1
|
Yin G, Song G, Xue S, Liu F. Adverse event signal mining and serious adverse event influencing factor analysis of fulvestrant based on FAERS database. Sci Rep 2024; 14:11367. [PMID: 38762547 PMCID: PMC11102440 DOI: 10.1038/s41598-024-62238-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 05/15/2024] [Indexed: 05/20/2024] Open
Abstract
Fulvestrant, as the first selective estrogen receptor degrader, is widely used in the endocrine treatment of breast cancer. However, in the real world, there is a lack of relevant reports on adverse reaction data mining for fulvestrant. To perform data mining on adverse events (AEs) associated with fulvestrant and explore the risk factors contributing to severe AEs, providing a reference for the rational use of fulvestrant in clinical practice. Retrieved adverse event report information associated with fulvestrant from the U.S. Food and Drug Administration (FDA) Adverse Event Reporting System (FAERS) database, covering the period from market introduction to September 30, 2023. Suspicious AEs were screened using the reporting odds ratio (ROR) and proportional reporting ratio methods based on disproportionality analysis. Univariate and multivariate logistic regression analyses were conducted on severe AEs to explore the risk factors associated with fulvestrant-induced severe AEs. A total of 6947 reports related to AEs associated with fulvestrant were obtained, including 5924 reports of severe AEs and 1023 reports of non-severe AEs. Using the disproportionality analysis method, a total of 210 valid AEs were identified for fulvestrant, with 45 AEs (21.43%) not listed in the product labeling, involving 11 systems and organs. The AEs associated with fulvestrant were sorted by frequency of occurrence, with neutropenia (325 cases) having the highest number of reports. By signal strength, injection site pruritus showed the strongest signal (ROR = 658.43). The results of the logistic regression analysis showed that concurrent use of medications with extremely high protein binding (≥ 98%) is an independent risk factor for severe AEs associated with fulvestrant. Age served as a protective factor for fulvestrant-related AEs. The co-administration of fulvestrant with CYP3A4 enzyme inhibitors did not show statistically significant correlation with the occurrence of severe AEs. Co-administration of drugs with extremely high protein binding (≥ 98%) may increase the risk of severe adverse reactions of fulvestrant. Meanwhile, age (60-74 years) may reduce the risk of severe AEs of fulvestrant. However, further clinical research is still needed to explore and verify whether there is interaction between fulvestrant and drugs with high protein binding through more clinical studies.
Collapse
Affiliation(s)
- Guisen Yin
- Department of Pharmacy, Yantai Hospital of Traditional Chinese Medicine, YantaiShandong, 264000, China
| | - Guiling Song
- Department of Chemical Medicine, Yantai Center for Food and Drug Control, YantaiShandong, 264003, China
| | - Shuyi Xue
- Department of Pharmacy, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, 266042, Shandong, China
| | - Fen Liu
- Department of Pharmacy, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, 410011, Hunan, China.
| |
Collapse
|
2
|
Schwartz G, Shee K, Romo B, Marotti J, Kisselev A, Lewis L, Miller T. Phase Ib Study of the Oral Proteasome Inhibitor Ixazomib (MLN9708) and Fulvestrant in Advanced ER+ Breast Cancer Progressing on Fulvestrant. Oncologist 2021; 26:467-e924. [PMID: 33641211 PMCID: PMC8176977 DOI: 10.1002/onco.13733] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 02/10/2021] [Indexed: 12/28/2022] Open
Abstract
LESSONS LEARNED Fulvestrant is a selective estrogen receptor (ER)-downregulating antiestrogen that blocks ER transcriptional activity and is approved for ER-positive breast cancer. Fulvestrant also induces accumulation of insoluble ER and activates an unfolded protein response; proteasome inhibitors have been shown to enhance these effects in preclinical models. BACKGROUND Fulvestrant is a selective estrogen receptor (ER)-downregulating antiestrogen that blocks ER transcriptional activity and is approved for ER-positive (+) breast cancer. Fulvestrant also induces accumulation of insoluble ER and activates an unfolded protein response; proteasome inhibitors have been shown to enhance these effects in preclinical models. METHODS This is a single-center phase Ib study with a 3+3 design of fulvestrant and the proteasome inhibitor ixazomib (MLN9708) in patients with advanced ER+ breast cancer that was progressing on fulvestrant. A dose-escalation design allowed establishment of the ixazomib maximum tolerated dose (MTD). Secondary objectives included progression-free survival, pharmacokinetics, and tumor molecular analyses. RESULTS Among nine evaluable subjects, treatment was well-tolerated without dose-limiting toxicities The MTD of ixazomib was 4 mg in combination with fulvestrant. Plasma concentrations of the active form of ixazomib (MLN2238) in the 4-mg dose cohort had a median (range) maximal concentration (Cmax ) of 155 (122-171) ng/mL, time of maximal concentration (Tmax ) of 1 (1-1.5) hour, terminal elimination half-life of 66.6 (57.3-102.6) hour after initial dose, and area under the curve (AUC) of 5,025 (4,160-5,345) ng*h/mL. One partial response was observed, and median progression-free survival was 51 days (range, 47-137). CONCLUSION This drug combination has a favorable safety profile and antitumor activity in patients with fulvestrant-resistant advanced ER+ breast cancer that justifies future testing.
Collapse
Affiliation(s)
- Gary Schwartz
- Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Kevin Shee
- Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Bianca Romo
- Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Jonathan Marotti
- Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | | | - Lionel Lewis
- Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| | - Todd Miller
- Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire, USA
| |
Collapse
|
3
|
Dongala T, Palakurthi AK, Vytla Y, Katari NK. A novel UPLC-PDA isocratic method for the quantification fulvestrant in oil-based pre-filled syringe injection matrix formulations. J Anal Sci Technol 2019. [DOI: 10.1186/s40543-019-0171-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
4
|
Jessl L, Lenz R, Massing FG, Scheider J, Oehlmann J. Effects of estrogens and antiestrogens on gonadal sex differentiation and embryonic development in the domestic fowl ( Gallus gallus domesticus). PeerJ 2018; 6:e5094. [PMID: 30002959 PMCID: PMC6034593 DOI: 10.7717/peerj.5094] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 06/05/2018] [Indexed: 01/11/2023] Open
Abstract
Since it is known that environmental contaminants have the potential to cause endocrine disorders in humans and animals, there is an urgent need for in vivo tests to assess possible effects of these endocrine disrupting chemicals (EDCs). Although there is no standardized guideline, the avian embryo has proven to be particularly promising as it responds sensitively to a number of EDCs preferentially impacting the reproductive axis. In the present study we examined the effects of in ovo exposure to fulvestrant and tamoxifen as antiestrogenic model compounds and co-exposure to both substances and the potent estrogen 17α-ethinylestradiol (EE2) regarding sex differentiation and embryonic development of the domestic fowl (Gallus gallus domesticus). The substances were injected into the yolk of fertilized eggs on embryonic day 1. On embryonic day 19 sex genotype and phenotype were determined, followed by gross morphological and histological examination of the gonads. Sole EE2-treatment (20 ng/g egg) particularly affected male gonads and resulted in an increased formation of female-like gonadal cortex tissue and a reduction of seminiferous tubules. In ovo exposure to tamoxifen (0.1/1/10 µg/g egg) strongly impaired the differentiation of female gonads, led to a significant size reduction of the left ovary and induced malformations of the ovarian cortex, while fulvestrant (0.1/1/10 µg/g egg) did not affect sexual differentiation. However, both antiestrogens were able to antagonize the feminizing effects of EE2in genetic males when administered simultaneously. Since both estrogens and antiestrogens induce concentration-dependent morphological alterations of the sex organs, the chick embryo can be regarded as a promising model for the identification of chemicals with estrogenic and antiestrogenic activity.
Collapse
Affiliation(s)
- Luzie Jessl
- Department Aquatic Ecotoxicology, Johann Wolfgang Goethe Universität Frankfurt am Main, Frankfurt am Main, Hesse, Germany.,R-Biopharm AG, Darmstadt, Hesse, Germany
| | - Rebecca Lenz
- Department Aquatic Ecotoxicology, Johann Wolfgang Goethe Universität Frankfurt am Main, Frankfurt am Main, Hesse, Germany.,Dr. Drexler + Dr. Fecher GmbH, Groß-Umstadt, Hesse, Germany
| | - Fabian G Massing
- Department Aquatic Ecotoxicology, Johann Wolfgang Goethe Universität Frankfurt am Main, Frankfurt am Main, Hesse, Germany.,ERM GmbH, Neu-Isenburg, Hesse, Germany
| | - Jessica Scheider
- Department Aquatic Ecotoxicology, Johann Wolfgang Goethe Universität Frankfurt am Main, Frankfurt am Main, Hesse, Germany
| | - Jörg Oehlmann
- Department Aquatic Ecotoxicology, Johann Wolfgang Goethe Universität Frankfurt am Main, Frankfurt am Main, Hesse, Germany
| |
Collapse
|
5
|
Abstract
Small-molecule drug discovery has traditionally focused on occupancy of a binding site that directly affects protein function, and this approach typically precludes targeting proteins that lack such amenable sites. Furthermore, high systemic drug exposures may be needed to maintain sufficient target inhibition in vivo, increasing the risk of undesirable off-target effects. Induced protein degradation is an alternative approach that is event-driven: upon drug binding, the target protein is tagged for elimination. Emerging technologies based on proteolysis-targeting chimaeras (PROTACs) that exploit cellular quality control machinery to selectively degrade target proteins are attracting considerable attention in the pharmaceutical industry owing to the advantages they could offer over traditional small-molecule strategies. These advantages include the potential to reduce systemic drug exposure, the ability to counteract increased target protein expression that often accompanies inhibition of protein function and the potential ability to target proteins that are not currently therapeutically tractable, such as transcription factors, scaffolding and regulatory proteins.
Collapse
Affiliation(s)
| | - Craig M. Crews
- Departments of Molecular, Cellular & Developmental Biology; Chemistry; Pharmacology, Yale University, New Haven, CT 06511, USA
| |
Collapse
|
6
|
Bogliolo S, Cassani C, Dominoni M, Orlandini A, Ferrero S, Iacobone AD, Viazzo F, Venturini PL, Spinillo A, Gardella B. The role of fulvestrant in endometrial cancer. Expert Opin Drug Metab Toxicol 2016; 13:537-544. [PMID: 27696906 DOI: 10.1080/17425255.2016.1244264] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Endometrial cancer is the most common malignancy of the female genital tract in industrialized countries. The traditional treatment of endometrial cancer is based on a surgical approach. In recent years, systemic endocrine therapy has demonstrated good efficacy in recurrent or metastatic setting, delaying progression, ameliorating quality of life and palliating symptoms. Areas covered: Phase I and II studies on selective estrogen receptor down-regulators used for the treatment of endometrial cancer treatment have been reviewed. The pharmacokinetic and pharmacodynamic features of selective receptor down-regulators have been also investigated. Expert opinion: Selective estrogen receptor down-regulators may exhibit clinical efficacy in the treatment of gynecological malignancies due to their pure estrogen receptor antagonist properties. However, up to now data are still limited and some unsolved questions remain. Fulvestrant has poor oral bioavailability and low pharmacodynamic characteristics. Further trials are required to examine new selective estrogen receptor down-regulator agents with better pharmacodynamic and pharmacokinetic profiles.
Collapse
Affiliation(s)
- Stefano Bogliolo
- a Department of Obstetrics and Gynaecology , IRCCS-Fondazione Policlinico San Matteo, University of Pavia , Pavia , Italy
| | - Chiara Cassani
- a Department of Obstetrics and Gynaecology , IRCCS-Fondazione Policlinico San Matteo, University of Pavia , Pavia , Italy
| | - Mattia Dominoni
- a Department of Obstetrics and Gynaecology , IRCCS-Fondazione Policlinico San Matteo, University of Pavia , Pavia , Italy
| | - Anna Orlandini
- a Department of Obstetrics and Gynaecology , IRCCS-Fondazione Policlinico San Matteo, University of Pavia , Pavia , Italy
| | - Simone Ferrero
- b Department of Obstetrics and Gynaecology , IRCCS Azienda Ospedaliera Universitaria San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, University of Genoa , Genoa , Italy
| | - Anna Daniela Iacobone
- a Department of Obstetrics and Gynaecology , IRCCS-Fondazione Policlinico San Matteo, University of Pavia , Pavia , Italy
| | - Franco Viazzo
- a Department of Obstetrics and Gynaecology , IRCCS-Fondazione Policlinico San Matteo, University of Pavia , Pavia , Italy
| | - Pier Luigi Venturini
- b Department of Obstetrics and Gynaecology , IRCCS Azienda Ospedaliera Universitaria San Martino, IST Istituto Nazionale per la Ricerca sul Cancro, University of Genoa , Genoa , Italy
| | - Arsenio Spinillo
- a Department of Obstetrics and Gynaecology , IRCCS-Fondazione Policlinico San Matteo, University of Pavia , Pavia , Italy
| | - Barbara Gardella
- a Department of Obstetrics and Gynaecology , IRCCS-Fondazione Policlinico San Matteo, University of Pavia , Pavia , Italy
| |
Collapse
|
7
|
Hertz DL, Barlow WE, Kidwell KM, Albain KS, Vandenberg TA, Dakhil SR, Tirumali NR, Livingston RB, Gralow J, Hayes DF, Hortobagyi GN, Mehta RS, Rae JM. Fulvestrant decreases anastrozole drug concentrations when taken concurrently by patients with metastatic breast cancer treated on SWOG study S0226. Br J Clin Pharmacol 2016; 81:1134-41. [PMID: 26859101 PMCID: PMC4876171 DOI: 10.1111/bcp.12904] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 01/25/2016] [Accepted: 02/03/2016] [Indexed: 11/29/2022] Open
Abstract
AIMS In the SWOG S0226 trial the combination of anastrozole plus fulvestrant (n = 349) was superior to anastrozole alone (n = 345) in hormone receptor (HR)-positive metastatic breast cancer. Here we report a pharmacokinetic subset analysis investigating a possible drug interaction between anastrozole and fulvestrant. METHODS Post-menopausal patients with HR-positive metastatic breast cancer were randomized to anastrozole with or without concurrent fulvestrant. Blood samples were collected at 2, 4, 6 and 8 months, just prior to receiving the next dose of anastrozole and fulvestrant. Drug concentrations were measured via LC/MS-MS. Anastrozole concentration was compared in patients on anastrozole alone vs. patients on concomitant fulvestrant. Comparisons were made at each time point using parametric tests and over time using a linear mixed effects model. RESULTS A total of 483 anastrozole concentration measurements were included, 224 samples from 64 patients on the anastrozole alone arm and 259 from 73 patients on the combination arm. The mean anastrozole concentration in the combination arm was significantly lower than that in the anastrozole alone arm at each sample collection time (all P < 0.01) and in the mixed effects model (an estimated difference of 9.85 ng ml(-1) (95% CI 5.69, 14.00 ng ml(-1) ), P < 0.001). CONCLUSION A significant pharmacokinetic drug interaction was detected, in which the addition of fulvestrant to anastrozole treatment decreased the trough anastrozole concentration. Further research is needed to verify whether this interaction affects treatment efficacy and to determine the pharmacological mechanism by which this interaction occurs.
Collapse
Affiliation(s)
- Daniel L Hertz
- College of Pharmacy, University of Michigan, Ann Arbor, MI, USA
| | | | - Kelley M Kidwell
- University of Michigan, School of Public Health, Ann Arbor, MI, USA
| | - Kathy S Albain
- Loyola University Chicago Stritch School of Medicine, Maywood, IL, USA
| | - Ted A Vandenberg
- London Health Sciences Center/National Cancer Institute of Canada Clinical, Trials Group, London, ON, Canada
| | | | | | | | - Julie Gralow
- Seattle Cancer Care Alliance, Fred Hutchinson Cancer Research Center, University of Washington, Seattle, WA, USA
| | - Daniel F Hayes
- Division of Hematology/Oncology, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI, USA
| | | | - Rita S Mehta
- University of California Irvine Medical Center, Chao Family Comprehensive, Cancer Center, Orange, CA, USA
| | - James M Rae
- Division of Hematology/Oncology, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI, USA
| |
Collapse
|
8
|
Abstract
Estrogen receptors (ERs) are a group of compounds named for their importance in both menstrual and estrous reproductive cycles. They are involved in the regulation of various processes ranging from tissue growth maintenance to reproduction. Their action is mediated through ER nuclear receptors. Two subtypes of the estrogen receptor, ERα and ERβ, exist and exhibit distinct cellular and tissue distribution patterns. In humans, both receptor subtypes are expressed in many cells and tissues, and they control key physiological functions in various organ systems. Estrogens attract great attention due to their wide applications in female reproductive functions and treatment of some estrogen-dependent cancers and osteoporosis. This paper provides a general review of ER ligands published in international journals patented between 2013 and 2015. The broad physiological profile of estrogens has attracted the attention of many researchers to develop new estrogen ligands as therapeutic molecules for various clinical purposes. After the discovery of the ERβ receptor, subtype-selective ligands could be used to elicit beneficial estrogen-like activities and reduce adverse side effects, based on the different distributions and relative levels of the two ER subtypes in different estrogen target tissues. Therefore, recent literature has focused on selective estrogen ligands as highly promising agents for the treatment of some types of cancer, as well as for cardiovascular, inflammatory, and neurodegenerative diseases. Estrogen receptors are nuclear transcription factors that are involved in the regulation of many complex physiological functions in humans. Selective estrogen ligands are highly promising targets for treatment of some types of cancer, as well as for cardiovascular, inflammatory and neurodegenerative diseases. Extensive structure-activity relationship studies of ER ligands based on small molecules indicate that many different structural scaffolds may provide high-affinity compounds, provided that some basic structural requirements are present.
Collapse
|
9
|
Scott LJ, Croxtall JD, McKeage K. Fulvestrant: a guide to its use in hormone receptor-positive metastatic breast cancer in postmenopausal women in the EU. DRUGS & THERAPY PERSPECTIVES 2013. [DOI: 10.1007/s40267-013-0098-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
10
|
Shoda T, Okuhira K, Kato M, Demizu Y, Inoue H, Naito M, Kurihara M. Design and synthesis of tamoxifen derivatives as a selective estrogen receptor down-regulator. Bioorg Med Chem Lett 2013; 24:87-9. [PMID: 24332630 DOI: 10.1016/j.bmcl.2013.11.078] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 11/23/2013] [Accepted: 11/27/2013] [Indexed: 01/08/2023]
Abstract
We designed and synthesized an estrogen receptor (ER) down-regulator (5), which is a derivative of tamoxifen with a long alkyl side chain. Compound 5 effectively reduced ER protein levels in MCF-7 cells and had an antagonistic effect.
Collapse
Affiliation(s)
- Takuji Shoda
- Division of Organic Chemistry, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan.
| | - Keiichiro Okuhira
- Division of Biochemistry and Molecular Biology, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan
| | - Masashi Kato
- Division of Organic Chemistry, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan; School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Yosuke Demizu
- Division of Organic Chemistry, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan
| | - Hideshi Inoue
- School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Mikihiko Naito
- Division of Biochemistry and Molecular Biology, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan
| | - Masaaki Kurihara
- Division of Organic Chemistry, National Institute of Health Sciences, 1-18-1 Kamiyoga, Setagaya-ku, Tokyo 158-8501, Japan; Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, 4259 Nagatsuta, Midori-ku, Yokohama, Kanagawa 226-8501, Japan.
| |
Collapse
|
11
|
Li J, Ma Z, Jiang RW, Wu B. Hormone-related pharmacokinetic variations associated with anti-breast cancer drugs. Expert Opin Drug Metab Toxicol 2013; 9:1085-95. [PMID: 23687971 DOI: 10.1517/17425255.2013.802771] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Breast cancer is the most common female cancer, with more than one million new patients diagnosed annually worldwide. Generally speaking, there are three types of drugs used in management of breast cancer namely: hormonal treatment, chemotherapeutic agents and target-based agents. There is increasing evidence that hormones play an important role in development of both hormone-dependent and hormone-independent breast cancers. AREAS COVERED This review summarizes the pharmacokinetics of various types of drugs used to treat breast cancer. Furthermore, the authors discuss hormone-related variations including: the menstrual status, gender and exogenous hormones influencing drug absorption, distribution, metabolism or excretion (ADME). The authors also describe the physiological factors such as body weight and age that affect the pharmacokinetics of several drugs. EXPERT OPINION The factors affecting the pharmacokinetics of anti-breast cancer drugs are multifaceted. Hormones appear to be a key factor determining the pharmacokinetics (and efficacy) of hormonal therapy due to their role in cancer progression. In chemotherapy, the effects of hormones on the drug pharmacokinetics are possibly mediated through P-glycoprotein (P-gp) efflux and/or cytochrome P450 metabolism. In many cases, dosing regimen should be adjusted for drugs used in treatment of breast cancers based on the hormone levels in the body.
Collapse
Affiliation(s)
- Juan Li
- Jinan University, Institute of Traditional Chinese Medicine and Natural Products, Guangzhou, Guangdong 510632, China
| | | | | | | |
Collapse
|
12
|
Grattan BJ, Freake HC. Zinc and cancer: implications for LIV-1 in breast cancer. Nutrients 2012; 4:648-75. [PMID: 22852056 PMCID: PMC3407987 DOI: 10.3390/nu4070648] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 06/07/2012] [Accepted: 06/27/2012] [Indexed: 01/21/2023] Open
Abstract
Zinc is a trace mineral which is vital for the functioning of numerous cellular processes, is critical for growth, and may play an important role in cancer etiology and outcome. The intracellular levels of this mineral are regulated through the coordinated expression of zinc transporters, which modulate both zinc influx as well as efflux. LIV-1 (ZIP6) was first described in 1988 as an estrogen regulated gene with later work suggesting a role for this transporter in cancer growth and metastasis. Despite evidence of its potential utility as a target gene for cancer prognosis and treatment, LIV-1 has received relatively little attention, with only three prior reviews being published on this topic. Herein, the physiological effects of zinc are reviewed in light of this mineral’s role in cancer growth with specific attention being given to LIV-1 and the potential importance of this transporter to breast cancer etiology.
Collapse
Affiliation(s)
- Bruce J. Grattan
- Department of Family Medicine, Stony Brook University Hospital Medical Center, Stony Brook, New York, NY 11597, USA
- Authors to whom correspondence should be addressed; (B.J.G.); (H.C.F.); Tel.: +1-631-444-8245; Fax: +1-631-444-7552
| | - Hedley C. Freake
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06268, USA
- Authors to whom correspondence should be addressed; (B.J.G.); (H.C.F.); Tel.: +1-631-444-8245; Fax: +1-631-444-7552
| |
Collapse
|
13
|
Croxtall JD, McKeage K. Fulvestrant: a review of its use in the management of hormone receptor-positive metastatic breast cancer in postmenopausal women. Drugs 2011; 71:363-80. [PMID: 21319872 DOI: 10.2165/11204810-000000000-00000] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Fulvestrant (Faslodex®) is an intramuscularly administered steroidal estrogen receptor antagonist that is devoid of any known estrogen agonist effects. It is indicated as second-line therapy for the treatment of postmenopausal women with hormone receptor-positive advanced breast cancer who have progressed following prior endocrine therapy. In well designed, randomized clinical trials, regimens of fulvestrant 250 and 500 mg provided effective second-line therapy for postmenopausal women with advanced breast cancer who had progressed following prior endocrine therapy. Moreover, fulvestrant 250 mg monthly (with or without a loading dose) was as effective as aromatase inhibitor therapy. However, fulvestrant is absorbed slowly, and greater steady-state concentrations are achieved more rapidly when using a higher dosage with a loading dose regimen. Consequently, a regimen of fulvestrant 500 mg monthly with a loading dose was significantly more effective than a regimen of 250 mg monthly in postmenopausal women with disease progression. Limited data also indicate a potential role for the fulvestrant 500 mg regimen as first-line therapy. Fulvestrant is generally well tolerated with no additional adverse events noted with the high-dose regimen compared with the 250 mg regimens. Furthermore, the incidence of joint disorders was shown to be significantly lower with fulvestrant 250 mg monthly than with anastrozole. Treatment with fulvestrant is not associated with any clinically significant effects on endometrial thickening, bone-specific turnover markers or sex hormone levels. In conclusion, a monthly regimen of intramuscular fulvestrant 500 mg with a loading dose provides effective and well tolerated second-line therapy for postmenopausal women with advanced breast cancer who have progressed following prior endocrine therapy and is now the approved optimal dose.
Collapse
|
14
|
Li YW, Zhu GY, Shen XL, Chu JH, Yu ZL, Fong WF. Furanodienone inhibits cell proliferation and survival by suppressing ERα signaling in human breast cancer MCF-7 cells. J Cell Biochem 2011; 112:217-24. [DOI: 10.1002/jcb.22922] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
15
|
Ishii Y, Papa L, Bahadur U, Yue Z, Aguirre-Ghiso J, Shioda T, Waxman S, Germain D. Bortezomib enhances the efficacy of fulvestrant by amplifying the aggregation of the estrogen receptor, which leads to a proapoptotic unfolded protein response. Clin Cancer Res 2011; 17:2292-300. [PMID: 21292820 DOI: 10.1158/1078-0432.ccr-10-1745] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE Fulvestrant is known to promote the degradation of the estrogen receptor (ER) in the nucleus. However, fulvestrant also promotes the aggregation of the newly synthesized ER in the cytoplasm. Accumulation of protein aggregates leads to cell death but this effect is limited as a result of their elimination by the proteasome. We tested whether combining fulvestrant with the proteasome inhibitor, bortezomib, could enhance the accumulation of ER aggregates and cause apoptotic cell death. EXPERIMENTAL DESIGN The rate of aggregation of the ER was monitored in ER(+) breast cancer cells lines, T47D, ZR-75.1, BT474, MDA-MB-361, MCF-7, fulvestrant resistance MCF-7, and tamoxifen-resistant T47D-cyclin D1 cells. Activation of the unfolded protein response, apoptosis, and metabolic rate were also monitored in these cell lines following treatment with fulvestrant, bortezomib, or bortezomib in combination with fulvestrant. RESULTS We found that bortezomib enhances the fulvestrant-mediated aggregation of the ER in the cytoplasm without blocking the degradation of the ER in the nucleus. Further, these aggregates activate a sustained unfolded protein response leading to apoptotic cell death. Further, we show that the combination induced tumor regression in a breast cancer mouse model of tamoxifen resistance. CONCLUSIONS Adding bortezomib to fulvestrant enhances its efficacy by taking advantage of the unique ability of fulvestrant to promote cytoplasmic aggregates of the ER. As this effect of fulvestrant is independent of the transcriptional activity of the ER, these results suggest that this novel combination may be effective in breast cancers that are ER(+) but estrogen independent.
Collapse
Affiliation(s)
- Yuki Ishii
- Division of Hematology/Oncology, Tisch Cancer Institute and Department of Neurology and Neuroscience, Mount Sinai School of Medicine, New York, New York, USA
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Kocanova S, Mazaheri M, Caze-Subra S, Bystricky K. Ligands specify estrogen receptor alpha nuclear localization and degradation. BMC Cell Biol 2010; 11:98. [PMID: 21143970 PMCID: PMC3009626 DOI: 10.1186/1471-2121-11-98] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2010] [Accepted: 12/10/2010] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The estrogen receptor alpha (ERα) is found predominately in the nucleus, both in hormone stimulated and untreated cells. Intracellular distribution of the ERα changes in the presence of agonists but the impact of different antiestrogens on the fate of ERα is a matter of debate. RESULTS A MCF-7 cell line stably expressing GFP-tagged human ERα (SK19 cell line) was created to examine the localization of ligand-bound GFP-ERα. We combined digitonin-based cell fractionation analyses with fluorescence and immuno-electron microscopy to determine the intracellular distribution of ligand-bound ERα and/or GFP-ERα.Using fluorescence- and electron microscopy we demonstrate that both endogenous ERα and GFP-ERα form numerous nuclear focal accumulations upon addition of agonist, 17β-estradiol (E2), and pure antagonists (selective estrogen regulator disruptor; SERD), ICI 182,780 or RU58,668, while in the presence of partial antagonists (selective estrogen regulator modulator; SERM), 4-hydroxytamoxifen (OHT) or RU39,411, diffuse nuclear staining persisted.Digitonin based cell fractionation analyses confirmed that endogenous ERα and GFP-ERα predominantly reside in the nuclear fraction. Overall ERα protein levels were reduced after estradiol treatment. In the presence of SERMs ERα was stabilized in the nuclear soluble fraction, while in the presence of SERDs protein levels decreased drastically and the remaining ERα was largely found in a nuclear insoluble fraction. mRNA levels of ESR1 were reduced compared to untreated cells in the presence of all ligands tested, including E2. E2 and SERDs induced ERα degradation occurred in distinct nuclear foci composed of ERα and the proteasome providing a simple explanation for ERα sequestration in the nucleus. CONCLUSIONS Our results indicate that chemical structure of ligands directly affect the nuclear fate and protein turnover of the estrogen receptor alpha independently of their impact on transcription. These findings provide a molecular basis for the selection of antiestrogen compounds issue from pharmacological studies aimed at improving treatment of breast cancer.
Collapse
Affiliation(s)
- Silvia Kocanova
- Université de Toulouse; UPS; Laboratoire de Biologie Moléculaire Eucaryote; F-31062 Toulouse, France
| | | | | | | |
Collapse
|
17
|
Demir K, Unuvar T, Eren S, Abaci A, Bober E. Tamoxifen as first-line treatment in a premenarchal girl with juvenile breast hypertrophy. J Pediatr Adolesc Gynecol 2010; 23:e133-6. [PMID: 20813327 DOI: 10.1016/j.jpag.2009.11.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2009] [Revised: 11/04/2009] [Accepted: 11/05/2009] [Indexed: 10/19/2022]
Abstract
BACKGROUND Juvenile breast hypertrophy is uncommon and is characterized by excessive breast enlargement in the peripubertal period. The clinical entity is thought to result from increased sensitivity of mammary tissue to normal levels of circulating hormones. CASE Here, we report a female patient, aged 12 years and 6 months, suffering from juvenile breast hypertrophy, who presented at the third month of symptoms and benefited from tamoxifen treatment. COMMENTS In experienced clinics, use of tamoxifen in the treatment of juvenile breast hypertrophy during the brisk growth period may become a medical alternative to reconstructive surgery.
Collapse
Affiliation(s)
- Korcan Demir
- Department of Pediatric Endocrinology, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| | | | | | | | | |
Collapse
|
18
|
Alternative strategies for the treatment of classical congenital adrenal hyperplasia: pitfalls and promises. INTERNATIONAL JOURNAL OF PEDIATRIC ENDOCRINOLOGY 2010; 2010:670960. [PMID: 20652035 PMCID: PMC2905899 DOI: 10.1155/2010/670960] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Accepted: 05/11/2010] [Indexed: 01/09/2023]
Abstract
Despite decades of different treatment algorithms, the management of congenital adrenal hyperplasia (CAH) remains clinically challenging. This is due to the inherent difficulty of suppressing adrenal androgen production using near physiological dosing of glucocorticoids (GC). As a result, alternating cycles of androgen versus GC excess can occur and may lead to short stature, obesity, virilization, and alterations in puberty. Novel therapeutic alternatives, including new and more physiological means of GC delivery, inhibitors at the level of CRH or ACTH secretion and/or action, as well as “rescue strategies”, such as GnRH analogs, anti-androgens, aromatase inhibitors, and estrogen receptor blockers, are available; many of these agents, however, still require active investigation in CAH. Bilateral adrenalectomy is effective but it is also still an experimental approach. Gene therapy and stem cells, to provide functional adrenal cortical tissue, are at preclinical stage but provide exciting avenues for a potential cure for CAH.
Collapse
|
19
|
Kabos P, Borges VF. Fulvestrant: a unique antiendocrine agent for estrogen-sensitive breast cancer. Expert Opin Pharmacother 2010; 11:807-16. [PMID: 20151846 DOI: 10.1517/14656561003641982] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
IMPORTANCE OF THE FIELD The role of estrogen deprivation for the treatment of breast cancer has been understood since the 1800s. Pharmacologic advances in the field in the past decades, including tamoxifen and the aromatase inhibitors, have contributed significantly to the reduced mortality of estrogen-sensitive breast cancer. However, this subtype of breast cancer still presents with relapses and, once metastatic, progression to hormone-refractory state and loss of disease control remain an expected disease course. Fulvestrant, a pure estrogen receptor downregulator, is a new addition to the antiestrogen therapeutic armamentarium since its FDA approval in 2002. Its unique mechanism of action offers potential advantages over other estrogen targeted therapies. AREAS COVERED IN THIS REVIEW Published scientific literature, including presented abstracts, on fulvestrant from 1985 to the present were reviewed with selected publications included. WHAT THE READER WILL GAIN This review addresses current issues and therapies for estrogen-sensitive breast cancer, highlights the role of fulvestrant in current treatment guidelines and outlines some of the ongoing investigations of this compound. TAKE HOME MESSAGE Fulvestrant is an effective and well-tolerated drug for treatment of metastatic estrogen-sensitive breast cancer. Work is underway to enhance its clinical benefit to patients as a single agent and in combination with other therapies.
Collapse
Affiliation(s)
- Peter Kabos
- Division of Medical Oncology, University of Colorado - Denver, 12801 E 17th Avenue, Mailstop 8117, Aurora, CO 80045, USA.
| | | |
Collapse
|
20
|
Inhibition of drug metabolizing cytochrome P450s by the aromatase inhibitor drug letrozole and its major oxidative metabolite 4,4'-methanol-bisbenzonitrile in vitro. Cancer Chemother Pharmacol 2009; 64:867-75. [PMID: 19198839 DOI: 10.1007/s00280-009-0935-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2008] [Accepted: 01/08/2009] [Indexed: 12/21/2022]
Abstract
PURPOSE To determine the inhibitory potency of letrozole and its main human metabolite, 4,4'-methanol-bisbenzonitrile, on the activities of eight cytochrome P450 (CYP) enzymes. METHODS Letrozole and its metabolite were incubated with human liver microsomes (HLMs) (or expressed CYP isoforms) and NADPH in the absence (control) and presence of the test inhibitor. RESULTS Letrozole was a potent competitive inhibitor of CYP2A6 (K (i) 4.6 +/- 0.05 microM and 5.0 +/- 2.4 microM in HLMs and CYP2A6, respectively) and a weak inhibitor of CYP2C19 (K (i) 42.2 microM in HLMs and 33.3 microM in CYP2C19), while its metabolite showed moderate inhibition of CYP2C19 and CYP2B6. Letrozole or its metabolite had negligible effect on other CYPs. CONCLUSIONS Based on the in vitro K (i) values, letrozole is predicted to be a weak inhibitor of CYP2A6 in vivo. Letrozole and its major human metabolite show inhibitory activity towards other CYPs, but clinically relevant drug interactions seem less likely as the K (i) values are above the therapeutic plasma concentrations of letrozole.
Collapse
|
21
|
|
22
|
Jansen GH, Franke HR, Wolbers F, Brinkhuis M, Vermes I. Effects of fulvestrant alone or combined with different steroids in human breast cancer cells in vitro. Climacteric 2008; 11:315-21. [PMID: 18645697 DOI: 10.1080/13697130802232500] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVES Fulvestrant is an estrogen receptor (ER) antagonist that binds, blocks and degrades the estrogen receptor and is currently used in adjuvant treatment in postmenopausal women with ER-positive breast cancer as an alternative for tamoxifen. As an antagonist, it may induce or aggravate climacteric symptoms. In order to alleviate these symptoms, one could consider hormone therapy. The objective of this study was to analyze the effect of fulvestrant alone or in combination with different steroids in human breast cancer cells in vitro, and to demonstrate whether these steroids will compromise the efficacy of fulvestrant in ER-positive breast cancer cells. METHODS We performed experiments in vitro with various hormone therapy preparations (estradiol (E2), dihydrodydrogesterone (DHD) and tibolone) at a concentration of 10(-6) mol/l alone or combined with fulvestrant in different breast cancer cell lines, ER-positive and ER-negative. After an incubation of 144 h, proliferation and apoptosis were measured. The first was measured by quantification of the expression of cyclin D1 mRNA, the latter by the Nicoletti fragmentation assay. RESULTS This in vitro study revealed clear differences in results when various hormone therapy preparations, alone or combined with fulvestrant, are added to ER-positive and ER-negative breast cancer cell lines. CONCLUSIONS Our study demonstrated that fulvestrant, an ER antagonist used in the treatment of ER-positive breast cancer, combined with E2 and DHD or in combination with tibolone, is not compromised in its efficacy in inducing apoptosis in ER-positive breast cancer cell lines in vitro.
Collapse
Affiliation(s)
- G H Jansen
- Department of Obstetrics, Twente Hospital Group, Enschede, The Netherlands
| | | | | | | | | |
Collapse
|
23
|
Abstract
Recent clinical data on selective estrogen receptor modulators (SERMs) have provided the basis for reassessment of the SERM concept. The molecular basis of SERM activity involves binding of the ligand SERM to the estrogen receptor (ER), causing conformational changes which facilitate interactions with coactivator or corepressor proteins, and subsequently initiate or suppress transcription of target genes. SERM activity is intrinsic to each ER ligand, which accomplishes its unique profile by specific interactions in the target cell, leading to tissue selective actions. We discuss the estrogenic and anti-estrogenic effects of early SERMs, such as clomiphene citrate, used for treatment of ovulation induction, and the triphenylethylene, tamoxifen, which has ER antagonist activity in the breast, and is used for prevention and treatment of ER-positive breast cancer. Since the development of tamoxifen, other triphenylethylene SERMs have been studied for breast cancer prevention, including droloxifene, idoxifene, toremifene, and ospemifene. Other SERMs have entered clinical development more recently, including benzothiophenes (raloxifene and arzoxifene), benzopyrans (ormeloxifene, levormeloxifene, and EM-800), lasofoxifene, pipendoxifene, bazedoxifene, HMR-3339, and fulvestrant, an anti-estrogen which is approved for breast cancer treatment. SERMs have effects on tissues containing ER, such as the breast, bone, uterine and genitourinary tissues, and brain, and on markers of cardiovascular risk. Current evidence indicates that each SERM has a unique array of clinical activities. Differences in the patterns of action of SERMs suggest that each clinical end point must be evaluated individually, and conclusions about any particular SERM can only be established through appropriate clinical trials.
Collapse
|
24
|
Wittmann BM, Sherk A, McDonnell DP. Definition of functionally important mechanistic differences among selective estrogen receptor down-regulators. Cancer Res 2007; 67:9549-60. [PMID: 17909066 DOI: 10.1158/0008-5472.can-07-1590] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
One subclass of antiestrogens, the selective estrogen receptor down-regulators (SERDs), have received considerable attention of late as they competitively inhibit estrogen binding and induce a rapid, proteasome-dependent degradation of the receptor. Contained within this class of molecules is the steroidal antiestrogen ICI182,780 (faslodex), recently approved for the treatment of metastatic cancer, and GW5638/DPC974, a SERD that is currently being evaluated in the clinic. Given that mechanistic differences between different selective estrogen receptor modulators have been translated into important clinical profiles, it was of interest to determine if the SERD subclass of ligands were likewise functionally or mechanistically distinguishable. In this study, we show that although the steroidal and nonsteroidal SERDs target ERalpha for degradation, the underlying mechanism(s) are different. Of note was the identification of a specific protein-protein interaction surface presented on ERalpha in the presence of the ICI182,780-activated receptor which is required for degradation. Interestingly, this surface is also presented on ERalpha in the presence of RU58,668, a SERD that is chemically distinct from ICI182,780. This surface is not required for GW5638-mediated degradation, and thus, this SERD seems to affect ERalpha down-regulation by a different mechanism. These data suggest that sequencing of therapies using drugs of this class is likely to be possible. Finally, because of the unmet need for orally active SERDS that function similarly to ICI182,780, we have used the insights from these mechanistic studies to develop and validate a high-throughput screen for compounds of this class with improved pharmaceutical properties.
Collapse
Affiliation(s)
- Bryan M Wittmann
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, 27710, USA
| | | | | |
Collapse
|
25
|
Abstract
HsEg5 (Eg5) is a kinesin required for proper execution of mitosis. Several compounds that specifically block Eg5 are in clinical development and have the potential to be used in the treatment of breast cancer. In this study, we investigated the interaction between Eg5 and estrogen receptor signaling. We observed decreased Eg5 expression after treatment of estrogen receptor-positive human breast cancer MCF-7 cells with the estrogen receptor downregulator fulvestrant. Downregulation of Eg5 expression in response to fulvestrant was also observed in another estrogen receptor-positive cell line ZR-75, but not in the estrogen receptor-negative breast cancer cell line MDA-231. Moreover, in MCF-7 cells previously arrested in the G0/G1 phase of the cell cycle by fulvestrant, addition of estrogen increased Eg5 expression. This upregulation correlated with progression through S-phase. Nevertheless, the effect of fulvestrant in Eg5 expression could not be explained solely by cell cycle arrest, because treatments that blocked cell cycle progression did not consistently decrease Eg5 expression. Pharmacological inhibition of Eg5 function, with either S-trityl-L-cysteine or monastrol, prevented growth of estrogen-treated MCF-7 cells with an IC50 of 0.46 and 29.71 micromol/l, respectively. Simultaneous inhibition of estrogen receptor function with fulvestrant increased the IC50 for S-trityl-L-cysteine to 2.30 micromol/l and for monastrol to 112.69 micromol/l. Our results suggest that pharmacological inhibition of Eg5 may be an effective treatment for estrogen receptor-positive breast cancer, even without concomitant hormonal therapy.
Collapse
Affiliation(s)
- Maricarmen D Planas-Silva
- Department of Pharmacology (H078), Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA.
| | | |
Collapse
|