1
|
Mukherjee AG, Wanjari UR, Nagarajan D, K K V, V A, P JP, T TP, Chakraborty R, Renu K, Dey A, Vellingiri B, Gopalakrishnan AV. Letrozole: Pharmacology, toxicity and potential therapeutic effects. Life Sci 2022; 310:121074. [DOI: 10.1016/j.lfs.2022.121074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 10/02/2022] [Accepted: 10/10/2022] [Indexed: 11/07/2022]
|
2
|
Yang C, Li P, Li Z. Clinical application of aromatase inhibitors to treat male infertility. Hum Reprod Update 2021; 28:30-50. [PMID: 34871401 DOI: 10.1093/humupd/dmab036] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 10/14/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Infertility affects 15% of men and contributes to nearly half of all cases of infertility. Infertile men usually have impaired spermatogenesis, presenting as azoospermia or various degrees of asthenospermia and oligozoospermia. Spermatogenesis is a complex and coordinated process, which is under precise modulation by the hypothalamic-pituitary-gonadal (HPG) axis. An aberrant hormone profile, especially an imbalance between testosterone (T) and estradiol (E2), plays an essential role in male infertility. In the male, E2 is produced mainly from the conversion of T by the aromatase enzyme. Theoretically, reducing an abnormally elevated T:E2 ratio using aromatase inhibitors (AIs) could restore the balance between T and E2 and optimize the HPG axis to support spermatogenesis. For decades, AIs have been used to treat male infertility empirically. However, owing to the lack of large-scale randomized controlled studies and basic research, the treatment efficacy and safety of AIs in male infertility remain controversial. Therefore, there is a need to summarize the clinical trials and relevant basic research on the application of AIs in the treatment of male infertility. OBJECTIVE AND RATIONALE In this narrative review, we summarized the application of AIs in the treatment of male infertility, including the pharmacological mechanisms involved, clinical trials focused on patients with different types of infertility, factors affecting treatment efficacy and the side-effects. SEARCH METHODS A literature search was performed using MEDLINE/PubMed and EMBASE, focusing on publications in the past four decades concerning the use of AIs for treating male infertility. The search terms included AI, male infertility, letrozole, anastrozole, testolactone, azoospermia, oligozoospermia, aromatase polymorphisms, obesity and antiestrogens, in various combinations. OUTCOMES Clinical studies demonstrate that AIs, especially nonsteroidal letrozole and anastrozole, could significantly inhibit the production of E2 and its negative feedback on the HPG axis, resulting in increased T and FSH production as well as improved semen parameters in infertile men. Large-scale surveys suggest that obesity may result in symptoms of hypogonadism in both fertile and infertile males, such as decreased semen quality and attenuated sexual function, which can be improved by AIs treatment. Polymorphisms of the aromatase gene CYP19A1, including single nucleotide polymorphisms and tetranucleotide TTTA repeats polymorphism (TTTAn), also influence hormone profiles, semen quality and treatment efficacy of AIs in male hypogonadotropic hypogonadism and infertility. The side-effects of AIs in treating male infertility are various, but most are mild and well tolerated. WIDER IMPLICATIONS The application of AIs in treating male infertility has been off-label and empirical for decades. This narrative review has summarized the target patients, dose, treatment duration and side-effects of AIs. Polymorphisms of CYP19A1 that may affect AIs treatment efficacy were also summarized, but a full understanding of the mechanisms involved in AIs action requires further study.
Collapse
Affiliation(s)
- Chao Yang
- Department of Andrology, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Peng Li
- Department of Andrology, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Li
- Department of Andrology, Urologic Medical Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Li Y, Kong X, Xuan L, Wang Z, Huang YH. Prolactin and endocrine therapy resistance in breast cancer: The next potential hope for breast cancer treatment. J Cell Mol Med 2021; 25:10327-10348. [PMID: 34651424 PMCID: PMC8581311 DOI: 10.1111/jcmm.16946] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/21/2021] [Accepted: 09/19/2021] [Indexed: 12/20/2022] Open
Abstract
Breast cancer, a hormone‐dependent tumour, generally includes four molecular subtypes (luminal A, luminal B, HER2 enriched and triple‐negative) based on oestrogen receptor, progesterone receptor and human epidermal growth factor receptor‐2. Multiple hormones in the body regulate the development of breast cancer. Endocrine therapy is one of the primary treatments for hormone‐receptor‐positive breast cancer, but endocrine resistance is the primary clinical cause of treatment failure. Prolactin (PRL) is a protein hormone secreted by the pituitary gland, mainly promoting mammary gland growth, stimulating and maintaining lactation. Previous studies suggest that high PRL levels can increase the risk of invasive breast cancer in women. The expression levels of PRL and PRLR in breast cancer cells and breast cancer tissues are elevated in most ER+ and ER− tumours. PRL activates downstream signalling pathways and affects endocrine therapy resistance by combining with prolactin receptor (PRLR). In this review, we illustrated and summarized the correlations between endocrine therapy resistance in breast cancer and PRL, as well as the pathophysiological mechanisms and clinical practices. The study on PRL and its receptor would help explore reversing endocrine therapy‐resistance for breast cancer.
Collapse
Affiliation(s)
- Yuan Li
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiangyi Kong
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lixue Xuan
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhongzhao Wang
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yen-Hua Huang
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
4
|
Alimardani M, Moghbeli M, Rastgar-Moghadam A, Shandiz FH, Abbaszadegan MR. Single nucleotide polymorphisms as the efficient prognostic markers in breast cancer. Curr Cancer Drug Targets 2021; 21:768-793. [PMID: 34036920 DOI: 10.2174/1568009621666210525151846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 03/15/2021] [Accepted: 04/19/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Breast cancer (BC) is known as the most common malignancy in women. Environmental and genetic factors are associated with BC progression. Genetic polymorphisms have been reported as important risk factors of BC prognosis and drug response. Main body: Therefore, in the present review, we have summarized all single nucleotide polymorphisms (SNPs) which have been significantly associated with drug response in BC patients around the world. We have also categorized the reported SNPs based on their related genes functions to clarify the molecular biology of drug responses in BC. CONCLUSION The majority of SNPs were reported in detoxifying enzymes, which introduced such genes as the main genetic risk factors during BC drug responses. This review paves the way for introducing a prognostic panel of SNPs for the BC patients in the world.
Collapse
Affiliation(s)
- Maliheh Alimardani
- Medical Genetics Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Azam Rastgar-Moghadam
- Medical Genetics Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Homaei Shandiz
- Department of Radiotherapy/Oncology, Omid Hospital, Mashhad University of Medical Science, Mashhad, Iran
| | - Mohammad Reza Abbaszadegan
- Medical Genetics Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
5
|
Umamaheswaran G, Kadambari D, Muthuvel SK, Kumar NAN, Dubashi B, Aibor Dkhar S, Adithan C. Polymorphisms of T- cell leukemia 1A gene loci are not related to the development of adjuvant letrozole-induced adverse events in breast cancer. PLoS One 2021; 16:e0247989. [PMID: 33760860 PMCID: PMC7990231 DOI: 10.1371/journal.pone.0247989] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 02/17/2021] [Indexed: 01/24/2023] Open
Abstract
Letrozole, an aromatase inhibitor (AI), is the first-line adjuvant drug for treating hormone receptor-positive (HR+) breast cancer in postmenopausal women. However, harmful adverse events (AEs) and significant differences in drug response among individuals remain a significant problem in clinical application. Current evidence suggests that the observed individual variation in the treatment outcomes of AI is conferred by genetic variants. Hence, in this study, we examined the association of TCL1A gene polymorphisms with letrozole-induced AEs. The study subjects were postmenopausal HR+ breast cancer patients who were receiving adjuvant letrozole. Genomic DNA was isolated by a routine standard phenol-chloroform method. In total, 198 South Indian patients were genotyped for four single nucleotide polymorphisms (SNPs) in the TCL1A gene loci by the TaqMan allelic discrimination assay using the RT-PCR system. We used the odds ratio and 95% confidence interval to assess the genetic association. Musculoskeletal (MS) AEs and vasomotor symptoms (VMSs) are the most common side effects observed in the study cohort. Among 198 patients, 81 experienced musculoskeletal toxicity, reporting MS-AEs, 57 had VMSs, and 33 of them had both. The most frequently identified polymorphic variants in the patient series were rs11849538 (G), with an allele frequency of about 27.3%, followed by rs7158782-G (27.3%), rs7159713-G (25.8%), and rs2369049-G (22.5%). The genetic association analysis indicated no significant difference in the proportion of TCL1A gene variants between patients with and without AEs on either MS-AEs or VMSs. Though we observed high LD in all patient groups, the inferred haplotypes displayed a non-significant association with letrozole-induced specific AEs. However, the SNP functionality analysis by RegulomeDB provided a 2b rank score for rs7158782, suggesting a potential biological function. Our findings suggest that TCL1A gene polymorphisms may not play any role in the prediction of letrozole-induced AEs in South Indian HR+ breast cancer patients.
Collapse
Affiliation(s)
- Gurusamy Umamaheswaran
- Department of Pharmacology, Centre for Advanced Research in Pharmacogenomics, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
- * E-mail:
| | - Dharanipragada Kadambari
- Departments of Surgery and Medical Education, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Suresh Kumar Muthuvel
- Center for Bioinformatics, School of Life Sciences, Pondicherry University, Puducherry, India
| | - Naveena A. N. Kumar
- Departments of Surgery and Medical Education, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Biswajit Dubashi
- Department of Medical Oncology, Regional Cancer Center, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Steven Aibor Dkhar
- Department of Pharmacology, Centre for Advanced Research in Pharmacogenomics, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Chandrasekaran Adithan
- Department of Pharmacology, Centre for Advanced Research in Pharmacogenomics, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
- Department of Clinical Pharmacology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| |
Collapse
|
6
|
Gagno S, D'Andrea MR, Mansutti M, Zanusso C, Puglisi F, Dreussi E, Montico M, Biason P, Cecchin E, Iacono D, Russo S, Cinausero M, Saracchini S, Gasparini G, Sartori D, Bari M, Collovà E, Meo R, Merkabaoui G, Spagnoletti I, Pellegrino A, Gianni L, Sandri P, Cretella E, Vattemi E, Rocca A, Serra P, Fabbri MA, Benedetti G, Foghini L, Medici M, Basso U, Amoroso V, Riccardi F, Baldelli AM, Clerico M, Bonura S, Saggia C, Innocenti F, Toffoli G. A New Genetic Risk Score to Predict the Outcome of Locally Advanced or Metastatic Breast Cancer Patients Treated With First-Line Exemestane: Results From a Prospective Study. Clin Breast Cancer 2019; 19:137-145.e4. [PMID: 30584056 DOI: 10.1016/j.clbc.2018.11.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 11/18/2018] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Approximately 50% of locally advanced or metastatic breast cancer (MBC) patients treated with first-line exemestane do not show objective response and currently there are no reliable biomarkers to predict the outcome of patients using this therapy. The constitutive genetic background might be responsible for differences in the outcome of exemestane-treated patients. We designed a prospective study to investigate the role of germ line polymorphisms as biomarkers of survival. PATIENTS AND METHODS Three hundred two locally advanced or MBC patients treated with first-line exemestane were genotyped for 74 germ line polymorphisms in 39 candidate genes involved in drug activity, hormone balance, DNA replication and repair, and cell signaling pathways. Associations with progression-free survival (PFS) and overall survival (OS) were tested with multivariate Cox regression. Bootstrap resampling was used as an internal assessment of results reproducibility. RESULTS Cytochrome P450 19A1-rs10046TC/CC, solute carrier organic anion transporter 1B1-rs4149056TT, adenosine triphosphate binding cassette subfamily G member 2-rs2046134GG, fibroblast growth factor receptor-4-rs351855TT, and X-ray repair cross complementing 3-rs861539TT were significantly associated with PFS and then combined into a risk score (0-1, 2, 3, or 4-6 risk points). Patients with the highest risk score (4-6 risk points) compared with ones with the lowest score (0-1 risk points) had a median PFS of 10 months versus 26.3 months (adjusted hazard ratio [AdjHR], 3.12 [95% confidence interval (CI), 2.18-4.48]; P < .001) and a median OS of 38.9 months versus 63.0 months (AdjHR, 2.41 [95% CI, 1.22-4.79], P = .012), respectively. CONCLUSION In this study we defined a score including 5 polymorphisms to stratify patients for PFS and OS. This score, if validated, might be translated to personalize locally advanced or MBC patient treatment and management.
Collapse
Affiliation(s)
- Sara Gagno
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano, IRCCS, Aviano, Italy
| | | | - Mauro Mansutti
- Department of Oncology, University Hospital of Udine, Udine, Italy
| | - Chiara Zanusso
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano, IRCCS, Aviano, Italy
| | - Fabio Puglisi
- Medical Oncology and Cancer Prevention Unit, Centro di Riferimento Oncologico di Aviano, IRCCS, Aviano, Italy; Medical Oncology, Department of Medicine, University of Udine, Udine, Italy
| | - Eva Dreussi
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano, IRCCS, Aviano, Italy
| | - Marcella Montico
- Scientific Directorate, Centro di Riferimento Oncologico di Aviano, IRCCS, Aviano, Italy
| | - Paola Biason
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano, IRCCS, Aviano, Italy; Medical Oncology Unit 1, Istituto Oncologico Veneto IOV IRCCS, Padova, Italy
| | - Erika Cecchin
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano, IRCCS, Aviano, Italy
| | - Donatella Iacono
- Department of Oncology, University Hospital of Udine, Udine, Italy
| | - Stefania Russo
- Department of Oncology, University Hospital of Udine, Udine, Italy
| | - Marika Cinausero
- Department of Oncology, University Hospital of Udine, Udine, Italy
| | - Silvana Saracchini
- Medical Oncology Unit, Santa Maria degli Angeli Hospital, Pordenone, Italy
| | | | - Donata Sartori
- Medical Oncology Department, General Hospital, Mirano, Italy
| | - Mario Bari
- Medical Oncology Department, General Hospital, Mirano, Italy
| | - Elena Collovà
- Oncology Operative Unit, ASST Ovest Milanese, Ospedale di Legnano, Legnano, Italy
| | - Rosa Meo
- Medical Oncology Unit, Presidio Ospedaliero Sant'Alfonso Maria dei Liguori, Cerreto Sannita, Italy
| | - Ghassan Merkabaoui
- Medical Oncology Unit, Azienda Ospedaliera Universitaria Federico II di Napoli, Napoli, Italy
| | - Ilaria Spagnoletti
- Medical Oncology Unit, Ospedale Sacro Cuore di Gesù, Fatebenefratelli, Benevento, Italy
| | - Arianna Pellegrino
- Medical Oncology Unit, Ospedale San Pietro Fatebenefratelli, Rome, Italy
| | | | - Paolo Sandri
- Medical Oncology Unit, San Vito al Tagliamento Hospital, Pordenone, Italy
| | | | - Emanuela Vattemi
- Medical Oncology, Azienda Sanitaria dell'Alto Adige, Bolzano, Italy
| | - Andrea Rocca
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), IRCCS, Meldola, Italy
| | - Patrizia Serra
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), IRCCS, Meldola, Italy
| | - Maria Agnese Fabbri
- Division of Oncology, Complesso Ospedaliero Belcolle, AUSL Viterbo, Viterbo, Italy
| | | | | | - Michele Medici
- Department of Medical Oncology, Azienda ULSS 3 Serenissima, Mestre, Italy
| | - Umberto Basso
- Medical Oncology Unit 1, Istituto Oncologico Veneto IOV IRCCS, Padova, Italy
| | - Vito Amoroso
- Medical Oncology Unit, Spedali Civili Hospital, Brescia, Italy
| | | | - Anna Maria Baldelli
- Medical Oncology Unit, Azienda Ospedaliera Ospedali Riuniti Marche Nord, San Salvatore Hospital, Pesaro, Italy
| | - Mario Clerico
- Department of Oncology, Ospedale degli Infermi, Biella, Italy
| | | | - Chiara Saggia
- Azienda Ospedaliero-Universitaria Maggiore della Carità, Novara, Italy
| | | | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano, IRCCS, Aviano, Italy.
| |
Collapse
|
7
|
Sen F, Aydiner A. Endocrine Therapy of Metastatic Breast Cancer. Breast Cancer 2019. [DOI: 10.1007/978-3-319-96947-3_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
8
|
Hamadeh IS, Patel JN, Rusin S, Tan AR. Personalizing aromatase inhibitor therapy in patients with breast cancer. Cancer Treat Rev 2018; 70:47-55. [PMID: 30086432 DOI: 10.1016/j.ctrv.2018.07.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 07/19/2018] [Accepted: 07/20/2018] [Indexed: 01/15/2023]
Abstract
BACKGROUND Aromatase inhibitors are the mainstay of therapy for patients with hormone receptor-positive breast cancer in both adjuvant and metastatic settings. Their use in clinical practice has been challenged by significant inter-individual variability in response and tolerability. Hence, the purpose of this paper is to provide a succinct review of the literature on the genetic factors contributing to this variability. DESIGN A systematic search in PUBMED was conducted to identify studies that investigated the association between germline polymorphisms and disposition, clinical response and toxicities of aromatase inhibitors, as well as those evaluating the implications of mutations in ESR1 on clinical response. RESULTS Polymorphisms in genes coding for phase I and phase II enzymes (pharmacokinetic genes) significantly modulated exposure to aromatase inhibitors; however, there is a paucity of data linking interindividual variability in drug exposure to clinical response. Furthermore, pharmacogenetic studies interrogating relationship between polymorphisms in CYP19A1 (the target site of aromatase inhibitors, i.e. a pharmacodynamic gene) and response yielded conflicting results. Acquired mutations in ESR1 receptors have been identified as the underlying mechanism of resistance to aromatase inhibitors, and likely predict drug response. Although some pharmacogenetic studies have implicated polymorphisms in CYP19A1 and ESR1 with drug-related side effects, the putative role of these genes in predicting toxicity warrants further validation. CONCLUSION Genetic polymorphisms in pharmacokinetic and pharmacodynamic genes appear to influence aromatase inhibitor disposition, response and/or toxicity; however, prospective interventional studies are needed to understand the application of genomics to personalize aromatase inhibitor therapy in breast cancer patients.
Collapse
Affiliation(s)
- Issam S Hamadeh
- Department of Cancer Pharmacology, Levine Cancer Institute, Atrium Health, Charlotte, NC, United States.
| | - Jai N Patel
- Department of Cancer Pharmacology, Levine Cancer Institute, Atrium Health, Charlotte, NC, United States; University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC, United States
| | - Stephanie Rusin
- Department of Cancer Pharmacology, Levine Cancer Institute, Atrium Health, Charlotte, NC, United States; University of North Carolina Eshelman School of Pharmacy, Chapel Hill, NC, United States
| | - Antoinette R Tan
- Department of Solid Tumor Oncology and Investigational Therapeutics, Levine Cancer Institute, Atrium Health, Charlotte, NC, United States
| |
Collapse
|
9
|
Sini V, Botticelli A, Lunardi G, Gori S, Marchetti P. Pharmacogenetics and aromatase inhibitor induced side effects in breast cancer patients. Pharmacogenomics 2017; 18:821-830. [PMID: 28592202 DOI: 10.2217/pgs-2017-0006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
This paper reviews genetic variations mainly related to the onset of adverse events during aromatase inhibitors in early breast cancer. Genetic variability could occur at different steps. The analysis included studies that involved breast cancer patients, treated with an aromatase inhibitor, genotyped for CYP19A1 and/or CYP17A1 and/or CYP27B1 and/or TCLA1, and/or RANK/RANKL/OPG and/or ESR1/ESR2, and assessed for toxicity profile. Twenty-two articles were included for the analysis. Three studies evaluated outcomes and adverse events; 19 studies assessed only side effects. Functional variations may be useful in predicting the onset of toxicities. The identification of polymorphisms at increased risk of toxicity may enable patient management. However, more data are needed to be applied in the individualization of treatment in daily practice.
Collapse
Affiliation(s)
- Valentina Sini
- Clinical & Molecular Medicine Department, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy.,Oncology Unit - ASL Roma 1 - Santo Spirito Hospital, Rome, Italy
| | - Andrea Botticelli
- Clinical & Molecular Medicine Department, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Gianluigi Lunardi
- Medical Oncology Unit, Sacro Cuore Don Calabria Hospital, Negrar VR, Italy
| | - Stefania Gori
- Medical Oncology Unit, Sacro Cuore Don Calabria Hospital, Negrar VR, Italy
| | - Paolo Marchetti
- Clinical & Molecular Medicine Department, Sant'Andrea Hospital, Sapienza University of Rome, Rome, Italy.,Oncology Unit, IDI - I.R.C.C.S., Rome, Italy
| |
Collapse
|
10
|
Glubb DM, O'Mara TA, Shamsani J, Spurdle AB. The Association of CYP19A1 Variation with Circulating Estradiol and Aromatase Inhibitor Outcome: Can CYP19A1 Variants Be Used to Predict Treatment Efficacy? Front Pharmacol 2017; 8:218. [PMID: 28487654 PMCID: PMC5403944 DOI: 10.3389/fphar.2017.00218] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 04/06/2017] [Indexed: 11/26/2022] Open
Abstract
After menopause, estradiol is primarily synthesized in peripheral tissues by the enzyme aromatase, encoded by CYP19A1. CYP19A1 variation associates with circulating estradiol in postmenopausal women and this variation is best represented by the intronic variant rs727479. This variation appears to have pleiotropic effects as it also associates with endometrial cancer risk. Indeed, estradiol plays an important role in the development of breast and endometrial cancer. Aromatase inhibitor (AI) drugs are used in the treatment of both diseases, however, response rates for AIs are low and there is currently no way to identify breast or endometrial cancer patients who are more likely to receive a clinical benefit. Multiple studies have proposed that genetic variation in CYP19A1 will have effects on AI efficacy: eight candidate variant studies of sample size greater than 50 describe associations between CYP19A1 variation and the outcome of patients treated with AIs. Nominally significant associations with patient outcome were reported for several variants, including rs727479. However, only an association between rs4646 and time to progression was replicated in an independent study. Moreover, rs4646 is also the only variant that has an association with patient outcome that passes a multiple testing threshold and this variant is in linkage disequilibrium with rs727479, supporting the hypothesis that associations with patient outcome may be driven through the effects on circulating estradiol. Despite this preliminary evidence, well phenotyped and comprehensively genotyped patient sets need to be studied before conclusions can be drawn about the effects of CYP19A1 variation on AI efficacy.
Collapse
Affiliation(s)
- Dylan M Glubb
- Molecular Cancer Epidemiology, Department of Genetics and Computational Biology, QIMR Berghofer Medical Research InstituteBrisbane, QLD, Australia
| | - Tracy A O'Mara
- Molecular Cancer Epidemiology, Department of Genetics and Computational Biology, QIMR Berghofer Medical Research InstituteBrisbane, QLD, Australia
| | - Jannah Shamsani
- Molecular Cancer Epidemiology, Department of Genetics and Computational Biology, QIMR Berghofer Medical Research InstituteBrisbane, QLD, Australia
| | - Amanda B Spurdle
- Molecular Cancer Epidemiology, Department of Genetics and Computational Biology, QIMR Berghofer Medical Research InstituteBrisbane, QLD, Australia
| |
Collapse
|
11
|
Hertz DL, Henry NL, Rae JM. Germline genetic predictors of aromatase inhibitor concentrations, estrogen suppression and drug efficacy and toxicity in breast cancer patients. Pharmacogenomics 2017; 18:481-499. [PMID: 28346074 PMCID: PMC6219438 DOI: 10.2217/pgs-2016-0205] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 12/21/2016] [Indexed: 02/07/2023] Open
Abstract
The third-generation aromatase inhibitors (AIs), anastrozole, letrozole and exemestane, are highly effective for the treatment of estrogen receptor-positive breast cancer in postmenopausal women. AIs inhibit the aromatase (CYP19A1)-mediated production of estrogens. Most patients taking AIs achieve undetectable blood estrogen concentrations resulting in drug efficacy with tolerable side effects. However, some patients have suboptimal outcomes, which may be due, in part, to inherited germline genetic variants. This review summarizes published germline genetic associations with AI treatment outcomes including systemic AI concentrations, estrogenic response to AIs, AI treatment efficacy and AI treatment toxicities. Significant associations are highlighted with commentary about prioritization for future validation to identify pharmacogenetic predictors of AI treatment outcomes that can be used to inform personalized treatment decisions in patients with estrogen receptor-positive breast cancer.
Collapse
Affiliation(s)
- Daniel L Hertz
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI 48109-1065, USA
| | - N Lynn Henry
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84103, USA
| | - James M Rae
- Breast Oncology Program, University of Michigan Comprehensive Cancer Center, Ann Arbor, MI 48109-1065, USA
| |
Collapse
|
12
|
Polymorphisms in drug-metabolizing enzymes and steady-state exemestane concentration in postmenopausal patients with breast cancer. THE PHARMACOGENOMICS JOURNAL 2016; 17:521-527. [PMID: 27549341 PMCID: PMC5323433 DOI: 10.1038/tpj.2016.60] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Revised: 06/13/2016] [Accepted: 06/15/2016] [Indexed: 12/18/2022]
Abstract
Discovery of clinical and genetic predictors of exemestane pharmacokinetics was attempted in 246 post-menopausal patients with breast cancer enrolled on a prospective clinical study. A sample was collected two hours after exemestane dosing at a 1 or 3 month study visit to measure drug concentration. The primary hypothesis was that patients carrying the low-activity CYP3A4*22 (rs35599367) SNP would have greater exemestane concentration. Additional SNPs in genes relevant to exemestane metabolism (CYP1A1/2, CYP1B1, CYP3A4, CYP4A11, AKR1C3/4, AKR7A2) were screened in secondary analyses and adjusted for clinical covariates. CYP3A4*22 was associated with a 54% increase in exemestane concentration (p<0.01). Concentration was greater in patients who reported White race, had elevated aminotransferases, renal insufficiency, lower body mass index, and had not received chemotherapy (all p<0.05), and CYP3A4*22 maintained significance after adjustment for covariates (p<0.01). These genetic and clinical predictors of exemestane concentration may be useful for treatment individualization in patients with breast cancer.
Collapse
|
13
|
Liu X, Low SK, Boddy AV. The implications of genetic variation for the pharmacokinetics and pharmacodynamics of aromatase inhibitors. Expert Opin Drug Metab Toxicol 2016; 12:851-63. [PMID: 27253864 DOI: 10.1080/17425255.2016.1196189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Breast cancer is the most common female cancer and remains a serious public health concern worldwide. Third-generation aromatase inhibitors (AIs) are widely used in postmenopausal women with estrogen receptor positive breast cancer. However, there is marked interindividual variability in terms of the efficacy and incidence of adverse events following treatment with AIs. Pharmacogenetics has the potential to predict clinical outcomes based on patients' genetic information, paving the way towards personalized treatment. AREAS COVERED This article reviews pharmacogenetic studies of AIs, including pharmacokinetic and pharmacodynamic aspects, highlighting those studies where the efficacy and adverse events of AIs have been examined using both candidate gene and genome-wide approaches. EXPERT OPINION Pharmacogenetics is a promising approach to develop personalized medicine with AIs. However, the application of pharmacogenetics to predict therapeutic efficacy and adverse events in breast cancer patients is still far from implementation in routine clinical practice. Large, comprehensive, multicenter studies that simultaneously evaluate multiple genes and pathways, including rare variants, are warranted in order to produce reliable and informative results. The ultimate aim is to develop clinically-relevant guidelines for breast cancer therapy.
Collapse
Affiliation(s)
- Xiaoman Liu
- a Faculty of Pharmacy , University of Sydney , Sydney , Australia
| | - Siew-Kee Low
- a Faculty of Pharmacy , University of Sydney , Sydney , Australia
| | - Alan V Boddy
- a Faculty of Pharmacy , University of Sydney , Sydney , Australia
| |
Collapse
|
14
|
Papakonstantinou A, Foukakis T, Rodriguez-Wallberg KA, Bergh J. Is Estradiol Monitoring Necessary in Women Receiving Ovarian Suppression for Breast Cancer? J Clin Oncol 2016; 34:1573-9. [DOI: 10.1200/jco.2015.65.3493] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The Oncology Grand Rounds series is designed to place original reports published in the Journal into clinical context. A case presentation is followed by a description of diagnostic and management challenges, a review of the relevant literature, and a summary of the authors’ suggested management approaches. The goal of this series is to help readers better understand how to apply the results of key studies, including those published in Journal of Clinical Oncology, to patients seen in their own clinical practice. A 36-year-old premenopausal woman had been diagnosed with stage III breast cancer. After an initial biopsy confirmed breast cancer, she underwent mastectomy and axillary node dissection for a left-sided breast cancer, measuring 7 cm. The tumor had lobular histology and was considered grade 2 of 3. Metastatic carcinoma was identified in 10 of 13 axillary nodes. Immunohistochemical studies showed that the tumor was strongly positive for estrogen and progesterone receptor expression and had a Ki-67 score of 15% (> 20% is considered high according to a Swedish quality control study and the St Gallen Expert Consensus). 1 , 2 There was no amplification of the HER2/neu gene. Staging scans were negative for metastatic disease. In the adjuvant setting, she received three cycles of anthracycline-cyclophosphamide combination chemotherapy followed by three cycles of taxane chemotherapy and then locoregional radiotherapy. After completion of chemotherapy, she developed amenorrhea. As adjuvant endocrine therapy, she began monthly goserelin administration to achieve ovarian function suppression (OFS), in combination with the aromatase inhibitor (AI) exemestane. She experienced menopausal symptoms including hot flashes, vaginal dryness, and sexual dysfunction. After two monthly treatments with goserelin and exemestane, a sensitive assay for serum estradiol was checked and returned at 16 pg/mL (61 pmol/L); postmenopausal range for sensitive assay is less than 15 pg/mL (< 50 pmol/L). The patient has now been referred to our unit to discuss further management.
Collapse
Affiliation(s)
| | | | | | - Jonas Bergh
- Karolinska Institutet and University Hospital, Stockholm, Sweden
| |
Collapse
|
15
|
Mazzuca F, Botticelli A, Mazzotti E, La Torre M, Borro M, Marchetti L, Maddalena C, Gentile G, Simmaco M, Marchetti P. CYP19A1 Genetic Polymorphisms rs4646 and Osteoporosis in Patients Treated with Aromatase Inhibitor-Based Adjuvant Therapy. Eurasian J Med 2016; 48:10-4. [PMID: 27026757 PMCID: PMC4792488 DOI: 10.5152/eurasianjmed.2015.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 02/19/2015] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE Third-generation aromatase inhibitors (AI) are potent suppressors of aromatase activity. The aim of this study was to measure the incidence of adverse effects in breast cancer patients treated with AI-based adjuvant therapy and the relationship with the CYP19A1 genotypes. MATERIALS AND METHODS Forty-five postmenopausal breast cancer patients (46-85 yrs) in AI adjuvant treatment were genotyped for the rs4646 polymorphisms of CYP19A1 gene and three variations were identified. Toxicities were registered at each follow-up medical examination, and classified in accord with the Common Terminology Criteria for Adverse Events. RESULTS Twenty-four (53.3%) patients presented the GG genotype; 19 (42.2%) the GT, and 2 (4.4%) the TT. The AI treatment was Anastrazole for 35 patients (77.8%) and Letrozole for the others (n=10; 22.2%). Osteoporosis was significantly associated with the GG genotype (p=0.001). Treatment discontinuation (TD) was observed in 6 cases (13.3%). The only parameter able to predict TD was the appearance of severe arthralgia/myalgia (Odds Ratio, OR=23.75; p=0.009), when adjusted for age and AI treatment. CONCLUSION Our results suggest that CYP19A1 polymorphic variants may influence susceptibility to develop AI-related side effects. Further prospective studies are needed to confirm the role of the aromatase gene (CYP19A1) polymorphisms in predicting adverse effects to AI-based therapy.
Collapse
Affiliation(s)
- Federica Mazzuca
- Department of Clinical Oncology, Sant’Andrea Hospital, School of Medicine and Psychology, University of Rome La Sapienza, Rome, Italy
| | - Andrea Botticelli
- Department of Clinical Oncology, Sant’Andrea Hospital, School of Medicine and Psychology, University of Rome La Sapienza, Rome, Italy
| | - Eva Mazzotti
- Department of Emergency Medicine, Acıbadem University School of Medicine, İstanbul, Turkey
| | - Marco La Torre
- Department of General Surgery, Sant’Andrea Hospital, School of Medicine and Psychology, University of Rome La Sapienza, Rome, Italy
| | - Marina Borro
- Department of Advanced Molecular Diagnosis Unit (DiMA), Sant’Andrea Hospital Department of Medical-Surgical Sciences and Translational Medicine, Faculty of Medicine and Psychology, University of Rome La Sapienza, Rome, Italy
| | - Luca Marchetti
- Department of Clinical Oncology, Policlinico Umberto I, School of Medicine and Psychology, University of Rome La Sapienza, Rome, Italy
| | - Chiara Maddalena
- Department of Clinical Oncology, Sant’Andrea Hospital, School of Medicine and Psychology, University of Rome La Sapienza, Rome, Italy
| | - Giovanna Gentile
- Department of Advanced Molecular Diagnosis Unit (DiMA), Sant’Andrea Hospital Department of Medical-Surgical Sciences and Translational Medicine, Faculty of Medicine and Psychology, University of Rome La Sapienza, Rome, Italy
| | - Maurizio Simmaco
- Department of Advanced Molecular Diagnosis Unit (DiMA), Sant’Andrea Hospital Department of Medical-Surgical Sciences and Translational Medicine, Faculty of Medicine and Psychology, University of Rome La Sapienza, Rome, Italy
| | - Paolo Marchetti
- Department of Clinical Oncology, Sant’Andrea Hospital, School of Medicine and Psychology, University of Rome La Sapienza, Rome, Italy
| |
Collapse
|
16
|
Rumiato E, Brunello A, Ahcene-Djaballah S, Borgato L, Gusella M, Menon D, Pasini F, Amadori A, Saggioro D, Zagonel V. Predictive markers in elderly patients with estrogen receptor-positive breast cancer treated with aromatase inhibitors: an array-based pharmacogenetic study. THE PHARMACOGENOMICS JOURNAL 2015; 16:525-529. [PMID: 26503812 DOI: 10.1038/tpj.2015.73] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 08/27/2015] [Accepted: 09/08/2015] [Indexed: 11/09/2022]
Abstract
So far, no reliable predictive clinicopathological markers of response to aromatase inhibitors (AIs) have been identified, and little is known regarding the role played by host genetics. To identify constitutive predictive markers, an array-based association study was performed in a cohort of 55 elderly hormone-dependent breast cancer (BC) patients treated with third-generation AIs. The array used in this study interrogates variants in 225 drug metabolism and disposition genes with documented functional significance. Six variants emerged as associated with response to AIs: three located in ABCG1, UGT2A1, SLCO3A1 with a good response, two in SLCO3A1 and one in ABCC4 with a poor response. Variants in the AI target CYP19A1 resulted associated with a favourable response only as haplotype; haplotypes with increased response association were also detected for ABCG1 and SLCO3A1. These results highlight the relevance of host genetics in the response to AIs and represent a first step toward precision medicine for elderly BC patients.
Collapse
Affiliation(s)
- E Rumiato
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - A Brunello
- Medical Oncology 1 Unit, Department of Clinical and Experimental Oncology, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - S Ahcene-Djaballah
- Medical Oncology 1 Unit, Department of Clinical and Experimental Oncology, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - L Borgato
- Hemato-Oncology Unit, Medical Science Department ULSS 13, Mirano, Venezia, Italy
| | - M Gusella
- Division of Oncology, Rovigo General Hospital, ULSS 18, Rovigo, Italy
| | - D Menon
- Division of Oncology, Rovigo General Hospital, ULSS 18, Rovigo, Italy
| | - F Pasini
- Division of Oncology, Rovigo General Hospital, ULSS 18, Rovigo, Italy
| | - A Amadori
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy.,Department of Surgery, Oncology, and Gastroenterology, Oncology Section, University of Padova, Padova, Italy
| | - D Saggioro
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - V Zagonel
- Medical Oncology 1 Unit, Department of Clinical and Experimental Oncology, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| |
Collapse
|
17
|
Abstract
Breast cancer is a heterogeneous disease that necessitates proper patient classification to direct surgery, pharmacotherapy, and radiotherapy. Despite patients within the same subgroup receiving similar pharmacotherapy, substantial variation in clinical outcomes is observed. Pharmacogenetic variations with direct effect on pharmacokinetics and pharmacodynamics play a central role in clinical outcomes. Pharmacogenetic markers associated with clinical outcome are known as biomarkers. They are termed prognostic biomarkers when their presence is associated with a specific clinical outcome. If the presence of such biomarkers guides treatment, they are termed predictive biomarkers. A number of pharmacogenetic markers have been described in relation to breast cancer pharmacotherapy both in the adjuvant and neoadjuvant setting. CYP2D6 allelic variants produce variable rates of tamoxifen metabolism and are associated with survival outcomes. Other biomarkers have been described in relation to other forms of endocrine therapy and trastuzumab. In neoadjuvant and adjuvant breast cancer chemotherapy, specific biomarkers were correlated with clinical outcomes and risk of drug toxicity. This review highlights key biomarkers in breast cancer pharmacotherapy with the potential of translating such study outcomes into clinical practice.
Collapse
|
18
|
Artigalás O, Vanni T, Hutz MH, Ashton-Prolla P, Schwartz IV. Influence of CYP19A1 polymorphisms on the treatment of breast cancer with aromatase inhibitors: a systematic review and meta-analysis. BMC Med 2015; 13:139. [PMID: 26067721 PMCID: PMC4475294 DOI: 10.1186/s12916-015-0373-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Accepted: 05/18/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Many clinical trials have shown the efficacy of aromatase inhibitors (AIs) in the management of breast cancer (BC). There is growing evidence that CYP19A1 single-nucleotide polymorphisms (SNPs) are associated with clinical response (CR) and adverse effects (AEs) among BC patients treated with AIs. The aim of this study was to analyze the association between CYP19A1 polymorphisms and AI treatment in BC patients. METHODS A systematic review was performed in MEDLINE, EMBASE, and LILACS. A meta-analysis was conducted to compare the association between CYP19A1 variants and treatment response among BC patients. RESULTS A total of 12 studies were included in the final analysis. There was significant variation among the populations studied and the SNPs and outcomes investigated. A meta-analysis was only possible for the evaluation of SNP rs4646 vs. the wild-type variant with respect to time to progression (TTP) among metastatic BC patients treated with AI. TTP was significantly increased in patients with the rs4646 variant compared with the wild-type gene (hazard ratio (HR) = 0.51 [95 % confidence interval (CI), 0.33-0.78], P = 0.002). Seven studies analyzed the association between AEs with different polymorphisms of CYP19A1. Although there was a statistically significant association with musculoskeletal adverse events (rs934635, rs60271534, rs700518rs, and haplotype M_3_5) and with vasomotor symptoms (rs934635, rs1694189, rs7176005, and haplotype M_5_3) in individual studies, similar associations were not observed in further studies. No statistically significant association between musculoskeletal AEs and SNPs rs4646, rs10046, rs727479, and rs1062033 was found. CONCLUSIONS These findings suggest that the presence of the rs4646 variant may be a predictive factor of the benefit of AI treatment for BC. The effects of CYP19A1 polymorphisms on clinical outcomes were most often detected in individual studies, suggesting that longer-term studies will better clarify these associations. Additional studies are needed to clarify the predictive value of other SNPs and whether CYP19A1 genotyping should be used to guide AI treatment.
Collapse
Affiliation(s)
- Osvaldo Artigalás
- Postgraduate Program in Genetics and Molecular Biology, Department of Genetics, UFRGS, Av. Bento Gonçalves, 9500 - Prédio 43323M CEP: 91501-970 - Caixa Postal 15053, Porto Alegre, Rio Grande do Sul, Brazil. .,Genetics Unit, Children's Hospital, Grupo Hospitalar Conceição, GHC, Av. Francisco Trein, 596, CEP 91350-200, Porto Alegre, RS, Brazil.
| | - Tazio Vanni
- Coordenação Geral de Avaliação de Tecnologias em Saúde - CGATS, Department of Science and Technology, Ministry of Health, SCN Quadra 02 Projeção C Subsolo Sala T-004, CEP: 70712-902, Brasília, DF, Brazil.
| | - Mara Helena Hutz
- Postgraduate Program in Genetics and Molecular Biology, Department of Genetics, UFRGS, Av. Bento Gonçalves, 9500 - Prédio 43323M CEP: 91501-970 - Caixa Postal 15053, Porto Alegre, Rio Grande do Sul, Brazil.
| | - Patricia Ashton-Prolla
- Postgraduate Program in Genetics and Molecular Biology, Department of Genetics, UFRGS, Av. Bento Gonçalves, 9500 - Prédio 43323M CEP: 91501-970 - Caixa Postal 15053, Porto Alegre, Rio Grande do Sul, Brazil. .,Medical Genetics Service, Hospital de Clinicas de Porto Alegre, HCPA, Rua Ramiro Barcelos, 2350, CEP: 90035-903, Porto Alegre, RS, Brazil.
| | - Ida Vanessa Schwartz
- Postgraduate Program in Genetics and Molecular Biology, Department of Genetics, UFRGS, Av. Bento Gonçalves, 9500 - Prédio 43323M CEP: 91501-970 - Caixa Postal 15053, Porto Alegre, Rio Grande do Sul, Brazil. .,Medical Genetics Service, Hospital de Clinicas de Porto Alegre, HCPA, Rua Ramiro Barcelos, 2350, CEP: 90035-903, Porto Alegre, RS, Brazil.
| |
Collapse
|
19
|
Yamamoto-Ibusuki M, Arnedos M, André F. Targeted therapies for ER+/HER2- metastatic breast cancer. BMC Med 2015; 13:137. [PMID: 26059247 PMCID: PMC4462184 DOI: 10.1186/s12916-015-0369-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 05/14/2015] [Indexed: 12/31/2022] Open
Abstract
The majority of breast cancers present with estrogen receptor (ER)-positive and human epidermal growth factor receptor (HER2)-negative features and might benefit from endocrine therapy. Although endocrine therapy has notably evolved during the last decades, the invariable appearance of endocrine resistance, either primary or secondary, remains an important issue in this type of tumor. The improvement of our understanding of the cancer genome has identified some promising targets that might be responsible or linked to endocrine resistance, including alterations affecting main signaling pathways like PI3K/Akt/mTOR and CCND1/CDK4-6 as well as the identification of new ESR1 somatic mutations, leading to an array of new targeted therapies that might circumvent or prevent endocrine resistance. In this review, we have summarized the main targeted therapies that are currently being tested in ER+ breast cancer, the rationale behind them, and the new agents and combinational treatments to come.
Collapse
Affiliation(s)
- Mutsuko Yamamoto-Ibusuki
- Department of Breast and Endocrine Surgery, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan.
| | - Monica Arnedos
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France.
- INSERM Unit U981, Gustave Roussy Cancer Campus, Villejuif, France.
| | - Fabrice André
- Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France.
- INSERM Unit U981, Gustave Roussy Cancer Campus, Villejuif, France.
- Department of Medical Oncology and INSERM Unit U981, Gustave Roussy Cancer Campus, 114 Rue Edouard Vaillant, Villejuif, 94800, France.
| |
Collapse
|
20
|
Leyland-Jones B, Gray KP, Abramovitz M, Bouzyk M, Young B, Long B, Kammler R, Dell'Orto P, Biasi MO, Thürlimann B, Lyng MB, Ditzel HJ, Harvey VJ, Neven P, Treilleux I, Rasmussen BB, Maibach R, Price KN, Coates AS, Goldhirsch A, Pagani O, Viale G, Rae JM, Regan MM. CYP19A1 polymorphisms and clinical outcomes in postmenopausal women with hormone receptor-positive breast cancer in the BIG 1-98 trial. Breast Cancer Res Treat 2015; 151:373-84. [PMID: 25935582 DOI: 10.1007/s10549-015-3378-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 04/08/2015] [Indexed: 12/11/2022]
Abstract
To determine whether CYP19A1 polymorphisms are associated with abnormal activity of aromatase and with musculoskeletal and bone side effects of aromatase inhibitors. DNA was isolated from tumor specimens of 4861 postmenopausal women with hormone receptor-positive breast cancer enrolled in the BIG 1-98 trial to receive tamoxifen and/or letrozole for 5 years. Tumors were genotyped for six CYP19A1 polymorphisms using PCR-based methods. Associations with breast cancer-free interval (BCFI), distant recurrence-free interval (DRFI), musculoskeletal and bone adverse events (AEs) were assessed using Cox proportional hazards models. All statistical tests were two-sided. No association between the CYP19A1 genotypes and BCFI or DRFI was observed overall. A reduced risk of a breast cancer event for tamoxifen-treated patients with rs700518 variants was observed (BCFI CC/TC vs. TT: HR 0.53, 95 % CI 0.34-0.82, interaction P = 0.08), but not observed for letrozole-treated patients. There was an increased risk of musculoskeletal AEs for patients with rs700518 variants CC/TC versus TT (HR 1.22, 95 % CI 1.03-1.45, P = 0.02), regardless of treatment. Tamoxifen-treated patients with rs4646 variants had a reduced risk of bone AEs (AA/CA vs. CC: HR 0.76, 95 % CI 0.59-0.98), whereas an increase of minor allele (C) of rs10046 was associated with an increased risk of bone AEs (HR 1.28, 95 % CI 1.07-1.52). rs936308 variants were associated with a reduced risk of bone AEs in letrozole-treated patients (GG/GC vs. CC: HR 0.73, 95 % CI 0.54-0.99), different from in tamoxifen-treated patients (GG/GC vs. CC: HR 1.32, 95 % CI 0.92-1.90, interaction P = 0.01). CYP19A1 rs700518 variants showed associations with BCFI, DRFI, in tamoxifen treated patients and musculoskeletal AEs regardless of treatment. SNPs rs4646, rs10046, and rs936308 were associated with bone AEs.
Collapse
|
21
|
Shao X, Guo Y, Xu X, Zheng Y, Wang J, Chen Z, Huang J, Huang P, Cai J, Wang X. The CYP19 RS4646 polymorphism IS related to the prognosis of stage I-II and operable stage III breast cancer. PLoS One 2015; 10:e0121535. [PMID: 25793413 PMCID: PMC4368615 DOI: 10.1371/journal.pone.0121535] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 02/03/2015] [Indexed: 12/03/2022] Open
Abstract
Purpose Aromatase, encoded by the CYP19 gene, catalyzes the final step of the conversion of androgens to estrogens. Given the critical role of CYP19 in estrogen synthesis, the potential influence of CYP19 rs4646 polymorphism on breast cancer survival, deserves further study. Methods Genotyping for CYP19 rs4646 variants was performed on 406 Chinese women with stage I–II and operable stage III breast cancer. Associations were evaluated between CYP19 rs4646 genotypes and disease-free survival (DFS). Results In premenopausal patients, women who are homozygous for the minor allele (AA) have a longer DFS compared with those carrying the major allele (CC or AC) (87 months versus 48.7 months; Hazard ratio (HR) = 0.56, 95 % CI = 0.318-0.985, P = 0.041). These differences were further demonstrated by a multivariate analysis (HR = 0.456, 95 % CI = 0.249-0.836, P = 0.011). Conversely, the same variant (AA) was estimated to be associated with a poorer DFS in postmenopausal women (AA versus AC or CC: 13.7 months versus 56.3 months; HR = 2.758, 95 % CI = 1.432-5.313, P = 0.002). Furthermore, the differences were confirmed by the COX proportional hazards model (HR = 2.983, 95% CI =1.494-5.955, P = 0.002). Conclusions The present study indicates that CYP19 rs4646 polymorphism is related to DFS in early breast cancer and that the prognosis index of the homozygous for the minor allele (AA) may depend on menopause status. The findings are novel, if confirmed, rs4646 genotypes may provide useful information for routine management in breast cancer.
Collapse
Affiliation(s)
- Xiying Shao
- Department of Medical Oncology, The Affiliated Zhejiang Cancer Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Yong Guo
- Department of Oncology, The First Affiliated Hospital of Zhejiang Traditional Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Xiaohong Xu
- Clinical laboratory, The Affiliated Zhejiang Cancer Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Yabing Zheng
- Department of Medical Oncology, The Affiliated Zhejiang Cancer Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Jiwen Wang
- Department of Oncology, Zhejiang Cancer Hospital, Hangzhou, Zhejiang Province, China
| | - Zhanhong Chen
- Department of Medical Oncology, The Affiliated Zhejiang Cancer Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Jian Huang
- Department of Medical Oncology, The Affiliated Zhejiang Cancer Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Ping Huang
- Department of Medical Oncology, The Affiliated Zhejiang Cancer Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Jufen Cai
- Department of Medical Oncology, The Affiliated Zhejiang Cancer Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
| | - Xiaojia Wang
- Department of Medical Oncology, The Affiliated Zhejiang Cancer Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang Province, China
- * E-mail:
| |
Collapse
|
22
|
Blackburn HL, Ellsworth DL, Shriver CD, Ellsworth RE. Role of cytochrome P450 genes in breast cancer etiology and treatment: effects on estrogen biosynthesis, metabolism, and response to endocrine therapy. Cancer Causes Control 2015; 26:319-32. [PMID: 25554091 DOI: 10.1007/s10552-014-0519-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Accepted: 12/19/2014] [Indexed: 10/24/2022]
Abstract
PURPOSE The cytochrome P450 (CYP) genes are oxygenases involved in estrogen biosynthesis and metabolism, generation of DNA damaging procarcinogens, and response to anti-estrogen therapies. Since lifetime estrogen exposure is an established risk factor for breast cancer, determining the role of CYP genes in breast cancer etiology may provide critical information for understanding tumorigenesis and response to treatment. METHODS This review summarizes literature available in PubMed published between 1993 and 2013 that focuses on studies evaluating the effects of DNA variants in CYP genes on estrogen synthesis, metabolism, and generation of procarcinogens in addition to response to anti-estrogen therapies. RESULTS Evaluation of DNA variants in estrogen metabolism genes was largely inconclusive. Meta-analyses of data from CYP19A1 support an association between the number of (TTTA) n repeats in intron 4 and breast cancer risk, but the biological mechanism for this relationship is unknown. Associations between single nucleotide polymorphism in CYP1B1 and DNA damage caused by procarcinogenic estrogen metabolites were ambiguous. Variants in CYP2D6 are associated with altered metabolism tamoxifen; however, current data do not support widespread clinical testing. The effect of variants in CYP19A1 in response to aromatase inhibitors is also questionable. CONCLUSION Evaluation of DNA variants in CYP genes involved with estrogen metabolism or treatment response has been inconclusive, reflecting small samples sizes, tumor heterogeneity, and differences between populations. Better-powered studies that account for genetic backgrounds and tumor phenotypes are thus necessary.
Collapse
|
23
|
Umamaheswaran G, Kadambari D, Kumar ASA, Revathy M, Anjana R, Adithan C, Dkhar SA. Polymorphic genetic variations of cytochrome P450 19A1 and T-cell leukemia 1A genes in the Tamil population. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2015; 39:102-113. [PMID: 25481307 DOI: 10.1016/j.etap.2014.10.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 10/23/2014] [Accepted: 10/27/2014] [Indexed: 06/04/2023]
Abstract
Aromatase inhibitors (AIs) are anti-neoplastic drugs widely used for the treatment of endocrine responsive breast carcinoma in postmenopausal women. Drug disposition, efficacy and tolerability of these agents are influenced by germ-line polymorphisms in the sequence of the genes encoding CYP19A1 and TCL1A proteins. In the current work, we aimed to determine the haplotype structures, linkage disequilibrium (LD) patterns, and allele and genotype frequency distribution of pharmacologically important variants from two genes (CYP19A1 and TCL1A) in Tamil population and assessed their ethnic differences. DNA derived from peripheral leukocytes of 111 healthy subjects were genotyped for 15 pharmacogenetic variants by real time thermocycler through allelic discrimination method using TaqMan 5' nuclease genotyping assay. The polymorphic variant allele frequencies of CYP19A1 were 42.3% (rs4646, T), 18% (rs10046, T), 36% (rs700519, T), 16.7% (rs700518, G), 26.1% (rs727479, G), 18% (rs4775936, T), 32% (rs10459592, G), 15.3% (rs1062033, C), 33.8% (rs749292, A), 40.1% (rs6493497, T) and 40.1% (rs7176005, G). TCL1A gene allele frequencies were 26.1% (rs7158782, G), 27% (rs7159713, G), 21.2% (rs2369049, G) and 27.5% (rs11849538, G). Comparing our data across the 5 HapMap populations (CEU, GIH, HCB, JPT and YRI) huge inter-ethnic differences were exhibited in the variant allele frequencies, LD patterns and haplotype blocks. Our results emphasize the importance of normative frequency documentation and will offer significant clinical relevance in personalizing AIs therapy.
Collapse
Affiliation(s)
- Gurusamy Umamaheswaran
- ICMR Centre for Advanced Research in Pharmacogenomics, Department of Pharmacology, Jawaharlal Institute of Postgraduate Medical Education & Research (JIPMER), Pondicherry 605006, India.
| | - Dharanipragada Kadambari
- Department of Surgery, Jawaharlal Institute of Postgraduate Medical Education & Research (JIPMER), Pondicherry 605006, India
| | - Annan Sudarsan Arun Kumar
- ICMR Centre for Advanced Research in Pharmacogenomics, Department of Pharmacology, Jawaharlal Institute of Postgraduate Medical Education & Research (JIPMER), Pondicherry 605006, India
| | - Mohan Revathy
- Department of Biotechnology and Biochemical Engineering, Sree Buddha College of Engineering, Alappuzha 690529, Kerala, India
| | - Raj Anjana
- Department of Biotechnology and Biochemical Engineering, Sree Buddha College of Engineering, Alappuzha 690529, Kerala, India
| | - Chandrasekaran Adithan
- ICMR Centre for Advanced Research in Pharmacogenomics, Department of Pharmacology, Jawaharlal Institute of Postgraduate Medical Education & Research (JIPMER), Pondicherry 605006, India
| | - Steven Aibor Dkhar
- ICMR Centre for Advanced Research in Pharmacogenomics, Department of Pharmacology, Jawaharlal Institute of Postgraduate Medical Education & Research (JIPMER), Pondicherry 605006, India
| |
Collapse
|
24
|
Lee HS, Kim KH, Hwang JS. Association of aromatase (TTTA)n repeat polymorphisms with central precocious puberty in girls. Clin Endocrinol (Oxf) 2014; 81:395-400. [PMID: 24612204 DOI: 10.1111/cen.12439] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 12/15/2013] [Accepted: 02/19/2014] [Indexed: 11/30/2022]
Abstract
OBJECTIVE Precocious puberty is characterized by early activation of the pituitary-gonadal axis. Oestrogen is the final key factor to start the onset of puberty. The cytochrome P450 19A1 (CYP19A1) gene encodes an aromatase that is responsible for the conversion of androgens to oestrogen, which is a key step in oestrogen biosynthesis. The aim of this study was to identify CYP19A1 gene mutations or polymorphisms in girls with central precocious puberty (CPP). METHODS We evaluated the frequency of allelic variants of the CYP19A1 exons and the tetranucleotide tandem repeat (TTTA)n in intron 4 in 203 idiopathic central precocious puberty (CPP) girls and 101 normal healthy women. RESULTS The genotype analysis of the CYP19A1 (TTTA)n polymorphism revealed six different alleles ranging from seven to 13 repeats. Among the six different repeat alleles detected in this study, the (TTTA)₁₃ repeat allele was only detected in the patient group and carriers of the (TTTA)₁₃ allele were significantly associated with an increased risk of CPP (OR = 1·509, 95% CI = 1·425-1·598, P = 0·033). Carriers of the (TTTA)₁₃ repeat allele were significantly younger at pubertal onset and had higher levels of oestrogen than noncarriers of the (TTTA)₁₃ repeat allele. Although nine polymorphisms were detected in exons of the CYP19A1 gene, no clinical significance was observed. CONCLUSION In this study, carriers of a higher repeat (TTTA)₁₃ polymorphism in intron 4 of the CYP19A1 gene had higher levels of oestrogen. Those carrying the (TTTA)₁₃ repeat allele may have a higher risk of developing CPP.
Collapse
Affiliation(s)
- Hae Sang Lee
- Department of Pediatrics, Ajou University School of Medicine, Ajou University Hospital, Suwon, Korea
| | | | | |
Collapse
|
25
|
Analysis of the rs10046 polymorphism of aromatase (CYP19) in premenopausal onset of human breast cancer. Int J Mol Sci 2014; 15:712-24. [PMID: 24402127 PMCID: PMC3907833 DOI: 10.3390/ijms15010712] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 12/23/2013] [Accepted: 12/25/2013] [Indexed: 12/04/2022] Open
Abstract
The CYP19 gene encodes aromatase, an enzyme catalyzing the conversion of androgens to estrogens. Studies analyzing associations between single nucleotide polymorphisms in CYP19 and breast cancer risk have shown inconsistent results. The rs10046 polymorphism is located in the 3′ untranslated region of the CYP19 gene, but the influence of this polymorphism on breast cancer risk is unclear. In this study, we investigated the impact of rs10046 SNP on breast cancer risk, age at onset and association with clinical characteristics in an Austrian population of 274 breast cancer patients and 253 controls. The results show that a significantly increased fraction of patients with the TT genotype of rs10046 develop breast cancer under the age of 50 (41.8% of TT patients, compared to 26.6% of C carriers; p = 0.018, Chi-square test). No rs10046 genotypes were significantly associated with increased breast cancer risk or patient characteristics other than age at onset. These results suggest that the rs10046 polymorphism in the CYP19 gene may have an effect on breast cancer susceptibility at an age under 50 in the investigated population.
Collapse
|
26
|
Umamaheswaran G, Dkhar SA, Kalaivani S, Anjana R, Revathy M, Jaharamma M, Shree KML, Kadambari D, Adithan C. Haplotype structures and functional polymorphic variants of the drug target enzyme aromatase (CYP19A1) in South Indian population. Med Oncol 2013; 30:665. [PMID: 23893151 DOI: 10.1007/s12032-013-0665-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Accepted: 07/12/2013] [Indexed: 10/26/2022]
Abstract
CYP19A1 gene product aromatase (CYP19A1) is a 58-kDa protein and belongs to the member of the cytochrome P450 superfamily, which facilitates the bioconversion of estrogens from androgens. Single-nucleotide polymorphisms (SNPs) of CYP19A1 affect the activity of the enzyme and have been implicated in the association of estrogen-dependent disease, prognosis, therapeutic efficacy, and toxicity of third-generation aromatase inhibitors (AIs). Based on ethnicity, the frequency distribution of CYP19A1 alleles will differ, and until now, no data are available for Indians. Using qRT-PCR with TaqMan assays, the frequencies of functionally important polymorphic variants of CYP19A1 gene were determined in 163 healthy subjects of South Indian origin. The observed frequencies of the CYP19A1 minor alleles for the SNPs rs4646 (T), rs10046 (T), rs700519 (T), rs700518 (G), rs727479 (G), rs4775936 (T), rs10459592 (G), rs749292 (A), rs6493497 (T), and rs7176005 (A) are 41.1 (35.8-46.4), 20.0 (15.6-24.3), 33.7 (28.6-38.9), 17.8 (13.6-21.9), 25.8 (21.0-30.5), 19.9 (15.6-24.3), 33.7 (28.6-38.9), 24.9 (20.2-29.5), 35.9 (30.7-41.1), and 35.9 (30.7-41.1), respectively. Strong linkage disequilibrium existed between CYP19A1 SNPs, and sixteen different haplotype structures with a frequency >1% were derived from all the 10 SNPs tested. The most common being the haplotype (H1) GCTATCTGTG with a frequency of about 17.8%. Gender-specific assessment showed significant difference in the allele frequency for rs749292 (p < 0.04), and greater inter-ethnic variation was detected in the distribution of CYP19A1 variants except for rs727479. Our results could provide preliminary insight for further pharmacogenetic investigations of AIs as well as for subsequent molecular epidemiological studies on the contribution of these variants to the occurrence and development of estrogen-dependent disease in South Indians.
Collapse
Affiliation(s)
- Gurusamy Umamaheswaran
- ICMR Centre for Advanced Research in Pharmacogenomics, Department of Pharmacology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Pondicherry 605006, India.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Westbrook K, Stearns V. Pharmacogenomics of breast cancer therapy: an update. Pharmacol Ther 2013; 139:1-11. [PMID: 23500718 DOI: 10.1016/j.pharmthera.2013.03.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 02/19/2013] [Indexed: 12/13/2022]
Abstract
Clinical and histopathologic characteristics of breast cancer have long played an important role in treatment decision-making. Well-recognized prognostic factors include tumor size, node status, presence or absence of metastases, tumor grade, and hormone receptor expression. High tumor grade, presence of hormone receptors, and HER2-positivity are a few predictive markers of response to chemotherapy, endocrine manipulations, and anti-HER2 agents, respectively. However, there is much heterogeneity of outcomes in patients with similar clinical and pathologic features despite equivalent treatment regimens. Some of the differences in response to specific therapies can be attributed to somatic tumor characteristics, such as degree of estrogen receptor expression and HER2 status. In recent years, there has been great interest in evaluating the role that pharmacogenetics/pharmacogenomics, or variations in germline DNA, play in alteration of drug metabolism and activity, thus leading to disparate outcomes among patients with similar tumor characteristics. The utility of these variations in treatment decision-making remains debated. Here we review the data available to date on genomic variants that may influence response to drugs commonly used to treat breast cancer. While none of the variants reported to date have demonstrated clinical utility, ongoing prospective studies and increasing understanding of pharmacogenetics will allow us to better predict risk of toxicity or likelihood of response to specific treatments and to provide a more personalized therapy.
Collapse
Affiliation(s)
- Kelly Westbrook
- Duke University Medical Center, Duke Cancer Institute, Breast Cancer Program, DUMC Box 3893, 10 Searle Dr., Sealy Mudd Bldg. Room 449A, Durham, NC 27710, United States.
| | | |
Collapse
|
28
|
Identification of candidate polymorphisms on stress oxidative and DNA damage repair genes related with clinical outcome in breast cancer patients. Int J Mol Sci 2012; 13:16500-13. [PMID: 23443115 PMCID: PMC3546704 DOI: 10.3390/ijms131216500] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Revised: 11/23/2012] [Accepted: 11/27/2012] [Indexed: 11/16/2022] Open
Abstract
Diverse polymorphisms have been associated with the predisposition to develop cancer. On fewer occasions, they have been related to the evolution of the disease and to different responses to treatment. Previous studies of our group have associated polymorphisms on genes related to oxidative stress (rs3736729 on GCLC and rs207454 on XDH) and DNA damage repair (rs1052133 on OGG1) with a predisposition to develop breast cancer. In the present work, we have evaluated the hypothesis that these polymorphisms also play a role in a patient's survival. A population-based cohort study of 470 women diagnosed with primary breast cancer and a median follow up of 52.44 months was conducted to examine the disease-free and overall survival in rs3736729, rs207454 and rs1052133 genetic variants. Adjusted Cox regression analysis was used to that end. The Kaplan-Meier analysis shows that rs3736729 on GCLC presents a significant association with disease-free survival and overall survival. The polymorphisms rs1052133 on OGG1 and rs207454 on XDH show a trend of association with overall survival. The analysis based on hormonal receptor status revealed a stronger association. The CC genotype on rs207454 (XDH) was significantly associated with lower time of disease free survival (p = 0.024) in progesterone receptor negative (PGR-) patients and rs3736729 (GCLC) was significantly associated with disease free survival (p = 0.001) and overall survival (p = 0.012) in the subgroup of estrogen receptor negative (ER-) patients. This work suggests that unfavorable genetic variants in the rs207454 (XDH) and rs3736729 (GCLC) polymorphisms may act as predictors of the outcome in negative progesterone receptor and negative estrogen receptor breast cancer patients, respectively.
Collapse
|
29
|
Plasma estrone sulfate concentrations and genetic variation at the CYP19A1 locus in postmenopausal women with early breast cancer treated with letrozole. Breast Cancer Res Treat 2012; 137:167-74. [PMID: 23129173 PMCID: PMC3528956 DOI: 10.1007/s10549-012-2306-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Accepted: 10/15/2012] [Indexed: 11/04/2022]
Abstract
Estrogen synthesis suppression induced by aromatase inhibitors in breast cancer (BC) patients may be affected by single nucleotide polymorphisms (SNPs) of the gene encoding aromatase enzyme, CYP19A1. We assessed the association between plasma estrone sulfate (ES), letrozole treatment, and four SNPs of CYP19A1 gene (rs10046 C>T, rs4646 G>T, rs749292 C>T, rs727479 T>G) which seem to be related to circulating estrogen levels. Patients were enrolled into a prospective, Italian multi-center clinical trial (Gruppo Italiano Mammella, GIM-5) testing the association of CYP19A1 SNPs with the efficacy of letrozole adjuvant therapy, in postmenopausal early BC patients. SNPs were identified from peripheral blood cell DNA. Plasma ES concentrations were evaluated by Radio Immuno Assay. Blood samples were obtained immediately before letrozole therapy (N = 204), at 6-weeks (N = 178), 6 (N = 152) and 12-months (N = 136) during treatment. Medians (IQR) of ES were 160 pg/mL (85–274) at baseline, 35 pg/mL (12–64) at 6-weeks, 29 pg/mL (17–48) at 6 months and 25 pg/mL (8–46) after 12 months treatment. No statistically significant association was evident between polymorphisms and ES circulating levels during letrozole therapy. Letrozole suppression of the aromatase enzyme function is not affected by polymorphisms of CYP19A1 gene in postmenopausal BC patients.
Collapse
|
30
|
|