1
|
Taheri H, Ahmed E, Hu P, Sparreboom A, Hu S. Determination and Disposition of the Aromatase Inhibitor Exemestane in CYP3A-Deficient Mice. Molecules 2025; 30:1440. [PMID: 40286062 PMCID: PMC11990089 DOI: 10.3390/molecules30071440] [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: 01/22/2025] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 04/29/2025] Open
Abstract
Exemestane, a steroidal aromatase inhibitor prescribed for post-menopausal women with estrogen receptor-positive breast cancer, is associated with debilitating musculoskeletal side effects and exhibits considerable interindividual variability in systemic exposure. Although exemestane is metabolized mainly by cytochrome P-450 3A4, the contribution of this metabolic pathway to the elimination of exemestane and its drug-drug interaction liabilities remains uncertain. Here, we developed a novel quantification method for exemestane and applied it to evaluate the role of CYP3A-mediated metabolism in the pharmacokinetics of exemestane using wild-type and Cyp3a-deficient mice. Liquid chromatography-mass spectrometry was used to quantify exemestane in selective reaction monitoring (SRM) mode, in which precursor ion and fragment ion data were obtained simultaneously. Validation results demonstrated that the developed method was accurate and precise, and sufficiently sensitive to be applied to murine pharmacokinetic studies involving serial blood sampling strategies. Although in vitro studies indicate that exemestane undergoes extensive metabolism in the liver to inactive metabolites by CYP3A4, complete Cyp3a deficiency in mice did not influence the systemic exposure to exemestane. This unequivocal evidence from genetic approaches using preclinical mouse models confirms that the potential for such DDI liabilities is very low. Our newly developed method provides a robust platform for further pharmacokinetic studies with exemestane in mice to delineate DDI liabilities and define the mechanisms of elimination.
Collapse
Affiliation(s)
| | | | | | | | - Shuiying Hu
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; (H.T.); (E.A.); (P.H.); (A.S.)
| |
Collapse
|
2
|
Taheri H, Li Y, Huang KM, Ahmed E, Jin Y, Drabison T, Yang Y, Kulp SK, Young NA, Li J, Cheng X, Corps KN, Coss CC, Vaughn JE, Lustberg MB, Sparreboom A, Hu S. OATP1B-type Transport Function Is a Determinant of Aromatase Inhibitor-Associated Arthralgia Susceptibility. CANCER RESEARCH COMMUNICATIONS 2025; 5:496-510. [PMID: 40062557 PMCID: PMC11948302 DOI: 10.1158/2767-9764.crc-24-0475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 01/10/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025]
Abstract
SIGNIFICANCE AIs are effective but often discontinued because of arthralgia. This study explores the role of OATP1B transporters in AI-related side effects and the potential usage of transporter biomarkers to predict and reduce the risk of arthralgia associated with AI treatment.
Collapse
Affiliation(s)
- Hanieh Taheri
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Yang Li
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Kevin M. Huang
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Eman Ahmed
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Yan Jin
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Thomas Drabison
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Yan Yang
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Samuel K. Kulp
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Nicholas A. Young
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Junan Li
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Xiaolin Cheng
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Kara N. Corps
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio
| | - Christopher C. Coss
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Jennifer E. Vaughn
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio
| | - Maryam B. Lustberg
- Yale Comprehensive Cancer Center, Yale School of Medicine, New Haven, Connecticut
| | - Alex Sparreboom
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Shuiying Hu
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| |
Collapse
|
3
|
Taheri H, Jin Y, Ahmed E, Hu P, Li Y, Sparreboom A, Hu S. Quantification of the aromatase inhibitor letrozole and its carbinol metabolite in mouse plasma by UHPLC-MS/MS. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1238:124106. [PMID: 38636136 DOI: 10.1016/j.jchromb.2024.124106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 03/17/2024] [Accepted: 03/25/2024] [Indexed: 04/20/2024]
Abstract
A liquid chromatography - electrospray ionization-mass spectrometry (LC-ESI-MS) method was developed for the quantification of letrozole, a third-generation aromatase inhibitor, and its main carbinol metabolite (CM) in support of murine pharmacokinetic studies. Using polarity switching, simultaneous ESI-MS measurement of letrozole and CM was achieved in positive and negative mode, respectively. The assay procedure involved a one-step protein precipitation and extraction of all analytes from mouse plasma requiring only 5 μL of sample. Separation was optimized on an Accucore aQ column with gradient elution at a flow rate of 0.4 mL/min in 5 min. Two calibration curves per day over four consecutive measurement days showed satisfactory linear responses (r2 > 0.99) over concentration ranges of 5-1000 ng/mL and 20-2000 ng/mL for letrozole and CM, respectively. No matrix effect was found, and the mean extraction recoveries were 103-108 % for letrozole and 99.8-107 % for CM. Precision and accuracy within a single run and over four consecutive measurement days were verified to be within acceptable limits. Application of the developed method to preclinical pharmacokinetic studies in mice receiving oral letrozole at a dose 1 or 10 mg/kg revealed that the systemic exposure to letrozole was dose-, formulation-, and strain-dependent. These findings may inform the future design of preclinical studies aimed at refining the pharmacological profile of this clinically important drug.
Collapse
Affiliation(s)
- Hanieh Taheri
- Division of Outcomes and Translational Sciences, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Yan Jin
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Eman Ahmed
- Division of Outcomes and Translational Sciences, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Peng Hu
- Division of Outcomes and Translational Sciences, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Yang Li
- Division of Outcomes and Translational Sciences, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Alex Sparreboom
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Shuiying Hu
- Division of Outcomes and Translational Sciences, College of Pharmacy, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
4
|
Michalska K, Balcerczak E, Jeleń A, Saed L, Pietrzak J, Żebrowska-Nawrocka M. Effects of the SLCO1B1 A388G single nucleotide polymorphism on the development, clinical parameters, treatment, and survival of multiple myeloma cases in a Polish population. Mol Biol Rep 2023; 50:1447-1458. [PMID: 36478296 PMCID: PMC9889417 DOI: 10.1007/s11033-022-08162-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Multiple myeloma is one of the most common hematological malignancies worldwide. Genetic alterations may lead to the progression from monoclonal gammopathy to multiple myeloma. Additionally, the genetic background of the disease might influence therapy outcomes, including survival time. SLCO1B1, belonging to the OATPs family, is a membrane protein that mediates the uptake of a wide range of endogenous and exogenous (including drugs) compounds. METHODS AND RESULTS In this study, the A388G single nucleotide polymorphism in the SLCO1B1 gene in Polish multiple myeloma patients was determined. This polymorphism affects the amino acid change of the protein, so it may be responsible for treatment effectiveness or risk of disease development. A388G was evaluated by the PCR-RFLP method. The presented study showed a statistically significant association between the GG genotype with longer survival of patients with multiple myeloma with Melphalan-Prednisone therapy compared to other treatment regimens (p = 0.0271). There was no statistically significant association in the frequency of genotypes (p = 0.8211) and alleles: allele A (p = 0.5442); allele G (p = 0.8020) between multiple myeloma patients and a control group. CONCLUSIONS The A388G polymorphism does not seem to affect the increased risk of the development of multiple myeloma. However, the occurrence of the GG genotype may prolong of patients overall survival in the case of Melphalan-Prednisone therapy.
Collapse
Affiliation(s)
- Katarzyna Michalska
- Laboratory of Molecular Diagnostics and Pharmacogenomics, Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
| | - Ewa Balcerczak
- Laboratory of Molecular Diagnostics and Pharmacogenomics, Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
| | - Agnieszka Jeleń
- Laboratory of Molecular Diagnostics and Pharmacogenomics, Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
| | - Lias Saed
- Laboratory of Molecular Diagnostics and Pharmacogenomics, Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
| | - Jacek Pietrzak
- Laboratory of Molecular Diagnostics and Pharmacogenomics, Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
| | - Marta Żebrowska-Nawrocka
- Laboratory of Molecular Diagnostics and Pharmacogenomics, Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Interfaculty Cathedral of Laboratory and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1, 90-151 Lodz, Poland
| |
Collapse
|
5
|
Božina T, Ganoci L, Karačić E, Šimičević L, Vrkić-Kirhmajer M, Klarica-Domjanović I, Križ T, Sertić Z, Božina N. ABCG2 and SLCO1B1 gene polymorphisms in the Croatian population. Ann Hum Biol 2022; 49:323-331. [PMID: 36382878 DOI: 10.1080/03014460.2022.2140826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Organic anion-transporting polypeptide 1B1 (OATP1B1) and the ATP-binding cassette subfamily G member 2, ABCG2, are important transporters involved in the transport of endogenous substrates and xenobiotics, including drugs. Genetic polymorphisms of these transporters have effect on transporter activity. There is significant interethnic variability in the frequency of allele variants. AIM To determined allele and genotype frequencies of ABCG2 and SLCO1B1 genes in Croatian populations of European descent. SUBJECTS AND METHODS A total of 905 subjects (482 women) were included. Genotyping for ABCG2 c.421C > A (rs2231142) and for SLCO1B1 c.521T > C (rs4149056), was performed by real-time polymerase chain reaction (PCR) using TaqMan® DME Genotyping Assays. RESULTS For ABCG2 c.421C > A, the frequency of CC, CA and AA genotypes was 81.4%, 17.8% and 0.8% respectively. The frequency of variant ABCG2 421 A allele was 9.7%. For SLCO1B1 c.521T > C, the frequency of TT, TC and CC genotypes was 61.7%, 34.8% and 3.5% respectively. The frequency of variant SLCO1B1 521 C allele was 20.9%. CONCLUSION The frequency of the ABCG2 and SLCO1B1 allelic variants and genotypes in the Croatian population is in accordance with other European populations. Pharmacogenetic analysis can serve to individualise drug therapy and minimise the risk of developing adverse drug reactions.
Collapse
Affiliation(s)
- Tamara Božina
- Department of Medical Chemistry, Biochemistry, and Clinical Chemistry, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Lana Ganoci
- Division of Pharmacogenomics and Therapy Individualization, Department of Laboratory Diagnostics, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Ena Karačić
- Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| | - Livija Šimičević
- Division of Pharmacogenomics and Therapy Individualization, Department of Laboratory Diagnostics, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Majda Vrkić-Kirhmajer
- Department of Cardiovascular Diseases Zagreb, University of Zagreb School of Medicine, University Hospital Centre Zagreb, Croatia
| | | | - Tena Križ
- Department of Ophthalmology, University Hospital Centre "Sestre milosrdnice", Zagreb, Croatia
| | - Zrinka Sertić
- Department of Emergency Medicine, University Hospital Centre Zagreb, Zagreb, Croatia
| | - Nada Božina
- Department of Pharmacology, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
6
|
Anabtawi N, Drabison T, Hu S, Sparreboom A, Talebi Z. The role of OATP1B1 and OATP1B3 transporter polymorphisms in drug disposition and response to anticancer drugs: a review of the recent literature. Expert Opin Drug Metab Toxicol 2022; 18:459-468. [PMID: 35983889 DOI: 10.1080/17425255.2022.2113380] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Members of the solute carrier family of organic anion transporting polypeptides are responsible for the cellular uptake of a broad range of endogenous compounds and xenobiotics in multiple tissues. In particular, the polymorphic transporters OATP1B1 and OATP1B3 are highly expressed in the liver and have been identified as critical regulators of hepatic eliminaton. As these transporters are also expressed in cancer cells, the function alteration of these proteins have important consequences for an individual's susceptibility to certain drug-induced side effects, drug-drug interactions, and treatment efficacy. AREAS COVERED In this mini-review, we provide an update of this rapidly emerging field, with specific emphasis on the direct contribution of genetic variants in OATP1B1 and OATP1B3 to the transport of anticancer drugs, the role of these carriers in regulation of their disposition and toxicity profiles, and recent advances in attempts to integrate information on transport function in patients to derive individualized treatment strategies. EXPERT OPINION Based on currently available data, it appears imperative that different aspects of disease, physiology, and drugs of relevance should be evaluated along with an individual's genetic signature, and that tools such as biomarker levels can be implemented to achieve the most reliable prediction of clinically relevant pharmacodynamic endpoints.
Collapse
Affiliation(s)
- Nadeen Anabtawi
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Thomas Drabison
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Shuiying Hu
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio.,Division of Outcomes and Translational Sciences, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Alex Sparreboom
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| | - Zahra Talebi
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio
| |
Collapse
|
7
|
Roncato R, Gerratana L, Palmero L, Gagno S, Poetto AS, Peruzzi E, Zanchetta M, Posocco B, De Mattia E, Canil G, Alberti M, Orleni M, Toffoli G, Puglisi F, Cecchin E. An Integrated Pharmacological Counselling Approach to Guide Decision-Making in the Treatment with CDK4/6 Inhibitors for Metastatic Breast Cancer. Front Pharmacol 2022; 13:897951. [PMID: 35942220 PMCID: PMC9356076 DOI: 10.3389/fphar.2022.897951] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
A wide inter-individual variability in the therapeutic response to cyclin-dependent kinases 4 and 6 inhibitors (CDKis) has been reported. We herein present a case series of five patients treated with either palbociclib or ribociclib referred to our clinical pharmacological counselling, including therapeutic drug monitoring (TDM), pharmacogenetics, and drug–drug interaction analysis to support clinicians in the management of CDKis treatment for metastatic breast cancer. Patients’ plasma samples for TDM analysis were collected at steady state and analyzed by an LC-MS/MS method for minimum plasma concentration (Cmin) evaluation. Under and overexposure to the drug were defined based on the mean Cmin values observed in population pharmacokinetic studies. Polymorphisms in selected genes encoding for proteins involved in drug absorption, distribution, metabolism, and elimination were analyzed (CYP3A4, CYP3A5, ABCB1, SLCO1B1, and ABCG2). Three of the five reported cases presented a CDKi plasma level above the population mean value and were referred for toxicity. One of them presented a low function ABCB1 haplotype (ABCB1-rs1128503, rs1045642, and rs2032582), possibly causative of both increased drug oral absorption and plasmatic concentration. Two patients showed underexposure to CDKis, and one of them was referred for early progression. In one patient, a CYP3A5*1/*3 genotype was found to be potentially responsible for more efficient drug metabolism and lower drug plasma concentration. This intensified pharmacological approach in clinical practice has been shown to be potentially effective in supporting prescribing oncologists with dose and drug selection and could be ultimately useful for increasing both the safety and efficacy profiles of CDKi treatment.
Collapse
Affiliation(s)
- Rossana Roncato
- Experimental and Clinical Pharmacology Unit-CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
- *Correspondence: Rossana Roncato, ; Sara Gagno,
| | - Lorenzo Gerratana
- Department of Medical Oncology-CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| | - Lorenza Palmero
- Department of Medical Oncology-CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
- Department of Medicine (DAME), University of Udine, Udine, Italy
| | - Sara Gagno
- Experimental and Clinical Pharmacology Unit-CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
- *Correspondence: Rossana Roncato, ; Sara Gagno,
| | - Ariana Soledad Poetto
- Experimental and Clinical Pharmacology Unit-CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| | - Elena Peruzzi
- Experimental and Clinical Pharmacology Unit-CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| | - Martina Zanchetta
- Experimental and Clinical Pharmacology Unit-CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| | - Bianca Posocco
- Experimental and Clinical Pharmacology Unit-CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| | - Elena De Mattia
- Experimental and Clinical Pharmacology Unit-CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| | - Giovanni Canil
- Experimental and Clinical Pharmacology Unit-CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| | - Martina Alberti
- Department of Medical Oncology-CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
- Department of Medicine (DAME), University of Udine, Udine, Italy
| | - Marco Orleni
- Experimental and Clinical Pharmacology Unit-CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit-CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| | - Fabio Puglisi
- Department of Medical Oncology-CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
- Department of Medicine (DAME), University of Udine, Udine, Italy
| | - Erika Cecchin
- Experimental and Clinical Pharmacology Unit-CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy
| |
Collapse
|
8
|
Daniyal A, Santoso I, Gunawan NHP, Barliana MI, Abdulah R. Genetic Influences in Breast Cancer Drug Resistance. BREAST CANCER (DOVE MEDICAL PRESS) 2021; 13:59-85. [PMID: 33603458 PMCID: PMC7882715 DOI: 10.2147/bctt.s284453] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/12/2021] [Indexed: 12/25/2022]
Abstract
Breast cancer is the most common cancer in adult women aged 20 to 50 years. The therapeutic regimens that are commonly recommended to treat breast cancer are human epidermal growth factor receptor 2 (HER2) targeted therapy, endocrine therapy, and systemic chemotherapy. The selection of pharmacotherapy is based on the characteristics of the tumor and its hormone receptor status, specifically, the presence of HER2, progesterone receptors, and estrogen receptors. Breast cancer pharmacotherapy often gives different results in various populations, which may cause therapeutic failure. Different types of congenital drug resistance in individuals can cause this. Genetic polymorphism is a factor in the occurrence of congenital drug resistance. This review explores the relationship between genetic polymorphisms and resistance to breast cancer therapy. It considers studies published from 2010 to 2020 concerning the relationship of genetic polymorphisms and breast cancer therapy. Several gene polymorphisms are found to be related to longer overall survival, worse relapse-free survival, higher pathological complete response, and increased disease-free survival in breast cancer patients. The presence of these gene polymorphisms can be considered in the treatment of breast cancer in order to shape personalized therapy to yield better results.
Collapse
Affiliation(s)
- Adhitiya Daniyal
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Indonesia
| | - Ivana Santoso
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Indonesia
| | - Nadira Hasna Putri Gunawan
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Indonesia
| | - Melisa Intan Barliana
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Jatinangor, Indonesia
- Department of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Indonesia
| | - Rizky Abdulah
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Indonesia
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Jatinangor, Indonesia
| |
Collapse
|
9
|
Zhu Y, Koleck TA, Bender CM, Conley YP. Genetic Underpinnings of Musculoskeletal Pain During Treatment With Aromatase Inhibitors for Breast Cancer: A Biological Pathway Analysis. Biol Res Nurs 2019; 22:263-276. [PMID: 31847542 DOI: 10.1177/1099800419895114] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
BACKGROUND Musculoskeletal pain (MSKP) is the most reported symptom during treatment with aromatase inhibitors (AIs) for breast cancer. The mechanisms underlying MSKP are multidimensional and not well understood. The goals of this biological pathway analysis were to (1) gain an understanding of the genetic variation and biological mechanisms underlying MSKP with AI therapy and (2) identify plausible biological pathways and candidate genes for future investigation. METHOD Genes associated with MSKP during AI therapy or genes involved in drug metabolism of and response to AIs were identified from the literature. Studies published through February 2019 were queried in PubMed®. The genes identified from the literature were entered into QIAGEN's Ingenuity® Pathway Analysis (IPA) software to generate canonical pathways, upstream regulators, and networks through a core analysis. RESULTS The 17 genes identified were ABCB1, ABCG1, CYP17A1, CYP19A1, CYP27B1, CYP2A6, CYP3A4, CYP3A5, ESR1, OATP1B1, OPG, RANKL, SLCO3A1, TCL1A, UGT2A1, UGT2B17, and VDR. These genes are involved in encoding bone-remodeling regulators, drug-metabolizing enzymes (cytochrome P450 family, UDP-glucuronosyltransferases family), or drug transporters (ATP-binding cassette transporters, organic anion transporters). Multiple plausible biological pathways (e.g., nicotine degradation, melatonin degradation) and candidate genes (e.g., NFKB, HSP90, AKT, ERK1/2, FOXA2) are proposed for future investigation based on the IPA results. CONCLUSION Multiple genes and molecular-level etiologies may contribute to MSKP with AI therapy in women with breast cancer. Our innovative combination of gene identification from the literature plus biological pathway analysis allowed for the emergence of novel candidate genes and biological pathways for future investigations.
Collapse
Affiliation(s)
- Yehui Zhu
- School of Nursing, University of Pittsburgh, PA, USA
| | | | | | | |
Collapse
|
10
|
Dempsey JM, Kidwell KM, Gersch CL, Pesch AM, Desta Z, Storniolo AM, Stearns V, Skaar TC, Hayes DF, Henry NL, Rae JM, Hertz DL. Effects of SLCO1B1 polymorphisms on plasma estrogen concentrations in women with breast cancer receiving aromatase inhibitors exemestane and letrozole. Pharmacogenomics 2019; 20:571-580. [PMID: 31190621 PMCID: PMC6891932 DOI: 10.2217/pgs-2019-0020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 03/15/2019] [Indexed: 12/31/2022] Open
Abstract
Aim: This study tested for associations between SLCO1B1 polymorphisms and circulating estrogen levels in women with breast cancer treated with letrozole or exemestane. Patients & methods: Postmenopausal women with hormone-receptor positive breast cancer were genotyped for SLCO1B1*5 (rs4149056) and rs10841753. Pretreatment and on-treatment plasma estrogens and aromatase inhibitor (AI) concentrations were measured. Regression analyses were performed to test for pharmacogenetic associations with estrogens and drug concentrations. Results:SLCO1B1*5 was associated with elevated pretreatment estrone sulfate and an increased risk of detectable estrone concentrations after 3 months of AI treatment. Conclusion: These findings suggest SLCO1B1 polymorphisms may have an effect on estrogenic response to AI treatment, and therefore may adversely impact the anticancer effectiveness of these agents.
Collapse
Affiliation(s)
- Jacqueline M Dempsey
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI 48109-1065, USA
| | - Kelley M Kidwell
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Christina L Gersch
- Department of Internal Medicine, Division of Hematology/Oncology, Medical School, University of Michigan, Ann Arbor, MI 48109, USA
| | - Andrea M Pesch
- Department of Internal Medicine, Division of Hematology/Oncology, Medical School, University of Michigan, Ann Arbor, MI 48109, USA
| | - Zeruesenay Desta
- Department of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | | | - Vered Stearns
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21231, USA
| | - Todd C Skaar
- Department of Medicine, Indiana University, Indianapolis, IN 46202, USA
| | - Daniel F Hayes
- Department of Internal Medicine, Division of Hematology/Oncology, Medical School, University of Michigan, Ann Arbor, MI 48109, USA
| | - N Lynn Henry
- Department of Internal Medicine, Division of Hematology/Oncology, Medical School, University of Michigan, Ann Arbor, MI 48109, USA
| | - James M Rae
- Department of Internal Medicine, Division of Hematology/Oncology, Medical School, University of Michigan, Ann Arbor, MI 48109, USA
| | - Daniel L Hertz
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI 48109-1065, USA
| |
Collapse
|
11
|
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
|