1
|
García-Sánchez J, Mafla-España MA, Torregrosa MD, Cauli O. Androstenedione and Follicle-Stimulating Hormone Concentration Predict the Progression of Frailty Syndrome at One Year Follow-Up in Patients with Localized Breast Cancer Treated with Aromatase Inhibitors. Biomedicines 2022; 10:biomedicines10071634. [PMID: 35884939 PMCID: PMC9312841 DOI: 10.3390/biomedicines10071634] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/05/2022] [Accepted: 07/05/2022] [Indexed: 11/16/2022] Open
Abstract
Background: The standard treatment in postmenopausal women with estrogen- and progesterone-positive localized breast cancer consists of aromatase inhibitors (AROi). The ability of AROi to promote or worsen frailty syndrome over time and the relationship with changes in gonadal hormones concentration in blood have not been investigated. Methods: A prospective study to evaluate the relationship between frailty syndrome and gonadal hormones concentrations in blood at baseline (prior to AROi treatment) and after 6 and 12 months under AROi treatment in post-menopausal women with breast cancer. Frailty syndrome was evaluated by the Fried’ criteria. We evaluated whether hormone concentration at baseline could predict frailty syndrome at follow-up. Results: Multinomial regression analysis showed that of the different hormones, those significantly (p < 0.05) associated to the worsening of frailty syndrome were high androstenedione levels and low follicle-stimulating hormone (FSH) levels in blood. Receiver operating characteristic curve analysis showed both androstenedione and FSH significantly (p < 0.05) discriminate patients who developed or presented worsening of frailty syndrome over time, with acceptable sensitivity (approximately 80% in both cases) but low specificity (40%). Conclusion: Hormonal concentrations before AROi treatment constitute possible biomarkers to predict the progression of frailty syndrome.
Collapse
Affiliation(s)
- Javier García-Sánchez
- Medical Oncology Department, Doctor Peset University Hospital, 46017 Valencia, Spain; (J.G.-S.); (M.D.T.)
- Medical Oncology Department, Hospital Center of Wallonie Picardy, 7500 Tournai, Belgium
| | - Mayra Alejandra Mafla-España
- Frailty Research Organized Group, University of Valencia, 46010 Valencia, Spain;
- Department of Nursing, University of Valencia, 46010 Valencia, Spain
| | - María Dolores Torregrosa
- Medical Oncology Department, Doctor Peset University Hospital, 46017 Valencia, Spain; (J.G.-S.); (M.D.T.)
| | - Omar Cauli
- Frailty Research Organized Group, University of Valencia, 46010 Valencia, Spain;
- Department of Nursing, University of Valencia, 46010 Valencia, Spain
- Correspondence:
| |
Collapse
|
2
|
van Dijk M, ter Laak AM, Wichard JD, Capoferri L, Vermeulen NPE, Geerke DP. Comprehensive and Automated Linear Interaction Energy Based Binding-Affinity Prediction for Multifarious Cytochrome P450 Aromatase Inhibitors. J Chem Inf Model 2017; 57:2294-2308. [PMID: 28776988 PMCID: PMC5615371 DOI: 10.1021/acs.jcim.7b00222] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Indexed: 11/30/2022]
Abstract
Cytochrome P450 aromatase (CYP19A1) plays a key role in the development of estrogen dependent breast cancer, and aromatase inhibitors have been at the front line of treatment for the past three decades. The development of potent, selective and safer inhibitors is ongoing with in silico screening methods playing a more prominent role in the search for promising lead compounds in bioactivity-relevant chemical space. Here we present a set of comprehensive binding affinity prediction models for CYP19A1 using our automated Linear Interaction Energy (LIE) based workflow on a set of 132 putative and structurally diverse aromatase inhibitors obtained from a typical industrial screening study. We extended the workflow with machine learning methods to automatically cluster training and test compounds in order to maximize the number of explained compounds in one or more predictive LIE models. The method uses protein-ligand interaction profiles obtained from Molecular Dynamics (MD) trajectories to help model search and define the applicability domain of the resolved models. Our method was successful in accounting for 86% of the data set in 3 robust models that show high correlation between calculated and observed values for ligand-binding free energies (RMSE < 2.5 kJ mol-1), with good cross-validation statistics.
Collapse
Affiliation(s)
- Marc van Dijk
- AIMMS
Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical
Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | | | - Jörg D. Wichard
- Bayer AG, Pharmaceuticals Division, Müllerstrasse
178, D-13353 Berlin, Germany
| | - Luigi Capoferri
- AIMMS
Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical
Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Nico P. E. Vermeulen
- AIMMS
Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical
Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| | - Daan P. Geerke
- AIMMS
Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical
Sciences, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
3
|
Simsa P, Mihalyi A, Kyama CM, Mwenda JM, Fülöp V, D'Hooghe TM. Selective Estrogen-Receptor Modulators and Aromatase Inhibitors: Promising New Medical Therapies for Endometriosis? WOMENS HEALTH 2016; 3:617-28. [DOI: 10.2217/17455057.3.5.617] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Endometriosis is an estrogen-dependent disease and estrogen-related pathways are imbalanced in women with endometriosis. One of the key enzymes in estrogen synthesis is aromatase. Inhibiting this pathway at several points is a promising idea for the treatment of endometriosis. The third generation of aromatase inhibitors is becoming more potent in efficacy, with fewer side effects than previous generations, but cotreatment with other hormones is needed to inhibit ovarian stimulation. Other components that promote estrogen synthesis such as COX-2 can also be potentially targeted. Selective estrogen-receptor modulators could also be interesting in view of their tissue-specific effect. However, all these new drugs are still in an early phase of development. At present, it is too early to conclude that aromatase inhibitors, COX-2 inhibitors or selective estrogen-receptor modulators really present any added value compared with the existing drugs that can be used to achieve hormonal suppression in the medical treatment of endometriosis.
Collapse
Affiliation(s)
- Peter Simsa
- Leuven University Fertility Centre, Department of Obstetrics & Gynaecology, University Hospital Gasthuisberg, Leuven, Belgium, Tel: +32 1634 3624; Fax: +32 1634 3607
- National Institute of Health, Budapest, Hungary
| | - Attila Mihalyi
- Leuven University Fertility Centre, Department of Obstetrics & Gynaecology, University Hospital Gasthuisberg, Leuven, Belgium, Tel: +32 1634 3624; Fax: +32 1634 3607
| | - Cleophas M Kyama
- Leuven University Fertility Centre, Department of Obstetrics & Gynaecology, University Hospital Gasthuisberg, Leuven, Belgium, Tel: +32 1634 3624; Fax: +32 1634 3607
- Institute of Primate Research, Division of Reproduction, PO Box 24481, Karen, Nairobi, Kenya
| | | | | | - Thomas M D'Hooghe
- Leuven University Fertility Centre, Department of Obstetrics & Gynaecology, University Hospital Gasthuisberg, Leuven, Belgium, Tel: +32 1634 3624; Fax: +32 1634 3607
- Institute of Primate Research, Division of Reproduction, PO Box 24481, Karen, Nairobi, Kenya
| |
Collapse
|
4
|
Modulation of Aromatase by Phytoestrogens. Enzyme Res 2015; 2015:594656. [PMID: 26798508 PMCID: PMC4699002 DOI: 10.1155/2015/594656] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 10/31/2015] [Accepted: 11/17/2015] [Indexed: 12/29/2022] Open
Abstract
The aromatase enzyme catalyzes the conversion of androgens to estrogens in many human tissues. Estrogens are known to stimulate cellular proliferation associated with certain cancers and protect against adverse symptoms during the peri- and postmenopausal intervals. Phytoestrogens are a group of plant derived naturally occurring compounds that have chemical structures similar to estrogen. Since phytoestrogens are known to be constituents of animal/human food sources, these compounds have received increased research attention. Phytoestrogens may contribute to decreased cancer risk by the inhibition of aromatase enzyme activity and CYP19 gene expression in human tissues. This review covers (a) the aromatase enzyme (historical descriptions on function, activity, and gene characteristics), (b) phytoestrogens in their classifications and applications to human health, and (c) a chronological coverage of aromatase activity modulated by phytoestrogens from the early 1980s to 2015. In general, phytoestrogens act as aromatase inhibitors by (a) decreasing aromatase gene expression, (b) inhibiting the aromatase enzyme itself, or (c) in some cases acting at both levels of regulation. The findings presented herein are consistent with estrogen's impact on health and phytoestrogen's potential as anticancer treatments, but well-controlled, large-scale studies are warranted to determine the effectiveness of phytoestrogens on breast cancer and age-related diseases.
Collapse
|
5
|
Pandya K, Wyatt D, Gallagher B, Shah D, Baker A, Bloodworth J, Zlobin A, Pannuti A, Green A, Ellis IO, Filipovic A, Sagert J, Rana A, Albain KS, Miele L, Denning MF, Osipo C. PKCα Attenuates Jagged-1-Mediated Notch Signaling in ErbB-2-Positive Breast Cancer to Reverse Trastuzumab Resistance. Clin Cancer Res 2015; 22:175-86. [PMID: 26350262 DOI: 10.1158/1078-0432.ccr-15-0179] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 08/25/2015] [Indexed: 02/06/2023]
Abstract
PURPOSE Breast cancer is the second leading cause of cancer mortality among women worldwide. The major problem with current treatments is tumor resistance, recurrence, and disease progression. ErbB-2-positive breast tumors are aggressive and frequently become resistant to trastuzumab or lapatinib. We showed previously that Notch-1 is required for trastuzumab resistance in ErbB-2-positive breast cancer. EXPERIMENTAL DESIGN Here, we sought to elucidate mechanisms by which ErbB-2 attenuates Notch signaling and how this is reversed by trastuzumab or lapatinib. RESULTS The current study elucidates a novel Notch inhibitory mechanism by which PKCα downstream of ErbB-2 (i) restricts the availability of Jagged-1 at the cell surface to transactivate Notch, (ii) restricts the critical interaction between Jagged-1 and Mindbomb-1, an E3 ligase that is required for Jagged-1 ubiquitinylation and subsequent Notch activation, (iii) reverses trastuzumab resistance in vivo, and (iv) predicts better outcome in women with ErbB-2-positive breast cancer. CONCLUSIONS The clinical impact of these studies is PKCα is potentially a good prognostic marker for low Notch activity and increased trastuzumab sensitivity in ErbB-2-positive breast cancer. Moreover, women with ErbB-2-positive breast tumors expressing high Notch activation and low PKCα expression could be the best candidates for anti-Notch therapy.
Collapse
Affiliation(s)
- Kinnari Pandya
- Molecular Biology Program, Louisiana State University, New Orleans, Louisiana
| | - Debra Wyatt
- Oncology Research Institute, Louisiana State University, New Orleans, Louisiana
| | - Brian Gallagher
- Oncology Research Institute, Louisiana State University, New Orleans, Louisiana
| | - Deep Shah
- Department of Molecular Pharmacology and Therapeutics, Louisiana State University, New Orleans, Louisiana
| | - Andrew Baker
- Integrated Cell Biology Program, Louisiana State University, New Orleans, Louisiana
| | - Jeffrey Bloodworth
- Molecular Biology Program, Louisiana State University, New Orleans, Louisiana
| | - Andrei Zlobin
- Oncology Research Institute, Louisiana State University, New Orleans, Louisiana
| | | | - Andrew Green
- Department of Histopathology, University of Nottingham and University Hospital NHS Trust, Nottingham, United Kingdom
| | - Ian O Ellis
- Department of Histopathology, University of Nottingham and University Hospital NHS Trust, Nottingham, United Kingdom
| | | | | | - Ajay Rana
- Department of Molecular Pharmacology and Therapeutics, Louisiana State University, New Orleans, Louisiana
| | - Kathy S Albain
- Department of Medicine/Hematology and Oncology, Cardinal Bernardin Cancer Center of Loyola University Chicago: Health Sciences Division, Maywood, Illinois
| | - Lucio Miele
- Louisiana State University, New Orleans, Louisiana
| | - Mitchell F Denning
- Oncology Research Institute, Louisiana State University, New Orleans, Louisiana. Department of Pathology, Cardinal Bernardin Cancer Center of Loyola University Chicago: Health Sciences Division, Maywood, Illinois
| | - Clodia Osipo
- Oncology Research Institute, Louisiana State University, New Orleans, Louisiana. Department of Pathology, Cardinal Bernardin Cancer Center of Loyola University Chicago: Health Sciences Division, Maywood, Illinois. Department of Microbiology and Immunology, Cardinal Bernardin Cancer Center of Loyola University Chicago: Health Sciences Division, Maywood, Illinois.
| |
Collapse
|
6
|
Young NA, Friedman AK, Kaffenberger B, Rajaram MVS, Birmingham DJ, Rovin BH, Hebert LA, Schlesinger LS, Wu LC, Jarjour WN. Novel estrogen target gene ZAS3 is overexpressed in systemic lupus erythematosus. Mol Immunol 2012. [PMID: 23178823 DOI: 10.1016/j.molimm.2012.10.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Systemic lupus erythematosus (SLE) is a prototypic, inflammatory autoimmune disease characterized by significant gender bias. Previous studies have established a role for hormones in SLE pathogenesis, including the sex hormone estrogen. Estrogen regulates gene expression by translocating estrogen receptors (ER) α and β into the nucleus where they induce transcription by binding to estrogen response elements (EREs) of target genes. The ZAS3 locus encodes a signaling and transcriptional molecule involved in regulating inflammatory responses. We show that ZAS3 is significantly up-regulated in SLE patients at both the protein and mRNA levels in peripheral blood mononuclear cells (PBMCs). Furthermore, estrogen stimulates the expression of ZAS3 in vitro in several leukocyte and breast cancer cell lines of both human and murine origin. In vivo estrogen treatment mediates induction of tissue specific ZAS3 expression in several lymphoid organs in mice. Estrogen stimulation also significantly up-regulates ZAS3 expression in primary PBMCs, while treatment with testosterone has no effect. Mechanistically, estrogen induces differential ERα binding to putative EREs within the ZAS3 gene and ERα knockdown with siRNA prevents estrogen induced ZAS3 up-regulation. In contrast, siRNA targeting IFNα has no effect. These data demonstrate that ZAS3 expression is directly regulated by estrogen and that ZAS3 is overexpressed in lupus. Since ZAS3 has been shown to regulate inflammatory pathways, its up-regulation by estrogen could play a critical role in female-biased autoimmune disorders.
Collapse
Affiliation(s)
- Nicholas A Young
- Division of Rheumatology and Immunology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Noh YH, Ko YJ, Cho SH, Ghim JL, Choe S, Jung JA, Kim UJ, Jin SJ, Park HJ, Song GS, Lim HS, Bae KS. Pharmacokinetic Comparison of 2 Formulations of Anastrozole (1 mg) in Healthy Korean Male Volunteers: A Randomized, Single-Dose, 2-Period, 2-Sequence, Crossover Study. Clin Ther 2012; 34:305-13. [DOI: 10.1016/j.clinthera.2012.01.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Revised: 12/13/2011] [Accepted: 01/03/2012] [Indexed: 11/26/2022]
|
8
|
Punetha A, Shanmugam K, Sundar D. Insight into the Enzyme-Inhibitor Interactions of the First Experimentally Determined Human Aromatase. J Biomol Struct Dyn 2011; 28:759-71. [DOI: 10.1080/07391102.2011.10508604] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
9
|
Chen JQ, Yager JD, Russo J. Regulation of mitochondrial respiratory chain structure and function by estrogens/estrogen receptors and potential physiological/pathophysiological implications. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2005; 1746:1-17. [PMID: 16169101 DOI: 10.1016/j.bbamcr.2005.08.001] [Citation(s) in RCA: 109] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2005] [Revised: 08/03/2005] [Accepted: 08/03/2005] [Indexed: 01/11/2023]
Abstract
It is well known that the biological and carcinogenic effects of 17beta-estradiol (E2) are mediated via nuclear estrogen receptors (ERs) by regulating nuclear gene expression. Several rapid, non-nuclear genomic effects of E2 are mediated via plasma membrane-bound ERs. In addition, there is accumulating evidence suggesting that mitochondria are also important targets for the action of estrogens and ERs. This review summarized the studies on the effects of estrogens via ERs on mitochondrial structure and function. The potential physiological and pathophysiological implications of deficiency and/or overabundance of these E2/ER-mediated mitochondrial effects in stimulation of cell proliferation, inhibition of apoptosis, E2-mediated cardiovascular and neuroprotective effects in target cells are also discussed.
Collapse
Affiliation(s)
- Jin-Qiang Chen
- Breast Cancer Research Laboratory, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111, USA.
| | | | | |
Collapse
|
10
|
Compounds for the treatment of oestrogen-dependent illnesses. Expert Opin Ther Pat 2005. [DOI: 10.1517/13543776.10.11.1771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
11
|
Zaidman BZ, Yassin M, Mahajna J, Wasser SP. Medicinal mushroom modulators of molecular targets as cancer therapeutics. Appl Microbiol Biotechnol 2005; 67:453-68. [PMID: 15726350 DOI: 10.1007/s00253-004-1787-z] [Citation(s) in RCA: 244] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2004] [Revised: 09/27/2004] [Accepted: 10/02/2004] [Indexed: 02/07/2023]
Abstract
Empirical approaches to discover anticancer drugs and cancer treatments have made limited progress in the past several decades in finding a cure for cancer. The expanded knowledge of the molecular basis of tumorigenesis and metastasis, together with the inherently vast structural diversity of natural compounds found in mushrooms, provided unique opportunities for discovering new drugs that rationally target the abnormal molecular and biochemical signals leading to cancer. This review focuses on mushroom low-molecular-weight secondary metabolites targeting processes such as apoptosis, angiogenesis, metastasis, cell cycle regulation, and signal transduction cascades. Also discussed in this review are high-molecular-weight polysaccharides or polysaccharide-protein complexes from mushrooms that appear to enhance innate and cell-mediated immune responses, exhibit antitumor activities in animals and humans, and demonstrate the anticancer properties of selenium compounds accumulated in mushrooms.
Collapse
Affiliation(s)
- Ben-Zion Zaidman
- Biodiversity and Biotechnology Center of Cryptogamic Plants and Fungi, The Institute of Evolution, University of Haifa, Mount Carmel, Israel
| | | | | | | |
Collapse
|
12
|
Abstract
In premenopausal women ovaries are the major sites of estrogen production, while in postmenopausal women estrogen is produced by aromatization of ovarian and adrenal androgens in extragonadal sites, mostly in adipose tissue. Aromatase is a cytochrome P450 hemoprotein-containing enzyme complex that catalyzes the rate-limiting step in the conversion of androstenedione and testosterone to estrone and estradiol (E2). Aromatase inhibitors (AIs) have been developed primarily for use in either natural or surgical postmenopausal patients. In premenopausal women, the ovary can overcome the estrogen blockade by reflex increments of luteinizing hormone (LH) and follicle stimulating hormone (FSH), so AIs must be combined with a gonadotropin releasing hormone (GnRH) agonist to prevent the reflex LH and FSH increments. In advanced hormone-dependent breast cancer treatment, AIs have been shown to be superior to tamoxifen. Preliminary evidence also suggests superiority in the adjuvant, neoadjuvant settings and also for breast cancer prevention. AIs have been used in infertility and can increase ovulation rate. Reducing FSH dose, estrogen levels, improving response to FSH, implantation rates, and developing multiple follicles that can be used in in vitro maturation procedures are potential areas that AIs might be used in in assisted reproductive technologies (ART), besides simple ovulation induction. AIs are reported to be successful in treatment of endometriosis, an estrogen-dependent process. The use of AIs in gynecomastia, puberte precox, leiomyoma uteri, some estrogen-dependent cancers (ovarian), endometrial cancer and male infertility are reported; some of the results are promising but more clinical trials are needed. AIs are predicted to become the gold standard in the treatment of estrogen-dependent diseases in reproductive medicine in the near future.
Collapse
Affiliation(s)
- Oznur Karaer
- Department of Obstetrics and Gynecology, Celal Bayar University Faculty of Medicine, Manisa, Turkey.
| | | | | |
Collapse
|
13
|
Howell A, Robertson JFR, Vergote I. A Review of the Efficacy of Anastrozole in Postmenopausal Women with Advanced Breast Cancer with Visceral Metastases. Breast Cancer Res Treat 2003; 82:215-22. [PMID: 14703069 DOI: 10.1023/b:brea.0000004375.17920.0b] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Tumors that have spread to the liver or lungs (visceral metastases) are associated with a worse prognosis than tumors in soft tissue and bone only. Here we review available efficacy data to address whether or not anastrozole, a non-steroidal aromatase inhibitor (AI), is effective in postmenopausal patients with advanced breast cancer (ABC) and visceral metastases. We include data from Phase III clinical trials, comparing clinical benefit (CB) with anastrozole versus tamoxifen as a first-line treatment, and versus megestrol acetate (MA) or fulvestrant as a second-line therapy. Patients in these trials had adequate organ function and the volume of disease had to be minimal or moderate for them to be eligible for inclusion. First-line treatment of patients with or without visceral metastases in the overall population resulted in CB rates of 49.5 and 62.3%, respectively, for anastrozole and 46.9 and 55.9%, respectively, for tamoxifen. In patients with confirmed hormone receptor (HR)-positive tumors, the CB rate was 51.9 and 65.7%, respectively, for anastrozole and 41.6 and 58.7%, respectively, for tamoxifen. In patients with or without visceral metastases, second-line treatment with anastrozole resulted in a CB rate of 31.4 and 51.8%, respectively, compared with 31.9 and 47.1%, respectively, for those treated with MA. Patients in the overall population with and without visceral metastases treated with anastrozole obtained a CB rate of 37.4 and 43.8%, respectively, while those treated with fulvestrant obtained a CB rate of 38.2 and 47.6%, respectively. In patients with confirmed HR-positive tumors, CB was seen in 37.6 and 41.5%, respectively, of patients treated with anastrozole and in 37.3 and 47.0%, respectively, of patients treated with fulvestrant. The results reveal anastrozole to be an effective and valuable first- and second-line therapy in postmenopausal women with ABC and visceral metastases, showing similar CB to other endocrine therapies.
Collapse
Affiliation(s)
- A Howell
- Cancer Research UK Department of Medical Oncology, Christie CRC Research Hospital, Christie Hospital National Health Service Trust, University of Manchester, Manchester, UK.
| | | | | |
Collapse
|
14
|
Schmid M, Jakesz R, Samonigg H, Kubista E, Gnant M, Menzel C, Seifert M, Haider K, Taucher S, Mlineritsch B, Steindorfer P, Kwasny W, Stierer M, Tausch C, Fridrik M, Wette V, Steger G, Hausmaninger H. Randomized trial of tamoxifen versus tamoxifen plus aminoglutethimide as adjuvant treatment in postmenopausal breast cancer patients with hormone receptor-positive disease: Austrian breast and colorectal cancer study group trial 6. J Clin Oncol 2003; 21:984-90. [PMID: 12637461 DOI: 10.1200/jco.2003.01.138] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To determine whether the addition of aminoglutethimide to tamoxifen is able to improve the outcome in postmenopausal patients with hormone receptor-positive, early-stage breast cancer. PATIENTS AND METHODS A total of 2,021 postmenopausal women were randomly assigned to receive either tamoxifen for 5 years alone or tamoxifen in combination with aminoglutethimide (500 mg/d) for the first 2 years of treatment. Tamoxifen was administered at 40 mg/d for the first 2 years and at 20 mg/d for 3 years. RESULTS All randomized and eligible patients were included in the analysis according to the intention-to-treat principle. After a median follow-up of 5.3 years, the 5-year disease-free survival in the aminoglutethimide plus tamoxifen group was 83.6% versus 83.7% in the monotherapy group (P =.89). The corresponding data for overall survival at 5 years were 91.4% and 91.2%, respectively (P =.74). More patients failed to complete combination treatment (13.7%) because of side effects as compared to tamoxifen alone (5.2%; P =.0001). CONCLUSION Aminoglutethimide given for 2 years in addition to tamoxifen for 5 years does not improve the prognosis of postmenopausal patients with receptor-positive, lymph node-negative or lymph node-positive breast cancer.
Collapse
Affiliation(s)
- Marianne Schmid
- Medical Department, Graz University, and Second Department of Surgery, Graz Hospital, Graz, Austria.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Affiliation(s)
- A Goldhirsch
- Department of Medicine, European Institute of Oncology, Milan, Italy
| | | | | |
Collapse
|
16
|
Buzdar AU, Robertson JFR, Eiermann W, Nabholtz JM. An overview of the pharmacology and pharmacokinetics of the newer generation aromatase inhibitors anastrozole, letrozole, and exemestane. Cancer 2002; 95:2006-16. [PMID: 12404296 DOI: 10.1002/cncr.10908] [Citation(s) in RCA: 176] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND The newer generation, nonsteroidal aromatase inhibitors (AIs) anastrozole and letrozole have shown superior efficacy compared with tamoxifen as first-line treatments and compared with megestrol acetate as second-line therapy in postmenopausal women with advanced breast carcinoma. In an open-label, Phase II trial, it was reported that exemestane showed numerical superiority compared with tamoxifen for objective response and clinical benefit. Because these agents ultimately may be administered for periods of up to 5 years in the adjuvant setting, it is of increasing importance to assess their tolerability and pharmacologic profiles. METHODS In the absence of data from direct clinical comparisons, the published literature was reviewed for the clinical pharmacology, pharmacokinetic characteristics, and selectivity profiles of anastrozole, letrozole, and exemestane. RESULTS At clinically administered doses, the plasma half-lives of anastrozole (1 mg once daily), letrozole (2.5 mg once daily), and exemestane (25 mg once daily) were 41-48 hours, 2-4 days, and 27 hours, respectively. The time to steady-state plasma levels was 7 days for both anastrozole and exemestane and 60 days for letrozole. Androgenic side effects have been reported only with exemestane. Anastrozole treatment had no impact on plasma lipid levels, whereas both letrozole and exemestane had an unfavorable effect on plasma lipid levels. In indirect comparisons, anastrozole showed the highest degree of selectivity compared with letrozole and exemestane in terms of a lack of effect on adrenosteroidogenesis. CONCLUSIONS All three AIs demonstrated clinical efficacy over preexisting treatments. However, there were differences in terms of pharmacokinetics and effects on lipid levels and adrenosteroidogenesis. The long-term clinical significance of these differences remains to be elucidated.
Collapse
Affiliation(s)
- Aman U Buzdar
- Department of Breast Medical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston 77030, USA.
| | | | | | | |
Collapse
|
17
|
Abstract
Systemic adjuvant therapy is recommended immediately following surgical removal of the primary tumour in the majority of patients with early breast cancer, to prevent the recurrence of distant metastases. Significant progress has been made in the development and evaluation of endocrine therapies for systemic adjuvant therapy. In pre- and perimenopausal women, ovarian ablation has proven to be a valuable treatment option, though not always desirable for young patients. Thus, reversible medical ovarian suppression with a luteinizing hormone releasing hormone agonist, such as goserelin (Zoladex), may provide an attractive alternative for such patients. International trials have indicated that goserelin provides an important addition to the choice of adjuvant therapies now available to pre- and perimenopausal patients. For postmenopausal patients, it is hoped that the ATAC (Arimidex, tamoxifen, alone or in combination) trial will reveal whether or not the benefits of anastrozole (Arimidex) observed in advanced disease, where it has proven to be well tolerated and at least as effective as tamoxifen in recent trials, will translate to the early setting to provide further management options for these patients. On the horizon is yet another exciting endocrine agent, ICI 182,780 (Fulvestrant), which has also been shown to be as effective as anastrozole in advanced disease. In terms of the future, these agents are likely to provide additional valuable treatment choices for early breast cancer across the patient spectrum.
Collapse
Affiliation(s)
- M Baum
- Department of Psycho-oncology, University College London Medical School, UK
| |
Collapse
|
18
|
Harper-Wynne C, Coombes RC. Anastrozole shows evidence of activity in postmenopausal patients who have responded or stabilised on formestane therapy. Eur J Cancer 1999; 35:744-6. [PMID: 10505035 DOI: 10.1016/s0959-8049(99)00015-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Formestane (Lentaron) and anastrozole (Arimidex) are in clinical use as second-line treatments for advanced breast cancer. Current practice is often to use an aromatase inhibitor only once before switching to third-line agents such as progestins. There are few clinical data on the sequential use of aromatase inhibitors. We therefore decided to study the clinical effects of anastrozole in postmenopausal patients with advanced breast cancer who had already received formestane. 21 patients were recruited. When receiving formestane 2/21 (10%) achieved a partial response (UICC criteria) and 10/21 (48%) stable disease. Of these 12 patients, 9 achieved further stable disease on anastrozole (78%; 7/9 oestrogen receptor positive). 4 of 9 patients who progressed on formestane also stabilised on anastrozole, of whom 3 had oestrogen receptor positive breast carcinomas. The explanation of this second stabilisation may relate to a further fall in oestradiol levels. We feel these results are of interest and warrant further clinical investigation.
Collapse
Affiliation(s)
- C Harper-Wynne
- Department of Cancer Medicine, Imperial College School of Medicine, Charing Cross Hospital, London, U.K
| | | |
Collapse
|
19
|
Miller W, Telford J, Love C, Leonard R, Hillier S, Gundacker H, Smith H, Dixon J. Effects of letrozole as primary medical therapy on in situ oestrogen synthesis and endogenous oestrogen levels within the breast. Breast 1998. [DOI: 10.1016/s0960-9776(98)90095-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
20
|
Affiliation(s)
- G Y Locker
- Evanston Northwestern Healthcare, Northwestern University, IL, USA
| |
Collapse
|
21
|
Abstract
Surgeons have been involved in the management of metastatic breast cancer since the technique of ovarian ablation was introduced in 1896. However, as newer hormonal and chemotherapeutic regimens were developed, drug therapy gradually replaced surgery as the preferred treatment for metastatic breast cancer. Thus, management of metastatic breast cancer has largely shifted from surgeons to medical oncologists. Advances in hormonal pharmacology have placed hormonal therapy alongside surgery and radiation therapy as a standard treatment option for women with advanced breast cancer. The purpose of this article is to update surgeons on the current use of hormonal agents for treatment of advanced breast cancer in postmenopausal women, and to review the aromatase inhibitors, a new line of hormonal agents for the treatment of advanced breast cancer.
Collapse
Affiliation(s)
- B J Roseman
- Department of Surgical Oncology, University of Texas M.D. Anderson Cancer Center, Houston 77030, USA
| | | | | |
Collapse
|
22
|
Geisler J, King N, Dowsett M, Ottestad L, Lundgren S, Walton P, Kormeset PO, Lønning PE. Influence of anastrozole (Arimidex), a selective, non-steroidal aromatase inhibitor, on in vivo aromatisation and plasma oestrogen levels in postmenopausal women with breast cancer. Br J Cancer 1996; 74:1286-91. [PMID: 8883419 PMCID: PMC2075919 DOI: 10.1038/bjc.1996.531] [Citation(s) in RCA: 249] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The effect of anastrozole ('Arimidex', ZD1033), a new, selective, non-steroidal aromatase inhibitor on in vivo aromatisation and plasma oestrogen levels was evaluated in post-menopausal women with breast cancer. Twelve patients progressing after treatment with tamoxifen were randomised to receive anastrozole 1 mg or 10 mg once daily for a 28 day period in a double-blinded crossover design. In vivo aromatisation and plasma oestrogen levels were determined before commencing treatment and at the end of each 4-week period. Treatment with anastrozole 1 and 10 mg reduced the percentage aromatisation from 2.25% to 0.074% and 0.043% (mean suppression of 96.7% and 98.1% from baseline) and suppressed plasma levels of oestrone, oestradiol and oestrone sulphate by > or = 86.5%, > or = 83.5% and > or = 93.5% respectively, irrespective of dose. Notably, several patients had their oestrone and oestradiol values suppressed beneath the sensitivity limit of the assays. In conclusion, anastrozole was found to be highly effective in inhibiting in vivo aromatisation with no difference in efficacy between the two drug doses. Contrary to previous studies on other aromatase inhibitors, this study revealed an internal consistency between the percentage aromatase inhibition and suppression of plasma oestrone sulphate.
Collapse
Affiliation(s)
- J Geisler
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Affiliation(s)
- W R Miller
- University Department of Clinical Oncology, Western General Hospital, Edinburgh, UK
| |
Collapse
|
24
|
Klijn JG, Setyono-Han B, Bontenbal M, Seynaeve C, Foekens J. Novel endocrine therapies in breast cancer. Acta Oncol 1996; 35 Suppl 5:30-7. [PMID: 9142962 DOI: 10.3109/02841869609083965] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Endocrine therapy of breast cancer consists of a variety of both medical and surgical ablative treatment modalities, but ablative therapy is increasingly replaced by medical treatment. Most endocrine therapies have more than one endocrine effect, frequently together with direct growth inhibitory actions via receptors. Endocrine therapy can be effective in all phases of the disease, but curative only in early disease while in advanced cancer it can only prolong survival. In the past decade the number of available endocrine agents has been drastically increased. Novel approaches in the endocrine therapy of breast cancer are application of new antiestrogens, antiprogestins, new potent aromatase inhibitors, analogues of luteinizing hormone-releasing hormone (LHRH-A) and somatostatin, inhibitors of prolactin secretion, vitamin A and D analogues, bisphosphonates, growth factor antagonists, tyrosine protein kinase inhibitors, protease inhibitors, inhibitors of angiogenesis, radiolabeled hormones and monoclonal antibodies. New cell biological factors such as oncogenes and suppressorgenes, secretory proteins and membrane receptors can be used not only as prognostic factors but also for prediction of type of response to endocrine and chemotherapy. Thus, these cell biological parameters can be used to select high and low risk patients, type of systemic treatment, and can also be used as targets for new treatment modalities. Future studies on treatment of all stages of disease will increasingly focus on promising combined treatment modalities.
Collapse
Affiliation(s)
- J G Klijn
- Department of Medical Oncology, Rotterdam Cancer Institute, Dr Daniel den Hoed Kliniek), The Netherlands
| | | | | | | | | |
Collapse
|