestane is recommended as an adjuvant treatment in postmenopausal women previously treated with tamoxifen in trials comparing 5 years of tamoxifen with 2-3 years of tamoxifen combined with 2-3 years of exemestane, which proved that treatment with exemestane provided better survival outcomes. Similarly, exemestane could be considered as a safe treatment option for neoadjuvant treatment, prevention of chemotherapy, and treatment of advanced BC either alone or in combination with other targeted therapy drugs in both pre- and postmenopausal women. CONCLUSION Exemestane could be considered as a reasonable therapeutic option in the treatment of ER+ BC at any stage in pre- and postmenopausal women.
Collapse
|
3
|
Gölz C, Kirchhoff FP, Westerhorstmann J, Schmidt M, Hirnet T, Rune GM, Bender RA, Schäfer MKE. Sex hormones modulate pathogenic processes in experimental traumatic brain injury. J Neurochem 2019; 150:173-187. [PMID: 30790293 DOI: 10.1111/jnc.14678] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 01/28/2019] [Accepted: 01/29/2019] [Indexed: 12/26/2022]
Abstract
Clinical and animal studies have revealed sex-specific differences in histopathological and neurological outcome after traumatic brain injury (TBI). The impact of perioperative administration of sex steroid inhibitors on TBI is still elusive. Here, we subjected male and female C57Bl/6N mice to the controlled cortical impact (CCI) model of TBI and applied pharmacological inhibitors of steroid hormone synthesis, that is, letrozole (LET, inhibiting estradiol synthesis by aromatase) and finasteride (FIN, inhibiting dihydrotestosterone synthesis by 5α-reductase), respectively, starting 72 h prior CCI, and continuing for a further 48 h after CCI. Initial gene expression analyses showed that androgen (Ar) and estrogen receptors (Esr1) were sex-specifically altered 72 h after CCI. When examining brain lesion size, we found larger lesions in male than in female mice, but did not observe effects of FIN or LET treatment. However, LET treatment exacerbated neurological deficits 24 and 72 h after CCI. On the molecular level, FIN administration reduced calpain-dependent spectrin breakdown products, a proxy of excitotoxicity and disturbed Ca2+ homeostasis, specifically in males, whereas LET increased the reactive astrocyte marker glial fibrillary acid protein specifically in females. Examination of neurotrophins (brain-derived neurotrophic factor, neuronal growth factor, NT-3) and their receptors (p75NTR , TrkA, TrkB, TrkC) revealed CCI-induced down-regulation of TrkB and TrkC protein expression, which was reduced by LET in both sexes. Interestingly, FIN decreased neuronal growth factor mRNA expression and protein levels of its receptor TrkA only in males. Taken together, our data suggest a sex-specific impact on pathogenic processes in the injured brain after TBI. Sex hormones may thus modulate pathogenic processes in experimental TBI.
Collapse
Affiliation(s)
- Christina Gölz
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Florian Paul Kirchhoff
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | | | - Matthias Schmidt
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Tobias Hirnet
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Gabriele M Rune
- Institute of Neuroanatomy, University Medical Center, Hamburg, Germany
| | - Roland A Bender
- Institute of Neuroanatomy, University Medical Center, Hamburg, Germany
| | - Michael K E Schäfer
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany.,Focus Program Translational Neurosciences, Mainz, Germany.,Research Center for Immunotherapy (FZI), Mainz, Germany
| |
Collapse
|
4
|
Abstract
The last 100 years have seen a dramatic alteration in the treatment of cancer. Aside from small molecule inhibitors of protein tyrosine kinases, monoclonal antibodies have also been found to provide valuable therapeutic approaches for modulating tumour pathophysiology. As our knowledge of cancer biology improves, the specificity of this new generation of drugs is generally delivering an improved therapeutic ratio compared to traditional cytotoxic agents. However, patient selection through the use of biomarkers is key in optimising efficacy and improving cost-effectiveness. The most recent wave of revolutionary new systemic therapy approaches to cancer has arrived in recent years in the form of immune checkpoint inhibitors, now clinically validated as modulators of immune-regulatory pathways. The future of oncology therapeutics includes a combination of cytotoxic agents, targeted therapies and immunotherapy.
Collapse
Affiliation(s)
| | - James Spicer
- King's Health Partners at Guy's Hospital, London, UK.
| |
Collapse
|
5
|
Investigation of genotoxicity risk and DNA repair capacity in breast cancer patients using anastrozole. North Clin Istanb 2018; 5:6-13. [PMID: 29607425 PMCID: PMC5864710 DOI: 10.14744/nci.2017.55822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 08/21/2017] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE: Breast cancer is the most common cancer in women worldwide and the incidence increases in postmenopausal women. Anastrozole is a non-steroidal (type II), third-generation aromatase inhibitor (AI) that is used in the treatment of postmenopausal estrogen-related breast cancer. Several studies have been conducted to assess the efficacy, safety, and superiority of AIs to tamoxifen; however, a literature search did not reveal a study that investigated the genotoxic potential of AIs. The aim of this study was to investigate the possible DNA damage risk profile and individual DNA repair capacity of patients using anastrozole with the modified alkaline comet assay in order to contribute to public health and health economics. METHODS: Women diagnosed with breast cancer after menopause comprised the study group. Six patients who had taken anastrozole for at least 6 months were retrospectively enrolled, and 12 patients who had not yet received treatment were prospectively enrolled as a control group. Peripheral blood lymphocytes were used to measure oxidized DNA damage using formamidopyrimidine DNA-glycosylase (FPG) and endonuclease III (endo III) in a modified comet assay. Individual DNA repair capacity was evaluated with the comet assay after a hydrogen peroxide (H2O2) challenge to examine the difference in DNA damage susceptibility. RESULTS: Analysis of DNA damage, oxidative base damage, susceptibility to DNA damage, and repair capacity revealed no significant difference between the control group and the patients taking anastrozole (p>0.05). Susceptibility to H2O2 damage was observed to increase with age (p<0.05). CONCLUSION: According to the results obtained in this study, anastrozole did not contribute to oxidative DNA damage. An H2O2 challenge with the comet assay is useful to evaluate circumstances of increased vulnerability to damage, such as aging and cancer.
Collapse
|
6
|
Abstract
The efficient production, folding, and secretion of proteins is critical for cancer cell survival. However, cancer cells thrive under stress conditions that damage proteins, so many cancer cells overexpress molecular chaperones that facilitate protein folding and target misfolded proteins for degradation via the ubiquitin-proteasome or autophagy pathway. Stress response pathway induction is also important for cancer cell survival. Indeed, validated targets for anti-cancer treatments include molecular chaperones, components of the unfolded protein response, the ubiquitin-proteasome system, and autophagy. We will focus on links between breast cancer and these processes, as well as the development of drug resistance, relapse, and treatment.
Collapse
Affiliation(s)
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, A320 Langley Hall, 4249 Fifth Ave, Pittsburgh, PA, 15260, USA.
| |
Collapse
|
7
|
Manna PR, Molehin D, Ahmed AU. Dysregulation of Aromatase in Breast, Endometrial, and Ovarian Cancers: An Overview of Therapeutic Strategies. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 144:487-537. [PMID: 27865465 DOI: 10.1016/bs.pmbts.2016.10.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Aromatase is the rate-limiting enzyme in the biosynthesis of estrogens, which play crucial roles on a spectrum of developmental and physiological processes. The biological actions of estrogens are classically mediated by binding to two estrogen receptors (ERs), ERα and ERβ. Encoded by the cytochrome P450, family 19, subfamily A, polypeptide 1 (CYP19A1) gene, aromatase is expressed in a wide variety of tissues, as well as benign and malignant tumors, and is regulated in a pathway- and tissue-specific manner. Overexpression of aromatase, leading to elevated systemic levels of estrogen, is unequivocally linked to the pathogenesis and growth of a number malignancies, including breast, endometrium, and ovarian cancers. Aromatase inhibitors (AIs) are routinely used to treat estrogen-dependent breast cancers in postmenopausal women; however, their roles in endometrial and ovarian cancers remain obscure. While AI therapy is effective in hormone sensitive cancers, they diminish estrogen production throughout the body and, thus, generate undesirable side effects. Despite the effectiveness of AI therapy, resistance to endocrine therapy remains a major concern and is the leading cause of cancer death. Considerable advances, toward mitigating these issues, have evolved in conjunction with a number of histone deacetylase (HDAC) inhibitors for countering an assortment of diseases and cancers, including the aforesaid malignancies. HDACs are a family of enzymes that are frequently dysregulated in human tumors. This chapter will discuss the current understanding of aberrant regulation and expression of aromatase in breast, endometrial, and ovarian cancers, and potential therapeutic strategies for prevention and treatment of these life-threatening diseases.
Collapse
Affiliation(s)
- P R Manna
- Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX, United States.
| | - D Molehin
- Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX, United States
| | - A U Ahmed
- Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX, United States
| |
Collapse
|
8
|
Briest S, Stearns V. Advances in the Adjuvant and Neoadjuvant Treatment of Breast Cancer. WOMENS HEALTH 2016; 3:325-39. [DOI: 10.2217/17455057.3.3.325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Several advances in the adjuvant systemic therapy of primary breast cancer have occurred in the last decade and contributed to a decline in disease-related mortality. These include the introduction of aromatase inhibitors, new chemotherapy agents, and the novel antibody trastuzumab. New supportive treatments, such as growth factors, have contributed to the optimization of chemotherapy dose and schedule, and have improved the efficacy and safety of the treatment. In this review we will outline some of the recent advances in the adjuvant and neoadjuvant treatment of breast cancer. We will also discuss ongoing and proposed clinical trials.
Collapse
Affiliation(s)
- Susanne Briest
- University of Leipzig, Department of Gynecology and Obstetrics, Leipzig Germany
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 1650 Orleans Street, CRB I, Room 186, Baltimore, MD 21231-1000, USA, Tel.: +1 410 502 3472; Fax: +1 410 614 9421
| | - Vered Stearns
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, 1650 Orleans Street, CRB I, Room 1M-53 Baltimore, MD 21231-1000, USA, Tel.: +1 443 287 6489; Fax: +1 410 955 0125
| |
Collapse
|
9
|
Dixon JM. Extended adjuvant therapy with letrozole: reducing the risk of recurrence. Expert Rev Anticancer Ther 2014; 6:849-59. [PMID: 16761928 DOI: 10.1586/14737140.6.6.849] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Patients with all stages of primary breast cancer are at continuing risk of relapse following 5 years of adjuvant tamoxifen therapy, even in the absence of lymph node involvement. Tamoxifen has been the standard therapy for reducing risk of recurrence, although more than 50% of relapses and deaths occur after completion of tamoxifen. Tamoxifen use is associated with an increased risk of serious side effects, and extended use beyond 5 years may have a negative impact on disease-free survival. Extended adjuvant letrozole therapy confers a significant benefit in relapse-free survival. The approval of letrozole for this indication in the USA and in many European countries introduces a new, safe and effective treatment for disease-free patients seeking to reduce their long-term risk of recurrence.
Collapse
Affiliation(s)
- J Michael Dixon
- Edinburgh Breast Unit, Western General Hospital, Edinburgh EH4 2XU, UK.
| |
Collapse
|
10
|
Brady N, Chuntova P, Bade LK, Schwertfeger KL. The FGF/FGFR axis as a therapeutic target in breast cancer. Expert Rev Endocrinol Metab 2013; 8:391-402. [PMID: 25400686 PMCID: PMC4228698 DOI: 10.1586/17446651.2013.811910] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Fibroblast growth factor receptor (FGFR) signaling is a vital component of both embryonic and postnatal mammary gland development, which has prompted researchers to investigate both its relevance to breast cancer and its potential as a therapeutic target. Deregulated FGFR signaling during breast cancer occurs through various mechanisms, including amplification of the receptor genes, aberrant ligand expression, receptor mutations and translocations. Recent experimental outcomes involving both animal models and human breast cancer cell lines have led to the initiation of multiple early clinical trials investigating the safety and efficacy of small molecule FGFR inhibitors. In this article we review both the most recent discoveries and the need for further investigation of the mechanisms through which FGF/FGFR signaling has emerged as an oncogenic driver.
Collapse
Affiliation(s)
- Nicholas Brady
- Microbiology, Immunology and Cancer Biology Graduate Program, University of Minnesota, 420 Delaware St. SE, MMC 609, Minneapolis, MN 55455
| | - Polly Chuntova
- Microbiology, Immunology and Cancer Biology Graduate Program, University of Minnesota, 420 Delaware St. SE, MMC 609, Minneapolis, MN 55455
| | - Lindsey K Bade
- Molecular, Cellular, Developmental Biology and Genetics Graduate Program, University of Minnesota, 420 Delaware St. SE, MMC 609, Minneapolis, MN 55455
| | - Kathryn L Schwertfeger
- Department of Laboratory Medicine and Pathology, University of Minnesota, 420 Delaware St. SE, MMC 609, Minneapolis, MN 55455
- Masonic Cancer Center, University of Minnesota, 420 Delaware St. SE, MMC 609, Minneapolis, MN 55455
| |
Collapse
|
11
|
Penney RB, Roy D. Thioredoxin-mediated redox regulation of resistance to endocrine therapy in breast cancer. Biochim Biophys Acta Rev Cancer 2013; 1836:60-79. [PMID: 23466753 DOI: 10.1016/j.bbcan.2013.02.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 02/18/2013] [Accepted: 02/19/2013] [Indexed: 12/27/2022]
Abstract
Resistance to endocrine therapy in breast carcinogenesis due to the redox regulation of the signal transduction system by reactive oxygen species (ROS) is the subject of this review article. Both antiestrogens and aromatase inhibitors are thought to prevent cancer through modulating the estrogen receptor function, but other mechanisms cannot be ruled out as these compounds also block metabolism and redox cycling of estrogen and are free radical scavengers. Endocrine therapeutic agents, such as, tamoxifen and other antiestrogens, and the aromatase inhibitor, exemestane, are capable of producing ROS. Aggressive breast cancer cells have high oxidative stress and chronic treatment with exemestane, fulvestrant or tamoxifen may add additional ROS stress. Breast cancer cells receiving long-term antiestrogen treatment appear to adapt to this increased persistent level of ROS. This, in turn, may lead to the disruption of reversible redox signaling that involves redox-sensitive phosphatases, protein kinases, such as, ERK and AKT, and transcription factors, such as, AP-1, NRF-1 and NF-κB. Thioredoxin modulates the expression of estrogen responsive genes through modulating the production of H2O2 in breast cancer cells. Overexpressing thioredoxine reductase 2 and reducing oxidized thioredoxin restores tamoxifen sensitivity to previously resistant breast cancer cells. In summary, it appears that resistance to endocrine therapy may be mediated, in part, by ROS-mediated dysregulation of both estrogen-dependent and estrogen-independent redox-sensitive signaling pathways. Further studies are needed to define the mechanism of action of thioredoxin modifiers, and their effect on the redox regulation that contributes to restoring the antiestrogen-mediated signal transduction system and growth inhibitory action.
Collapse
Affiliation(s)
- Rosalind Brigham Penney
- Department of Environmental and Occupational Health, Florida International University, Miami, FL 33199, USA
| | | |
Collapse
|
12
|
Fritz H, Seely D, McGowan J, Skidmore B, Fernandes R, Kennedy DA, Cooley K, Wong R, Sagar S, Balneaves LG, Fergusson D. Black Cohosh and Breast Cancer. Integr Cancer Ther 2013; 13:12-29. [DOI: 10.1177/1534735413477191] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Background. Many women use black cohosh as a natural treatment for menopausal symptoms. However, controversy exists around safety in breast cancer, because of its purported estrogenic activity. We conducted a systematic review of black cohosh use in women with or at risk of breast cancer. Methods. We searched MEDLINE, Embase, the Cochrane Library, and AMED from inception to July 2012 and October 2012 for human interventional or observational data pertaining to the safety and efficacy of black cohosh in patients with or at risk of breast cancer, including an assessment of the effect of black cohosh on estrogen responsive tissues. Results. Of 450 records, we included 26 articles: 14 randomized controlled trials, 7 uncontrolled trials, and 5 observational studies.The evidence on efficacy for ho t flashes is divided, with some benefits seen when compared with baseline, but not when compared with placebo. Two observational studies found no association between black cohosh and risk of breast cancer, whereas 2 studies reported significant reductions in risk of primary breast cancer among postmenopausal women (adjusted odds ratio = 0.47, 95% confidence interval = 0.27-0.82), and risk of recurrence (adjusted hazard ratio = 0.75, 95% confidence interval = 0.63-0.89). Seventeen trials showed no significant impact on circulating hormone levels or proliferation in estrogen responsive tissues. Conclusions. Current evidence does not support an association between black cohosh and increased risk of breast cancer. There is a lack of evidence supporting the efficacy of black cohosh for reduction of hot flashes in breast cancer patients. Given conflicting but promising results, and apparent safety, further research is warranted.
Collapse
Affiliation(s)
- Heidi Fritz
- The Canadian College of Naturopathic Medicine, Toronto, Ontario, Canada
| | - Dugald Seely
- The Canadian College of Naturopathic Medicine, Toronto, Ontario, Canada
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Ottawa Integrative Cancer Center, Ottawa, Ontario, Canada
| | - Jessie McGowan
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Becky Skidmore
- The Canadian College of Naturopathic Medicine, Toronto, Ontario, Canada
| | | | - Deborah A. Kennedy
- The Canadian College of Naturopathic Medicine, Toronto, Ontario, Canada
- The University of Toronto, Toronto, Ontario, Canada
| | - Kieran Cooley
- The Canadian College of Naturopathic Medicine, Toronto, Ontario, Canada
- The University of Toronto, Toronto, Ontario, Canada
| | - Raimond Wong
- Juravinski Cancer Center, Hamilton, Ontario, Canada
- McMaster University, Hamilton, Ontario, Canada
| | - Stephen Sagar
- Juravinski Cancer Center, Hamilton, Ontario, Canada
- McMaster University, Hamilton, Ontario, Canada
| | | | - Dean Fergusson
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| |
Collapse
|
13
|
González-Neira A. Pharmacogenetics of chemotherapy efficacy in breast cancer. Pharmacogenomics 2012; 13:677-90. [PMID: 22515610 DOI: 10.2217/pgs.12.44] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Large differences are observed in chemotherapy response between breast cancer patients, with a substantial part of this variability being explained by genetic factors. Polymorphisms in genes encoding drug-metabolizing enzymes, drug transporters and drug targets influence the pharmacokinetics and pharmacodynamics of these anticancer drugs, leading to differences in therapeutic efficacy. Pharmacogenetic investigations of breast cancer therapeutics focused on these candidate loci have been performed. This article summarizes the status of research to identify polymorphisms in genes that influence response to the chemotherapeutic agents used in breast cancer treatment and suggests future directions for this line of research. Understanding the genetic factors that predispose patients to poor treatment outcomes will help guide individualized therapeutic strategies to obtain maximal benefit.
Collapse
Affiliation(s)
- Anna González-Neira
- Human Genotyping Unit, Human Cancer Genetics Programme, Spanish National Cancer Research Centre, Madrid, Spain.
| |
Collapse
|
14
|
Development and validation of a liquid chromatography–tandem mass spectrometry method for the simultaneous quantification of tamoxifen, anastrozole, and letrozole in human plasma and its application to a clinical study. Anal Bioanal Chem 2010; 398:1791-800. [DOI: 10.1007/s00216-010-4075-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Revised: 07/02/2010] [Accepted: 07/30/2010] [Indexed: 11/26/2022]
|
15
|
Marsh S, Liu G. Pharmacokinetics and pharmacogenomics in breast cancer chemotherapy. Adv Drug Deliv Rev 2009; 61:381-7. [PMID: 19100797 DOI: 10.1016/j.addr.2008.10.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2008] [Accepted: 10/02/2008] [Indexed: 10/21/2022]
Abstract
Locally advanced or metastatic breast cancer is typically treated with chemotherapy. Multiple combinations of chemotherapy regimens are available, including anthracyclines, taxanes, antimetabolites, alkylating agents, platinum drugs and vinca alkaloids. This review discusses the pharmacokinetic and pharmacogenomic information available for commonly used breast cancer chemotherapy drugs. Pharmacogenomic associations for many drugs have yet to be identified or validated in breast cancer. Further work is needed to identify markers to screen breast cancer patients prior to therapy selection.
Collapse
|
16
|
Abstract
The use of aromatase inhibitors (AIs) as adjuvant endocrine therapy for hormone-sensitive breast cancer is increasing, as these drugs are more effective than tamoxifen alone in improving disease-free survival in breast cancer patients-whether used in lieu of tamoxifen as upfront therapy or after tamoxifen treatment periods of 2 years or longer. AIs differ from tamoxifen in their mechanism of action, effectively suppressing estrogen levels in postmenopausal women to near-undetectable levels. AI-associated adverse events largely mimic menopausal symptoms, including hot flashes, losses in bone mineral density, gynecologic symptoms, and arthralgias. The AIs lack the infrequent but potentially serious adverse events associated with tamoxifen (eg, endometrial cancer, thromboembolic events, and stroke). Large randomized studies of AIs in the adjuvant setting have not demonstrated an adverse effect on lipids and cardiovascular health, but postmenopausal women receiving AIs are at risk for age-related changes in lipid parameters and an increased risk for cardiovascular events. To optimize the overall benefits of adjuvant endocrine therapy with an AI, patients should be monitored for bone loss and cardiovascular risk factors, and symptoms such as joint pain and vaginal dryness should be anticipated and managed proactively.
Collapse
|
17
|
Abstract
Third-generation aromatase inhibitors (AIs), including letrozole, are now standard therapy for initial adjuvant endocrine treatment of postmenopausal women with early breast cancer. The International Breast Cancer Study Group's Breast International Group (BIG) 1-98 trial is examining efficacy and safety of letrozole or tamoxifen, whether used upfront or sequentially, for postmenopausal with hormone receptor-positive breast cancer. Women in the BIG 1-98 trial were randomized to either 5 years' monotherapy with tamoxifen or letrozole (trial arms A and B, respectively), or to 5 years' sequential therapy with 2 years of tamoxifen followed by 3 years of letrozole, or 2 years of letrozole followed by 3 years of tamoxifen (arms C and D, respectively). Results of the primary core analysis (PCA) of BIG 1-98, at a median follow-up of 25.8 months, showed a significant improvement in disease-free survival for patients treated with initial adjuvant letrozole relative to those on tamoxifen (P=0.003). Of interest, there was an important reduction in distant metastases, the most common and ultimately fatal type of early recurrence, for patients allocated to initial letrozole compared with initial tamoxifen. Since the PCA, there have been further analyses of BIG 1-98 with variations in the cohorts analyzed and follow-up period. The first and largest central pathology review in an adjuvant AI trial to date was also performed in the BIG 1-98 trial. In light of the impending sequence analysis, expected in late 2008, the goal of this article is to describe and summarize the breadth of information learned to date and highlight the key findings regarding the efficacy and safety of letrozole in the initial adjuvant setting.
Collapse
Affiliation(s)
- Andrew M Wardley
- Department of Medical Oncology, Christie Hospital, Manchester, UK.
| |
Collapse
|
18
|
Pennery E. The role of endocrine therapies in reducing risk of recurrence in postmenopausal women with hormone receptor-positive breast cancer. Eur J Oncol Nurs 2008; 12:233-43. [PMID: 18372213 DOI: 10.1016/j.ejon.2008.01.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2007] [Revised: 01/18/2008] [Accepted: 01/22/2008] [Indexed: 10/22/2022]
Abstract
Hormone receptor-positive breast cancer is increasingly considered a chronic disease, as there remains an ongoing risk of local and distant relapse for years after diagnosis. While early recurrence risk peaks 2-3 years post diagnosis, the majority of breast cancer recurrences and deaths occur following 5 years of adjuvant tamoxifen. Aromatase inhibitors have achieved greater relative reductions in recurrence risk than tamoxifen alone and are now widely recommended as adjuvant therapy for postmenopausal women with hormone-sensitive breast cancer. Although both anastrozole and letrozole have demonstrated superior disease-free survival compared with tamoxifen, letrozole to date offers the greatest significant reduction in the risk of distant metastases in patients with hormone-sensitive breast cancer. Anastrozole and exemestane also reduce local and distant recurrence risk in the "switch setting" following 2-3 years of tamoxifen. Extended adjuvant letrozole, given after 5 years of tamoxifen, significantly reduces local and distant recurrence as well as mortality in patients with node-positive disease. Specialist nurses and nurse practitioners facilitate informed choice for breast cancer patients through explaining treatment options and side effects; they thus need an understanding of which treatment strategies reduce recurrence risk, especially the risk of distant metastases.
Collapse
Affiliation(s)
- Emma Pennery
- Breast Cancer Care, Kiln House, London SE1 0NS, UK.
| |
Collapse
|
19
|
Paepke S, Jacobs VR, Ohlinger R, Warm M, Kümmel S, Thomas A, Harbeck N, Kiechle-Bahat M. Treatment strategies that effectively reduce early recurrence risk in postmenopausal women with endocrine-sensitive breast cancer: AIs upfront vs. switching. J Cancer Res Clin Oncol 2007; 133:905-16. [PMID: 17805570 DOI: 10.1007/s00432-007-0297-7] [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: 03/01/2007] [Accepted: 07/27/2007] [Indexed: 12/16/2022]
Abstract
Several large, well-controlled clinical trials have now established that the aromatase inhibitors (AIs), including letrozole, anastrozole, and exemestane, are more effective than tamoxifen when used as adjuvant endocrine therapy in postmenopausal women with breast cancer. Yet, it is an open question as to how these drugs should be best integrated into the adjuvant treatment regimen. Both letrozole and anastrozole have shown efficacy over tamoxifen when used as initial adjuvant therapy (initiated just following surgery for breast cancer), while exemestane and anastrozole have been used as switching adjuvant therapy, i.e., following 2-3 years of initial adjuvant tamoxifen therapy, with proven efficacy over continued tamoxifen. Studies demonstrate that recurrence risk peaks in the early period after surgery, and that distant metastases in particular, accounting for most of the early recurrences, have worse survival rates when compared with other types of recurrences. Treatments that reduce recurrences, especially distant metastases, in this early period are therefore likely to improve overall survival (OS) and reduce mortality from breast cancer. In this review, we discuss early recurrence risk among postmenopausal women with successfully treated early breast cancer, the efficacy of the different AIs in reducing early recurrences and distant metastases when incorporated into adjuvant therapy, and the evidence for increased OS when AIs are used as initial or switch adjuvant therapy.
Collapse
Affiliation(s)
- Stefan Paepke
- Interdisciplinary Breast Center, Operative Senology, Frauenklinik (OB/GYN), Technical University Munich, Ismaninger Strasse 22, 81675 Munich, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Su B, Landini S, Davis DD, Brueggemeier RW. Synthesis and biological evaluation of selective aromatase expression regulators in breast cancer cells. J Med Chem 2007; 50:1635-44. [PMID: 17315855 DOI: 10.1021/jm061133j] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Aromatase converts androgens to estrogens and is a particularly attractive target in the treatment of estrogen receptor positive breast cancer. The enzyme is encoded by the CYP19 gene, which is expressed in a tissue-specific manner. Prostaglandin E2 (PGE2), the major product of cyclooxygenase-2 (COX-2), stimulates aromatase gene expression via protein kinase A and C signaling pathways. Our previous study demonstrated that COX-2 selective inhibitor nimesulide decreased aromatase activity from the transcriptional level in breast cancer cells. In this manuscript, the synthesis and biological evaluation of a series of nimesulide analogues as potential selective aromatase expression regulators are described. Several novel sulfonanilide compounds demonstrate IC50 values from 0.33 to 2.68 microM in suppressing aromatase enzyme activity in SK-BR-3 breast cancer cells and are 10- to 80-fold more active than nimesulide. Also, the sulfonanilide compounds selectively decrease aromatase gene expression in breast cancer cells, without exhibiting cytotoxic or apoptotic effects at low micromole concentrations.
Collapse
Affiliation(s)
- Bin Su
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, and Ohio State Biochemistry Program, The Ohio State University, 500 W. 12th Avenue, Columbus, Ohio 43210, USA
| | | | | | | |
Collapse
|
21
|
Spicer J, Ellis P. Towards optimal endocrine therapy for hormone-sensitive breast cancer: Initial versus sequential adjuvant aromatase inhibition. Cancer Lett 2007; 248:165-74. [PMID: 16919870 DOI: 10.1016/j.canlet.2006.07.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2006] [Revised: 07/03/2006] [Accepted: 07/04/2006] [Indexed: 11/15/2022]
Abstract
Patients with hormone-receptor-positive early breast cancer have a significant risk of recurrence despite the use of adjuvant tamoxifen. The third-generation aromatase inhibitors letrozole, anastrozole, and exemestane offer promise as alternative or additional therapy to tamoxifen. As extended adjuvant therapy following completion of 5 years of tamoxifen, letrozole further decreased the risk of recurrence compared with placebo. Compared with tamoxifen, both letrozole and anastrozole significantly reduced the risk of recurrence when used as initial adjuvant therapy, and in the short term both were better tolerated than tamoxifen. Anastrozole and exemestane both reduced the risk of relapse when started after 2-3 years of tamoxifen compared with continued tamoxifen treatment; the results of letrozole in this setting are expected in 2008. These data establish adjuvant aromatase inhibitors as effective alternatives to tamoxifen. Only limited data are currently available to inform the choice between an aromatase inhibitor as either initial adjuvant therapy or sequentially after tamoxifen. Future results from the Breast International Group (BIG) 1-98 trial will further clarify strategies for the adjuvant use of aromatase inhibitors. Here, we critically review the evidence for adjuvant use of aromatase inhibitors. Comparison is made between initial aromatase inhibition, switching, and extended adjuvant strategies. Practical recommendations are given for the endocrine treatment of post-menopausal breast cancer.
Collapse
Affiliation(s)
- J Spicer
- Department of Medical Oncology, Academic Oncology Offices, 3rd Floor, Thomas Guy House, Guy's and St Thomas' Hospital, St Thomas Street, London, UK.
| | | |
Collapse
|
22
|
Kaufmann M, Rody A. Extended breast cancer treatment with an aromatase inhibitor (Letrozole) after tamoxifen: why, who and how long? Eur J Obstet Gynecol Reprod Biol 2006; 126:146-54. [PMID: 16621229 DOI: 10.1016/j.ejogrb.2006.03.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2005] [Revised: 02/10/2006] [Accepted: 03/13/2006] [Indexed: 10/24/2022]
Abstract
Breast cancer remains a leading cause of cancer death in women worldwide, and the risk for disease recurrence continues despite improvements in screening and treatment and the use of prophylactic estrogen-inhibiting therapies such as tamoxifen. A number of long-term studies now indicate a significant risk for breast cancer recurrence among patients who have undergone the currently recommended five years of tamoxifen adjuvant therapy following successful treatment of their initial disease. This ongoing recurrence risk extends even to patients commonly considered at low risk for relapse, that is, those with low-grade, small tumors, and/or node-negative disease. Treatment with tamoxifen for more than five years appears detrimental rather than beneficial and, therefore, tamoxifen is not indicated for use beyond the initial five years. Endometrial cancer and thromboembolism are among the serious adverse events that have been observed with long-term tamoxifen treatment. The aromatase inhibitors are able to reduce overall estrogen levels and appear to be better tolerated over a long term. Letrozole is the most potent aromatase inhibitor and has been available in Europe since 1996 and in the United States since 1997. Letrozole has been approved for first-line treatment of postmenopausal women with hormone-receptor-positive or hormone-receptor-unknown, advanced or metastatic breast cancer in the United States and Europe, as well as for neoadjuvant treatment (primary systemic therapy) of early breast cancer prior to surgery in many countries. The results of the pivotal MA-17 trial demonstrate that letrozole is unique in its ability to improve disease-free survival in breast cancer patients who have undergone tamoxifen therapy for five years. The MA-17 results indicate that extended adjuvant therapy with letrozole reduces risk of recurrence in this setting by 42%, reduces risk of distant recurrence (metastasis), and may improve patient survival in the node-positive patient population. The results also show letrozole to be well tolerated and safe over the length of follow-up. The trial outcomes have led to the approval of letrozole for the extended adjuvant indication in more than 40 countries worldwide. Re-randomization of letrozole-treated patients from this pivotal trial is underway to investigate if ten years of extended adjuvant endocrine therapy leads to further improvement, and the results of this extension study should aid in resolving several open questions regarding extended adjuvant therapy, including who should be treated and for how long.
Collapse
Affiliation(s)
- Manfred Kaufmann
- Department of Obstetrics and Gynaecology, Johann Wolfgang Goethe Universität, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| | | |
Collapse
|
23
|
O'Driscoll L, Clynes M. Molecular markers of multiple drug resistance in breast cancer. Chemotherapy 2006; 52:125-9. [PMID: 16612055 DOI: 10.1159/000092540] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2005] [Accepted: 07/27/2005] [Indexed: 11/19/2022]
Abstract
Breast cancer is a significant health problem in terms of both morbidity and mortality, with approximately 12% of women directly affected by this disease. Chemotherapy, given to patients with earlier stage disease, has a good survival impact and may contribute to cure. The failure of chemotherapeutic drugs to eradicate cancer cells in more advanced disease states may be due to intrinsic or acquired drug resistance, including multiple drug resistance. The drug resistance observed in breast cancer patients is likely to be multifactorial, involving mechanisms such as altered expression and/or activity of drug efflux pumps, nuclear DNA-binding enzymes, metabolizing and conjugating enzymes, and mismatch repair deficiency. More extensive transcriptomic and proteomic analyses of breast tumour and normal biopsies, followed by functional genomic studies in relevant cell line models, should increase our understanding of this phenomenon and lead to therapies being individualized for identifiable subgroups of breast cancer patients.
Collapse
Affiliation(s)
- L O'Driscoll
- National Institute for Cellular Biotechnology, Dublin City University, Ireland.
| | | |
Collapse
|
24
|
Dunn C, Keam SJ. Letrozole: a pharmacoeconomic review of its use in postmenopausal women with breast cancer. PHARMACOECONOMICS 2006; 24:495-517. [PMID: 16706574 DOI: 10.2165/00019053-200624050-00007] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Letrozole (Femara), an aromatase inhibitor that blocks estrogen synthesis by inhibiting the final step of the estrogen biosynthetic pathway, is approved for use in a wide range of breast cancer settings. Randomised clinical trials in postmenopausal women with hormone-responsive early-stage breast cancer have demonstrated that, as adjuvant therapy, letrozole has greater efficacy than tamoxifen. It is also more effective than placebo as extended adjuvant therapy after completion of tamoxifen therapy in these patients. In women with hormone-responsive advanced breast cancer, letrozole is superior to tamoxifen in prolonging the time to disease progression and time to treatment failure in a first-line setting, and is at least as effective as anastrozole and more effective than megestrol for some endpoints (in one of two trials) in a second-line setting. Letrozole is generally well tolerated, and in a health-related quality-of-life analysis from a large clinical trial, patient well-being with letrozole as extended adjuvant therapy did not differ from that with placebo. Modelled analyses from the UK and the US suggest that, in postmenopausal women with hormone-receptor-positive early-stage breast cancer, letrozole is likely to be a cost-effective alternative to tamoxifen as adjuvant therapy; moreover, using letrozole as extended adjuvant therapy after tamoxifen, rather than no further treatment, is also a cost-effective treatment strategy. Sensitivity analyses have shown these results to be robust. In terms of direct healthcare costs, pharmacoeconomic models suggest that letrozole is a cost-effective alternative to tamoxifen as first-line therapy in postmenopausal women with hormone-responsive advanced breast cancer from the perspectives of the UK NHS, the Canadian and Italian public healthcare systems and the Japanese national health insurance system. Incremental costs per QALY or progression-free year gained over tamoxifen were well within the recommended limits for acceptability of new agents that are more effective and more expensive than existing therapies in the UK, Japan and Canada. Modelled analyses from the UK and Canada have also suggested that letrozole is cost effective as second-line therapy for advanced breast cancer in postmenopausal women who have disease progression following anti-estrogen therapy. In conclusion, letrozole is an effective and well tolerated treatment for postmenopausal women with early-stage or advanced hormone-responsive breast cancer. Pharmacoeconomic analyses from UK and North American perspectives support the use of letrozole in hormone-responsive early-stage breast cancer in both the adjuvant and extended adjuvant settings. In addition, other modelled analyses conducted in a variety of healthcare systems across different countries consistently suggest that letrozole is cost effective in advanced treatment settings.
Collapse
|
25
|
Chow LWC, Loo WTY, Toi M. Current directions for COX-2 inhibition in breast cancer. Biomed Pharmacother 2005; 59 Suppl 2:S281-4. [PMID: 16507393 DOI: 10.1016/s0753-3322(05)80046-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Chemotherapy is effective against breast cancer. COX-2 has been implicated in the progression and angiogenesis of cancers. Celecoxib, a cyclooxygenase type 2 (COX-2) inhibitor, has both apoptotic and antiangiogenic activities, and may be of use in treatment of breast tumors which overexpress the COX-2 enzyme. Preliminary clinical trials have shown that the combination of chemotherapy with celecoxib has minimal additional toxicity and it may enhance the effects of the chemotherapy. Beside chemotherapy, celecoxib may promulgate the effect of aromatase inhibitor in breast cancer cells. Animal studies have shown that there are fewer and smaller tumors treated by combining exemestane and celecoxib. Larger clinical trials should be initiated to study the potential anti-cancer effects of celecoxib in breast cancer.
Collapse
Affiliation(s)
- L W C Chow
- Department Hung Chao Hong Integrated Center for Breast Diseases, University of Hong Kong Medical Center, Pokfulam, China.
| | | | | |
Collapse
|
26
|
Diesing D, Friedrich M, von Otte S. Nebenwirkungen der endokrinen Therapie beim Mammakarzinom. GYNAKOLOGISCHE ENDOKRINOLOGIE 2005. [DOI: 10.1007/s10304-005-0109-6] [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]
|
27
|
Lafky JM, Baron AT, Cora EM, Hillman DW, Suman VJ, Perez EA, Ingle JN, Maihle NJ. Serum Soluble Epidermal Growth Factor Receptor Concentrations Decrease in Postmenopausal Metastatic Breast Cancer Patients Treated with Letrozole. Cancer Res 2005; 65:3059-62. [PMID: 15833834 DOI: 10.1158/0008-5472.can-05-0067] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Previous studies have implicated estrogen as a regulator of epidermal growth factor receptor (EGFR) expression in breast tumors. We therefore speculated that estrogen might modulate serologic soluble EGFR (sEGFR) concentrations in breast cancer patients. Accordingly, we measured serum sEGFR concentrations in postmenopausal women with metastatic breast cancer (MBC) treated with letrozole, an aromatase inhibitor that blocks estrogen synthesis. Serum specimens were obtained prior to and following 1 and 3 months of letrozole therapy. We report that sEGFR concentrations do not differ between MBC patients prior to letrozole treatment and age- and postmenopause-matched healthy women (P = 0.468). In contrast, however, sEGFR concentrations decreased significantly in 76% of MBC patients after both 1 month (P = 0.006) and 3 months (P = 0.003) of letrozole therapy versus pretreatment concentrations. Within the limitations of this study, we found no evidence for an association between pretreatment sEGFR concentrations or decreased treatment sEGFR concentrations and either progression-free or overall survival. Nonetheless, we conclude that future prospective studies are warranted to determine if baseline and/or longitudinal serum sEGFR concentrations may be useful for predicting disease progression and survival, and/or for monitoring responsiveness to aromatase inhibitors or other endocrine therapies in breast cancer patients.
Collapse
Affiliation(s)
- Jacqueline M Lafky
- Tumor Biology Program, Cancer Center Statistics Unit, Department of Health Sciences Research, Mayo Clinic College of Medicine, Rochester, MN, USA
| | | | | | | | | | | | | | | |
Collapse
|