1
|
Abbes H, Soria-Chacartegui P, Omezzine A, Abad-Santos F, Zubiaur P. Impact of Genetic Variation in Adrenergic Receptors on β-Blocker Effectiveness and Safety in Cardiovascular Disease Management: A Systematic Review. Pharmaceuticals (Basel) 2025; 18:493. [PMID: 40283930 PMCID: PMC12030580 DOI: 10.3390/ph18040493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/21/2025] [Accepted: 03/26/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: A systematic review was conducted to compile all the evidence on the impact of ADRB1 and ADRB2 genetic variants on the response to β-blockers, used for the management of cardiovascular diseases. Methods: After searching in PubMed, PharmGKB, and the Cochrane Central Register of Controlled Trials including terms related to these drugs, genes, and pathologies, 1182 articles were retrieved, 29 of which met the inclusion criteria. A β-adrenoreceptor (ADRB) blockade qualitative variable was inferred for all the associations between genetic variants and clinical phenotypes. Results: The relationship between ADRB1 rs1801253 (G>C) C allele and higher receptor blockade showed a moderate overall level of evidence, reaching a high level on its relationship with higher reduction in the systolic (SBP) and diastolic blood pressure and heart rate (HR). The relationship between ADRB1 rs1801252 (A>G) G allele and lower receptor blockade reached an overall high level of evidence, considering its impact on the reduction in the SBP, HR, left ventricular end-diastolic diameter, and incidence of major cardiovascular events. The relationship between ADRB2 rs1042714 (G>C) C allele and lower receptor blockade reached a moderate overall level of evidence due to its impact on HR, pulmonary wedge pressure, and left ventricular ejection fraction response. The ADRB2 rs1042713 (G>A) A allele was associated with higher receptor blockade and higher HR reduction with a low level of evidence. Conclusions: The genotyping of both ADRB1 variants may be clinically useful; further investigation is required on the relevance of both ADRB2 variants. Further research is warranted to determine the clinical usefulness of ADRB preemptive genotyping.
Collapse
Affiliation(s)
- Houwaida Abbes
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IP), Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Biochemistry Department, LR12SP11, Sahloul University Hospital, Sousse 4054, Tunisia
- Faculty of Pharmacy of Monastir, University of Monastir, Monastir 5000, Tunisia
| | - Paula Soria-Chacartegui
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IP), Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
| | - Asma Omezzine
- Biochemistry Department, LR12SP11, Sahloul University Hospital, Sousse 4054, Tunisia
- Faculty of Pharmacy of Monastir, University of Monastir, Monastir 5000, Tunisia
| | - Francisco Abad-Santos
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IP), Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Pablo Zubiaur
- Clinical Pharmacology Department, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria La Princesa (IP), Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
| |
Collapse
|
2
|
Duarte JD, Thomas CD, Lee CR, Huddart R, Agundez JAG, Baye JF, Gaedigk A, Klein TE, Lanfear DE, Monte AA, Nagy M, Schwab M, Stein CM, Uppugunduri CRS, van Schaik RHN, Donnelly RS, Caudle KE, Luzum JA. Clinical Pharmacogenetics Implementation Consortium Guideline (CPIC) for CYP2D6, ADRB1, ADRB2, ADRA2C, GRK4, and GRK5 Genotypes and Beta-Blocker Therapy. Clin Pharmacol Ther 2024; 116:939-947. [PMID: 38951961 PMCID: PMC11502236 DOI: 10.1002/cpt.3351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/30/2024] [Indexed: 07/03/2024]
Abstract
Beta-blockers are widely used medications for a variety of indications, including heart failure, myocardial infarction, cardiac arrhythmias, and hypertension. Genetic variability in pharmacokinetic (e.g., CYP2D6) and pharmacodynamic (e.g., ADRB1, ADRB2, ADRA2C, GRK4, GRK5) genes have been studied in relation to beta-blocker exposure and response. We searched and summarized the strength of the evidence linking beta-blocker exposure and response with the six genes listed above. The level of evidence was high for associations between CYP2D6 genetic variation and both metoprolol exposure and heart rate response. Evidence indicates that CYP2D6 poor metabolizers experience clinically significant greater exposure and lower heart rate in response to metoprolol compared with those who are not poor metabolizers. Therefore, we provide therapeutic recommendations regarding genetically predicted CYP2D6 metabolizer status and metoprolol therapy. However, there was insufficient evidence to make therapeutic recommendations for CYP2D6 and other beta-blockers or for any beta-blocker and the other five genes evaluated (updates at www.cpicpgx.org).
Collapse
Affiliation(s)
- Julio D. Duarte
- Department of Pharmacotherapy and Translational ResearchUniversity of Florida College of PharmacyGainesvilleFloridaUSA
- Center for Pharmacogenomics and Precision MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Cameron D. Thomas
- Department of Pharmacotherapy and Translational ResearchUniversity of Florida College of PharmacyGainesvilleFloridaUSA
- Center for Pharmacogenomics and Precision MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Craig R. Lee
- Division of Pharmacotherapy and Experimental TherapeuticsUniversity of North Carolina Eshelman School of PharmacyChapel HillNorth CarolinaUSA
| | - Rachel Huddart
- Department of Biomedical Data ScienceStanford UniversityStanfordCaliforniaUSA
| | - Jose A. G. Agundez
- Institute of Molecular Pathology BiomarkersUniversity of ExtremaduraCáceresSpain
| | - Jordan F. Baye
- Department of Pharmacy PracticeSouth Dakota State University College of Pharmacy & Allied Health ProfessionsBrookingsSouth DakotaUSA
- Sanford ImageneticsSioux FallsSouth DakotaUSA
| | - Andrea Gaedigk
- Division of Clinical Pharmacology, Toxicology & Therapeutic InnovationChildren's Mercy Research Institute and School of Medicine, University of Missouri‐Kansas CityKansas CityMissouriUSA
| | - Teri E. Klein
- Department of Biomedical Data ScienceStanford UniversityStanfordCaliforniaUSA
| | - David E. Lanfear
- Center for Individualized and Genomic Medicine Research (CIGMA), Henry Ford HospitalDetroitMichiganUSA
- Heart and Vascular Institute, Henry Ford HealthDetroitMichiganUSA
| | - Andrew A. Monte
- Department of Emergency MedicineUniversity of Colorado School of MedicineAuroraColoradoUSA
| | - Mohamed Nagy
- Department of Pharmaceutical ServicesChildren's Cancer Hospital Egypt 57357CairoEgypt
- Personalized Medication Management UnitChildren's Cancer Hospital Egypt 57357CairoEgypt
| | - Matthias Schwab
- Dr. Margarete Fischer‐Bosch‐Institute of Clinical PharmacologyStuttgartGermany
- Department of Clinical PharmacologyUniversity Hospital TuebingenTuebingenGermany
- Department of Biochemistry and PharmacyUniversity TuebingenTuebingenGermany
| | - C. Michael Stein
- Division of Clinical Pharmacology, Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of PharmacologyVanderbilt University School of MedicineNashvilleTennesseeUSA
| | - Chakradhara Rao S. Uppugunduri
- Division of Pediatric Oncology and Hematology, Department of Women, Child and AdolescentUniversity Geneva HospitalsGenevaSwitzerland
- Department of Pediatrics, Gynecology and Obstetrics, Cansearch Research Platform for Pediatric Oncology and Hematology, Faculty of MedicineUniversity of GenevaGenevaSwitzerland
| | - Ron H. N. van Schaik
- Department of Clinical ChemistryErasmus MC University Medical CenterRotterdamThe Netherlands
| | - Roseann S. Donnelly
- Department of Pharmacy PracticeMassachusetts College of Pharmacy and Health SciencesBostonMassachusettsUSA
- Department of Pharmacy and Pharmaceutical SciencesSt. Jude Children's Research HospitalMemphisTennesseeUSA
| | - Kelly E. Caudle
- Department of Pharmacy and Pharmaceutical SciencesSt. Jude Children's Research HospitalMemphisTennesseeUSA
| | - Jasmine A. Luzum
- Center for Individualized and Genomic Medicine Research (CIGMA), Henry Ford HospitalDetroitMichiganUSA
- Department of Clinical PharmacyUniversity of Michigan College of PharmacyAnn ArborMichiganUSA
| |
Collapse
|
3
|
Walton M, Wagner JB. Pediatric Beta Blocker Therapy: A Comprehensive Review of Development and Genetic Variation to Guide Precision-Based Therapy in Children, Adolescents, and Young Adults. Genes (Basel) 2024; 15:379. [PMID: 38540438 PMCID: PMC10969836 DOI: 10.3390/genes15030379] [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: 02/21/2024] [Revised: 03/08/2024] [Accepted: 03/15/2024] [Indexed: 06/14/2024] Open
Abstract
Beta adrenergic receptor antagonists, known as beta blockers, are one of the most prescribed medications in both pediatric and adult cardiology. Unfortunately, most of these agents utilized in the pediatric clinical setting are prescribed off-label. Despite regulatory efforts aimed at increasing pediatric drug labeling, a majority of pediatric cardiovascular drug agents continue to lack pediatric-specific data to inform precision dosing for children, adolescents, and young adults. Adding to this complexity is the contribution of development (ontogeny) and genetic variation towards the variability in drug disposition and response. In the absence of current prospective trials, the purpose of this comprehensive review is to illustrate the current knowledge gaps regarding the key drivers of variability in beta blocker drug disposition and response and the opportunities for investigations that will lead to changes in pediatric drug labeling.
Collapse
Affiliation(s)
- Mollie Walton
- Ward Family Heart Center, Kansas City, MO 64108, USA
| | - Jonathan B. Wagner
- Ward Family Heart Center, Kansas City, MO 64108, USA
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children’s Mercy, 2401 Gillham Road, Kansas City, MO 64108, USA
- Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO 64108, USA
| |
Collapse
|
4
|
Fayed MS, Saleh MA, Sabri NA, Elkholy AA. β1-adrenergic receptor polymorphisms: a possible genetic predictor of bisoprolol response in acute coronary syndrome. Future Sci OA 2023; 9:FSO895. [PMID: 37753361 PMCID: PMC10518825 DOI: 10.2144/fsoa-2023-0113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 08/04/2023] [Indexed: 09/28/2023] Open
Abstract
Aim To investigate the association between beta1-adrenergic receptor (ADRB1) polymorphisms and response to bisoprolol treatment in beta-blocker naive patients with acute coronary syndrome (ACS). Patients & methods Seventy-seven patients received bisoprolol for four weeks. Blood pressure and heart rate were measured at baseline and during treatment. TaqMan allelic discrimination method was utilized for ADRB1 Ser49Gly and Arg389Gly genotyping. Results Arg389Arg carriers showed greater reductions in systolic and diastolic blood pressure (-8.5% ± 7.8% vs -0.76% ± 8.7%, p = 0.000218), and (-9.5% ± 9.7% vs -0.80% ± 11.5%, p = 0.000149), respectively, compared with Gly389 carriers. No statistical difference was found for study's outcomes based on codon 49. Conclusion Arg389Gly polymorphism is a promising bisoprolol response predictor in ACS patients.
Collapse
Affiliation(s)
- Mohamed S Fayed
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Mohamed Ayman Saleh
- Department of Cardiology, Faculty of Medicine, Ain Shams University, Cairo, 1181, Egypt
| | - Nagwa A Sabri
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Amal A Elkholy
- Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| |
Collapse
|
5
|
Moric-Janiszewska E, Smolik S, Szydłowski L, Kapral M. Associations between Selected ADRB1 and CYP2D6 Gene Polymorphisms in Children with Ventricular and Supraventricular Arrhythmias. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:2057. [PMID: 38138160 PMCID: PMC10744405 DOI: 10.3390/medicina59122057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/13/2023] [Accepted: 11/17/2023] [Indexed: 12/24/2023]
Abstract
Background and Objectives: Tachycardia is a common cardiovascular disease. Drugs blocking β1-adrenergic receptors (ADRB1) are used in the therapy of arrhythmogenic heart diseases. Disease-related polymorphisms can be observed within the ADRB1 gene. The two most important are Ser49Gly and Arg389Gly, and they influence the treatment efficacy. The family of the cytochrome P450 system consists of the isoenzyme CYP2D6 (Debrisoquine 4-hydroxylase), which is involved in phase I metabolism of almost 25% of clinically important drugs, including antiarrhythmic drugs. A study was conducted to detect the ADRB1 and CYP2D6 gene polymorphisms. Materials and Methods: The material for the test was whole blood from 30 patients with ventricular and supraventricular tachycardia and 20 controls. The samples were obtained from the Department of Pediatric Cardiology. The first to be made was the extraction of DNA using a GeneMATRIX Quick Blood DNA Purification Kit from EURx. The selected ADRB1 and CYP2D6 gene polymorphisms were detected by high-resolution melting polymerase chain reaction (HRM-PCR) analysis. Results: Based on the analysis of melt profile data for each PCR product, the identification of polymorphisms was carried out. Heterozygotes and homozygotes were found in the examined alleles. Conclusions: The frequency of the Arg389Gly polymorphism differs statistically significantly between the control group and patients with supraventricular and ventricular arrhythmias, as well as between these two groups of patients. Moreover, the Arg389Gly polymorphism was statistically more prevalent in the group of girls with SVT arrhythmia compared to girls with VT. A few carriers of homozygous and heterozygous systems of the S49G polymorphism were detected among patients with arrhythmias, as well as control group. The percentage of individuals carrying the CYP2D6 4 allele as either homozygous or heterozygous was observed in the study and control groups. The high prevalence of the CYP2D6*4 allele carriers in both groups prompts the optimization of beta-1 blocker therapy.
Collapse
Affiliation(s)
- Ewa Moric-Janiszewska
- Department of Biochemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Jedności 8B, 41-200 Sosnowiec, Poland
| | - Sławomir Smolik
- Department of Biochemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Jedności 8B, 41-200 Sosnowiec, Poland
| | - Lesław Szydłowski
- Department of Pediatric Cardiology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Medyków 16, 40-752 Katowice, Poland
| | - Małgorzata Kapral
- Department of Biochemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Jedności 8B, 41-200 Sosnowiec, Poland
| |
Collapse
|
6
|
Mboweni N, Maseko M, Tsabedze N. Heart failure with reduced ejection fraction and atrial fibrillation: a Sub-Saharan African perspective. ESC Heart Fail 2023; 10:1580-1596. [PMID: 36934444 PMCID: PMC10192282 DOI: 10.1002/ehf2.14332] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 01/06/2023] [Accepted: 02/07/2023] [Indexed: 03/20/2023] Open
Abstract
Cardiovascular diseases are a well-established cause of death in high-income countries. In the last 20 years, Sub-Saharan Africa (SSA) has seen one of the sharpest increases in cardiovascular disease-related mortality, superseding that of infectious diseases, including HIV/AIDS, in South Africa. This increase is evidenced by a growing burden of heart failure and atrial fibrillation (AF) risk factors. AF is a common comorbidity of heart failure with reduced ejection fraction (HFrEF), which predisposes to an increased risk of stroke, rehospitalizations, and mortality compared with patients in sinus rhythm. AF had the largest relative increase in cardiovascular disease burden between 1990 and 2010 in SSA and the second highest (106.4%) increase in disability-adjusted life-years (DALY) between 1990 and 2017. Over the last decade, significant advancements in the management of both HFrEF and AF have emerged. However, managing HFrEF/AF remains a clinical challenge for physicians, compounded by the suboptimal efficacy of guideline-mandated pharmacotherapy in this group of patients. There may be an essential role for racial differences and genetic influence on therapeutic outcomes of HFrEF/AF patients, further complicating our overall understanding of the disease and its pathophysiology. In SSA, the lack of accurate and up-to-date epidemiological data on this subgroup of patients presents a challenge in our quest to prevent and reduce adverse outcomes. This narrative review provides a contemporary overview of the epidemiology of HFrEF/AF in SSA. We highlight important differences in the demographic and aetiological profile and the management of this subpopulation, emphasizing what is currently known and, more importantly, what is still unknown about HFrEF/AF in SSA.
Collapse
Affiliation(s)
- Nonkanyiso Mboweni
- School of PhysiologyUniversity of the WitwatersrandJohannesburgSouth Africa
- Department of Internal Medicine, Division of Cardiology, Charlotte Maxeke Johannesburg Academic HospitalUniversity of the WitwatersrandGautengJohannesburgSouth Africa
| | - Muzi Maseko
- School of PhysiologyUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Nqoba Tsabedze
- Department of Internal Medicine, Division of Cardiology, Charlotte Maxeke Johannesburg Academic HospitalUniversity of the WitwatersrandGautengJohannesburgSouth Africa
| |
Collapse
|
7
|
Koniari I, Artopoulou E, Velissaris D, Mplani V, Anastasopoulou M, Kounis N, de Gregorio C, Tsigkas G, Karunakaran A, Plotas P, Ikonomidis I. Pharmacologic Rate versus Rhythm Control for Atrial Fibrillation in Heart Failure Patients. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:743. [PMID: 35744006 PMCID: PMC9228123 DOI: 10.3390/medicina58060743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 11/24/2022]
Abstract
Atrial fibrillation (AF) and Heart failure (HF) constitute two frequently coexisting cardiovascular diseases, with a great volume of the scientific research referring to strategies and guidelines associated with the best management of patients suffering from either of the two or both of these entities. The common pathophysiological paths, the adverse outcomes, the hospitalization rates, and the mortality rates that occur from various reports and trials indicate that a targeted therapy to the common background of these cardiovascular conditions may reverse the progression of their interrelating development. Among other optimal treatments concerning the prevalence of both AF and HF, the introduction of rhythm and rate control strategies in the guidelines has underlined the importance of sinus rhythm and heart rate control in the prevention of deleterious complications. The use of these strategies in the clinical practice has led to a debate about the superiority of rhythm versus rate control. The current guidelines as well as the published randomized trials and studies have not proved that rhythm control is more beneficial than the rate control treatments in the terms of survival, all-cause mortality, hospitalization rates, and quality of life. Therefore, the current therapeutic strategy is based on the therapy guidelines and the clinical judgment and experience. The aim of this review was to elucidate the endpoints of pharmacologic randomized clinical trials and the clinical data of each antiarrhythmic or rate-limiting medication, so as to promote their effective, individualized, evidence-based clinical use.
Collapse
Affiliation(s)
- Ioanna Koniari
- Department of Cardiology, University Hospital of South Manchester NHS Foundation Trust, Manchester M23 9LT, UK; (I.K.); (A.K.)
| | - Eleni Artopoulou
- Department of Internal Medicine, University Hospital of Patras, 26504 Patras, Greece; (E.A.); (D.V.)
| | - Dimitrios Velissaris
- Department of Internal Medicine, University Hospital of Patras, 26504 Patras, Greece; (E.A.); (D.V.)
| | - Virginia Mplani
- Department of Cardiology, University Hospital of Patras, 26504 Patras, Greece; (V.M.); (M.A.); (N.K.); (G.T.)
| | - Maria Anastasopoulou
- Department of Cardiology, University Hospital of Patras, 26504 Patras, Greece; (V.M.); (M.A.); (N.K.); (G.T.)
| | - Nicholas Kounis
- Department of Cardiology, University Hospital of Patras, 26504 Patras, Greece; (V.M.); (M.A.); (N.K.); (G.T.)
| | - Cesare de Gregorio
- Department of Clinical and Experimental Medicine Cardiology Unit, University Hospital of Messina, 98125 Messina, Italy;
| | - Grigorios Tsigkas
- Department of Cardiology, University Hospital of Patras, 26504 Patras, Greece; (V.M.); (M.A.); (N.K.); (G.T.)
| | - Arun Karunakaran
- Department of Cardiology, University Hospital of South Manchester NHS Foundation Trust, Manchester M23 9LT, UK; (I.K.); (A.K.)
| | - Panagiotis Plotas
- Laboratory Primary Health Care, School of Health Rehabilitation Sciences, University of Patras, 26504 Patras, Greece;
| | - Ignatios Ikonomidis
- Second Cardiology Department, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| |
Collapse
|
8
|
Zeng W, Chu TTW, Ho CS, Lo CWS, Chan ASL, Kong APS, Tomlinson B, Chan SW. Lack of Effects of Renin-Angiotensin-Aldosterone System Activity and Beta-Adrenoceptor Pathway Polymorphisms on the Response to Bisoprolol in Hypertension. Front Cardiovasc Med 2022; 9:842875. [PMID: 35433877 PMCID: PMC9010557 DOI: 10.3389/fcvm.2022.842875] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 02/10/2022] [Indexed: 01/02/2023] Open
Abstract
PURPOSE This study examined the effects of plasma renin activity (PRA), angiotensin II (Ang II) and aldosterone (PAC) concentrations as well as common polymorphisms in the β1-Adrenoceptor gene (ADRB1) and the G-protein α-Subunit (Gαs) protein gene the G protein α-Subunit 1 gene (GNAS) on the blood pressure (BP) and heart rate (HR) response to bisoprolol in Chinese patients with hypertension. METHODS Patients with sitting clinic systolic BP (SBP) 140-169 mmHg and/or diastolic BP (DBP) 90-109 mmHg after placebo run-in were treated with open-label bisoprolol 2.5 mg daily for 6 weeks. Patients diagnosed as having primary aldosteronism or renal artery stenosis were excluded. PRA, Ang II and PAC concentrations were measured after the placebo run-in and after 6 weeks of treatment. The Ser49Gly and Arg389Gly polymorphisms in ADRB1 and the c.393C > T polymorphism in GNAS were genotyped by the TaqMan® assay. RESULTS In 99 patients who completed the study, baseline PAC levels were significantly associated with baseline DBP and plasma potassium on univariate but not on multivariate linear regression analysis. PRA, Ang II, and PAC concentrations at baseline were not associated with changes in BP with bisoprolol treatment, but the values were all significantly reduced (PRA -0.141 ± 0.595 ng/mL/h, Ang II -2.390 ± 5.171 pmol/L and aldosterone -51.86 ± 119.1 pg/mL; all P < 0.05) following 6 weeks of bisoprolol treatment. There were no significant differences in BP or HR responses in patients with baseline PRA above or below the PRA cut-point of 0.65 ng/mL/h or the median value of 0.9 ng/ml/hour. There were no significant associations of the ADRB1 and GNAS polymorphisms with the clinic and ambulatory BP and HR responses to bisoprolol. CONCLUSION Baseline PRA, PAC and Ang II concentrations showed no significant association with the BP response to bisoprolol treatment, but all these parameters were reduced after 6 weeks of treatment with bisoprolol. The two common polymorphisms in ADRB1 and the c.393C > T polymorphism in GNAS had no significant association with the BP and HR response to bisoprolol in these patients.
Collapse
Affiliation(s)
- Weiwei Zeng
- Shenzhen Baoan Women’s and Children’s Hospital, Jinan University, Shenzhen, China
| | - Tanya T. W. Chu
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Chung Shun Ho
- Department of Chemical Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Clara W. S. Lo
- Department of Chemical Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Alan S. L. Chan
- Department of Chemical Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Alice P. S. Kong
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Brian Tomlinson
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Faculty of Medicine, Macau University of Science and Technology, Macau, Macau SAR, China
| | - Sze Wa Chan
- School of Health Sciences, Caritas Institute of Higher Education, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
9
|
Castaño-Amores C, Díaz-Villamarín X, Pérez-Gutiérrez AM, Antúnez-Rodríguez A, Pozo-Agundo A, Moreno-Escobar E, Sánchez-Ramos JG, Martínez-González LJ, Dávila-Fajardo CL. Pharmacogenetic polymorphisms affecting bisoprolol response. Biomed Pharmacother 2021; 142:112069. [PMID: 34470728 DOI: 10.1016/j.biopha.2021.112069] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/13/2021] [Accepted: 08/17/2021] [Indexed: 12/30/2022] Open
Abstract
β-blockers are commonly prescribed to treat multiple cardiovascular (CV) diseases, but, frequently, adverse drug reactions and intolerance limit their use in clinical practice. Interindividual variability in response to β-blockers may be explained by genetic differences. In fact, pharmacogenetic interactions for some of these drugs have been widely studied, such as metoprolol. But studies that explore genetic variants affecting bisoprolol response are inconclusive, limited or confusing because of mixed results with other β-Blockers, different genetic polymorphisms observed, endpoint studied etc. Because of this, we performed a systematic review in order to find relevant genetic variants affecting bisoprolol response. We have found genetic polymorphism in several genes, but most of the studies focused in ADRB variants. The ADRB1 Arg389Gly (rs1801253) was the most studied genetic polymorphism and it seems to influence the response to bisoprolol, although studies are inconclusive. Even, we performed a meta-analysis about its influence on systolic/diastolic blood pressure in patients treated with bisoprolol, but this did not show statistically significant results. In conclusion, many genetic polymorphisms have been assessed about their influence on patients´ response to bisoprolol and the ADRB1 Arg389Gly (rs1801253) seems the most relevant genetic polymorphism in this regard but results have not been confirmed with a meta-analysis. Our results support the need of further studies about the impact of genetic variants on bisoprolol response, considering different genetic polymorphisms and conducting single and multiple SNPs analysis, including other clinical parameters related to bisoprolol response in a multivariate study.
Collapse
Affiliation(s)
- Celia Castaño-Amores
- Pharmacy Unit; Hospital Universitario clínico San Cecilio - Instituto de investigación biosanitaria (ibs.Granada), Granada, Spain
| | - Xando Díaz-Villamarín
- Pharmacy Unit; Hospital Universitario clínico San Cecilio - Instituto de investigación biosanitaria (ibs.Granada), Granada, Spain; Genomics Unit; Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENyO), Granada, Spain.
| | - Ana María Pérez-Gutiérrez
- Department of Biochemistry and Molecular Biology II; School of Pharmacy, University of Granada, Granada, Spain
| | - Alba Antúnez-Rodríguez
- Genomics Unit; Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENyO), Granada, Spain
| | - Ana Pozo-Agundo
- Genomics Unit; Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENyO), Granada, Spain
| | - Eduardo Moreno-Escobar
- Cardiology Unit; Hospital Universitario clínico San Cecilio - Instituto de investigación biosanitaria (ibs.Granada), Granada, Spain
| | - Jesús Gabriel Sánchez-Ramos
- Cardiology Unit; Hospital Universitario clínico San Cecilio - Instituto de investigación biosanitaria (ibs.Granada), Granada, Spain
| | - Luis Javier Martínez-González
- Genomics Unit; Pfizer-University of Granada-Junta de Andalucía Centre for Genomics and Oncological Research (GENyO), Granada, Spain
| | - Cristina Lucía Dávila-Fajardo
- Pharmacy Unit; Hospital Universitario Virgen de las Nieves - Instituto de investigación biosanitaria (ibs.Granada), Granada, Spain
| |
Collapse
|
10
|
AlHabeeb W, Mrabeti S, Abdelsalam AAI. Therapeutic Properties of Highly Selective β-blockers With or Without Additional Vasodilator Properties: Focus on Bisoprolol and Nebivolol in Patients With Cardiovascular Disease. Cardiovasc Drugs Ther 2021; 36:959-971. [PMID: 34106365 PMCID: PMC9519665 DOI: 10.1007/s10557-021-07205-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/20/2021] [Indexed: 02/07/2023]
Abstract
Bisoprolol and nebivolol are highly selective β1-adrenoceptor antagonists, with clinical indications in many countries within the management of heart failure with reduced left ventricular ejection fraction (HFrEF), ischaemic heart disease (IHD), and hypertension. Nebivolol has additional vasodilator actions, related to enhanced release of NO in the vascular wall. In principle, this additional mechanism compared with bisoprolol might lead to more potent vasodilatation, which in turn might influence the effectiveness of nebivolol in the management of HFrEF, IHD and hypertension. In this article, we review the therapeutic properties of bisoprolol and nebivolol, as representatives of “second generation” and “third generation” β-blockers, respectively. Although head-to-head trials are largely lacking, there is no clear indication from published studies of an additional effect of nebivolol on clinical outcomes in patients with HFrEF or the magnitude of reductions of BP in patients with hypertension.
Collapse
Affiliation(s)
- Waleed AlHabeeb
- Cardiac Sciences Department, King Saud University, Riyadh, 12372, Saudi Arabia.
| | - Sanaa Mrabeti
- General Medicine and Endocrinology, Medical Affairs EMEA, Merck Serono Middle East FZ-LLC, Dubai, United Arab Emirates
| | | |
Collapse
|
11
|
Alkhatib N, Sweitzer NK, Lee CS, Erstad B, Slack M, Gharaibeh M, Karnes J, Klimecki W, Ramos K, Abraham I. Ex Ante Economic Evaluation of Arg389 Genetically Targeted Treatment with Bucindolol versus Empirical Treatment with Carvedilol in NYHA III/IV Heart Failure. Am J Cardiovasc Drugs 2021; 21:205-217. [PMID: 32710439 DOI: 10.1007/s40256-020-00425-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
OBJECTIVE The Beta-Blocker Evaluation Survival Trial showed no survival benefit for bucindolol in New York Heart Association (NYHA) class III/IV heart failure (HF) with reduced ejection fraction, but subanalyses suggested survival benefits for non-Black subjects and Arg389 homozygotes. We conducted an ex ante economic evaluation of Arg389 targeted treatment with bucindolol versus carvidolol, complementing a previous ex ante economic evaluation of bucindolol preceded by genetic testing for the Arg389 polymorphism, in which genetic testing prevailed economically over no testing. METHODS A decision tree analysis with an 18-month time horizon was performed to estimate the cost effectiveness/cost utility of trajectories of 100%, 50%, and 0% of patients genetically tested for Arg389 and comparing bucindolol with empirical carvedilol treatment as per prior BEST subanalyses. Incremental cost-effectiveness/cost-utility ratios (ICERs/ICURs) were estimated. RESULTS Race-based analyses for non-White subjects at 100% testing showed a loss of (0.04) life-years and (0.03) quality-adjusted life-years (QALYs) at an incremental cost of $2185, yielding a negative ICER of ($54,625)/life-year and ICUR of ($72,833)/QALY lost; at 50%, the analyses showed a loss of (0.27) life-years and (0.16) QALYs at an incremental cost of $1843, yielding a negative ICER of ($6826)/life-year and ICUR of ($11,519)/QALY lost; at 0%, the analyses showed a loss of (0.33) life-years and (0.30) QALYs at an incremental cost of $1459, yielding a negative ICER of ($4421)/life-year and ICUR of ($4863)/QALY lost. Arg389 homozygote analyses at 100% testing showed incremental gains of 0.02 life-years and 0.02 QALYs at an incremental cost of $378, yielding an ICER of 18,900/life-year and ICUR of $18,900/QALY gained; at 50%, the analyses showed a loss of (0.24) life-years and (0.09) QALYs at an incremental cost of $1039, yielding a negative ICER of ($4329)/life-year and ICUR of ($9336)/QALY lost; at 0%, the analyses showed a loss of (0.33) life-years and (0.30) QALYs at an incremental cost of $1459, yielding a negative ICER of ($4421)/life-year and ICUR of ($4863)/QALY lost. CONCLUSION This independent ex ante economic evaluation suggests that genetically targeted treatment with bucindolol is unlikely to yield clinicoeconomic benefits over empirical treatment with carvedilol in NYHA III/IV HF.
Collapse
|
12
|
Thomas CD, Johnson JA. Pharmacogenetic factors affecting β-blocker metabolism and response. Expert Opin Drug Metab Toxicol 2020; 16:953-964. [PMID: 32726152 PMCID: PMC7606773 DOI: 10.1080/17425255.2020.1803279] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 07/27/2020] [Indexed: 02/06/2023]
Abstract
INTRODUCTION β-blockers are among the most widely prescribed of all drugs, used for treatment of a large number of cardiovascular diseases. Herein we evaluate literature pertaining to pharmacogenetics of β-blocker therapy, provide insight into the robustness of the genetic associations, and determine the appropriateness for translating these genetic associations into clinical practice. AREAS COVERED A literature search was conducted using PubMed to collate evidence on associations between CYP2D6, ADRB1, ADRB2, and GRK5 genetic variation and drug-response outcomes in the presence of β-blocker exposure. Pharmacokinetic, pharmacodynamic, and clinical outcomes studies were included if genotype data and β-blocker exposure were documented. EXPERT OPINION Substantial data suggest that specific ADRB1 and GRK5 genotypes are associated with improved β-blocker efficacy and have potential for use to guide therapy decisions in the clinical setting. While the data do not justify ordering a CYP2D6 pharmacogenetic test, if CYP2D6 genotype is available in the electronic health record, there may be clinical utility for understanding dosing of β-blockers.
Collapse
Affiliation(s)
- Cameron D. Thomas
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| | - Julie A. Johnson
- Department of Pharmacotherapy and Translational Research and Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to provide an update on the recent advances in the genetics and genomics of dilated cardiomyopathy and heart failure. RECENT FINDINGS Over the last decade, the approach to the discovery of the genetic contribution to heart failure has evolved from investigation of rare variants implicated in Mendelian cardiomyopathies through linkage studies and candidate gene studies to the exploration of the contribution of common variants through large-scale genome-wide association and genome-first studies. The combination and integration of multiple of case-control heart failure cohorts, refinement of the heart failure phenotype, and utilization of large biobanks linked to electronic health records have advanced the understanding of the heritability of heart failure.
Collapse
Affiliation(s)
- Nosheen Reza
- Center for Inherited Cardiovascular Disease, Division of Cardiovascular Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 11 South Tower, Room 11-145 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA.
| | - Anjali Tiku Owens
- Center for Inherited Cardiovascular Disease, Division of Cardiovascular Medicine, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 11 South Tower, Room 11-145 3400 Civic Center Boulevard, Philadelphia, PA, 19104, USA
| |
Collapse
|
14
|
Van Driest SL, Sleeper LA, Gelb BD, Morris SA, Dietz HC, Forbus GA, Goldmuntz E, Hoskoppal A, James J, Lee TM, Levine JC, Li JS, Loeys BL, Markham LW, Meester JAN, Mital S, Mosley JD, Olson AK, Renard M, Shaffer CM, Sharkey A, Young L, Lacro RV, Roden DM. Variants in ADRB1 and CYP2C9: Association with Response to Atenolol and Losartan in Marfan Syndrome. J Pediatr 2020; 222:213-220.e5. [PMID: 32586526 PMCID: PMC7323908 DOI: 10.1016/j.jpeds.2020.03.064] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 03/03/2020] [Accepted: 03/31/2020] [Indexed: 12/26/2022]
Abstract
OBJECTIVE To test whether variants in ADRB1 and CYP2C9 genes identify subgroups of individuals with differential response to treatment for Marfan syndrome through analysis of data from a large, randomized trial. STUDY DESIGN In a subset of 250 white, non-Hispanic participants with Marfan syndrome in a prior randomized trial of atenolol vs losartan, the common variants rs1801252 and rs1801253 in ADRB1 and rs1799853 and rs1057910 in CYP2C9 were analyzed. The primary outcome was baseline-adjusted annual rate of change in the maximum aortic root diameter z-score over 3 years, assessed using mixed effects models. RESULTS Among 122 atenolol-assigned participants, the 70 with rs1801253 CC genotype had greater rate of improvement in aortic root z-score compared with 52 participants with CG or GG genotypes (Time × Genotype interaction P = .005, mean annual z-score change ± SE -0.20 ± 0.03 vs -0.09 ± 0.03). Among participants with the CC genotype in both treatment arms, those assigned to atenolol had greater rate of improvement compared with the 71 of the 121 assigned to losartan (interaction P = .002; -0.20 ± 0.02 vs -0.07 ± 0.02; P < .001). There were no differences in atenolol response by rs1801252 genotype or in losartan response by CYP2C9 metabolizer status. CONCLUSIONS In this exploratory study, ADRB1-rs1801253 was associated with atenolol response in children and young adults with Marfan syndrome. If these findings are confirmed in future studies, ADRB1 genotyping has the potential to guide therapy by identifying those who are likely to have greater therapeutic response to atenolol than losartan.
Collapse
Affiliation(s)
- Sara L. Van Driest
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA,Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lynn A. Sleeper
- Department of Cardiology, Boston Children’s Hospital; and Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Bruce D. Gelb
- Mindich Child Health and Development Institute, Departments of Pediatrics and Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shaine A. Morris
- Division of Cardiology, Department of Pediatrics, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX, USA
| | - Harry C. Dietz
- Institute of Genetic Medicine, Johns Hopkins University School of Medicine and Howard Hughes Medical Institute, Baltimore, MD, USA
| | - Geoffrey A. Forbus
- Department of Pediatrics, Division of Pediatric Cardiology, Medical University of South Carolina, Charleston, SC, USA
| | - Elizabeth Goldmuntz
- Division of Cardiology, Children’s Hospital of Philadelphia, Department of Pediatrics University of Pennsylvania Perlman School of Medicine, Philadelphia, PA, USA
| | - Arvind Hoskoppal
- Departments of Pediatrics and Internal Medicine, University of Utah and Intermountain Healthcare, Salt Lake City, UT, USA
| | - Jeanne James
- Department of Pediatrics, Section of Cardiology, Medical College of Wisconsin and Children’s Hospital of Wisconsin, Milwaukee, WI, USA
| | - Teresa M. Lee
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA
| | - Jami C. Levine
- Department of Cardiology, Boston Children’s Hospital; and Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Jennifer S. Li
- Department of Pediatrics, Division of Cardiology, Duke University Medical Center, Durham, NC, USA
| | - Bart L. Loeys
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Larry W. Markham
- Department of Pediatrics, Division of Pediatric Cardiology, Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Josephina A. N. Meester
- Center of Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Seema Mital
- Department of Pediatrics, Division of Cardiology, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Jonathan D. Mosley
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Aaron K. Olson
- Department of Pediatrics, Seattle Children’s Hospital, Seattle, WA, USA
| | - Marjolijn Renard
- Center for Medical Genetics, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Christian M. Shaffer
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Angela Sharkey
- Department of Pediatrics, Washington University, St. Louis, MO, USA
| | - Luciana Young
- Department of Pediatrics, Ann & Robert H. Lurie Children’s Hospital, Chicago, IL, USA
| | - Ronald V. Lacro
- Department of Cardiology, Boston Children’s Hospital; and Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Dan M. Roden
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA,Departments of Pharmacology and Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW We explore the pharmacogenomics of the beta-blocker bucindolol by discussing relevant beta-1 adrenergic receptor (ADRB1) polymorphisms and recent beta-blocker studies. Through this, we will understand how bucindolol may help patients with atrial fibrillation and heart failure with reduced ejection fraction (AF-HFrEF), which carries poor prognosis. RECENT FINDINGS Retrospective study of the Heart Failure: A Controlled Trial Investigating Outcomes of Exercise Training trial revealed the interaction between the optimal beta-blocker dose and the ADRB1 Arg389 genotype for HFrEF clinical outcomes. Further, a combinatorial genotype analysis in the Beta-Blocker Evaluation of Survival Trial showed that the Arg389Arg genotype, but not the Gly carrier, was associated with 40% lower mortality risk with bucindolol. Finally, the AF-HFrEF subgroup with the ADRB1 Arg389Arg genotype had greater heart rate reduction and suggestion for mortality benefit. Therapeutic response to beta-blockers varies by beta-blocker mechanism, ADRB1 Arg389 genotype, and clinical setting (AF, HFrEF, AF-HFrEF). The ongoing trial A Genotype-Directed Comparative Effectiveness Trial of Bucindolol and Toprol-XL for Prevention of Symptomatic Atrial Fibrillation/Atrial Flutter in Patients with Heart Failure prospectively identifies AF-HFrEF patients with favorable genotype for bucindolol to prevent AF recurrence.
Collapse
Affiliation(s)
- Kishan S Parikh
- Duke Clinical Research Institute, Durham, NC, USA. .,Duke University Medical Center, DUMC 3428, Durham, NC, 27710, USA.
| | | |
Collapse
|
16
|
Salehifar E, Ebrahim S, Shiran MR, Faramarzi F, Askari Rad H, Avan R, Mohseni Kiasari A, Ebrahimi P. Pharmacokinetic Parameters and Over-Responsiveness of Iranian Population to Propranolol. Adv Pharm Bull 2017; 7:195-202. [PMID: 28761821 PMCID: PMC5527233 DOI: 10.15171/apb.2017.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 04/28/2017] [Accepted: 04/30/2017] [Indexed: 11/24/2022] Open
Abstract
Purpose: Propranolol is the most widely used treatment for cardiovascular diseases. Dosage range in our patients is usually less than the amount mentioned in references. The aim of the present study was to clarify whether pharmacokinetic differences are able to justify the need for the fewer doses in our patients or not. Methods: Twenty healthy volunteers (10 male) at heart center of Mazandaran University of Medical Sciences were studied. Samples of blood were collected before a single oral dose (40 mg) of Propranolol. Blood samples were taken up to 9 hours after dose. Total plasma concentration of Propranolol was measured by HPLC. Population Pharmacokinetic analysis was performed using population pharmacokinetics modeling software P-Pharm. Results: The mean value for oral plasma clearance (CL/F) was 126.59 ml/hr. The corresponding values for apparent volume of distribution (V/F), t1/2 beta, maximum blood concentration (C max), and time to reach the maximum blood concentration (T max) were 334.12 Lit, 1.98 hr, 40.25 ng/ml, and 1.68 hr, respectively. The observed mean values of V/F of propranolol in the present study were comparable with those reported in the literature. However, the mean values of CL/F of propranolol in current study was significantly higher than those reported in other population (P-value<0.001). Conclusion: This study has confirmed that the pharmacokinetic differences are not able to justify over-responsiveness of Iranian population to propranolol. Pharmacodynamic differences in responding to beta blocker drugs by Renin secretion or having a different sensibility to beta receptors might play a role in making our population have a different response to propranolol.
Collapse
Affiliation(s)
- Ebrahim Salehifar
- Pharmaceutical Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shima Ebrahim
- Student Research Committee, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad-Reza Shiran
- Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Fatemeh Faramarzi
- Student Research Committee, Department of Clinical Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Hossein Askari Rad
- Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Razieh Avan
- Student Research Committee, Department of Clinical Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Pouneh Ebrahimi
- Department of Chemistry, Faculty of Basic Sciences, Golestan University, Gorgan, Iran
| |
Collapse
|
17
|
Li L, Zhang L, Binkley PF, Sadee W, Wang D. Regulatory Variants Modulate Protein Kinase C α (PRKCA) Gene Expression in Human Heart. Pharm Res 2017; 34:1648-1657. [PMID: 28120175 PMCID: PMC7315374 DOI: 10.1007/s11095-017-2102-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 01/06/2017] [Indexed: 11/26/2022]
Abstract
PURPOSE Protein kinase C α (PRKCA) is involved in multiple functions and has been implicated in heart failure risks and treatment outcomes. This study aims to identify regulatory variants affecting PRKCA expression in human heart, and evaluate attributable risk of heart disease. METHODS mRNA expression quantitative trait loci (eQTLs) were extracted from the Genotype and Tissue Expression Project (GTEx). Allelic mRNA ratios were measured in 51 human heart tissues to identify cis-acting regulatory variants. Potential regulatory regions were tested with luciferase reporter gene assays and further evaluated in GTEx and genome-wide association studies. RESULTS Located in a region with robust enhancer activity in luciferase reporter assays, rs9909004 (T > C, minor allele frequency =0.47) resides in a haplotype displaying strong eQTLs for PRKCA in heart (p = 1.2 × 10-23). The minor C allele is associated with both decreased PRKCA mRNA expression and decreased risk of phenotypes characteristic of heart failure in GWAS analyses (QT interval p = 3.0 × 10-14). While rs9909004 is the likely regulatory variant, other variants in high linkage disequilibrium cannot be excluded. Distinct regulatory variants appear to affect expression in other tissues. CONCLUSIONS The haplotype carrying rs9909004 influences PRKCA expression in the heart and is associated with traits linked to heart failure, potentially affecting therapy of heart failure.
Collapse
Affiliation(s)
- Liang Li
- Center for Pharmacogenomics and Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, 1005 BRT, 460 West 12th Ave, Columbus, Ohio, 43210, USA
| | - Lizhi Zhang
- Center for Pharmacogenomics and Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, 1005 BRT, 460 West 12th Ave, Columbus, Ohio, 43210, USA
| | - Philip F Binkley
- Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, Ohio, 43210, USA
| | - Wolfgang Sadee
- Center for Pharmacogenomics and Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, 1005 BRT, 460 West 12th Ave, Columbus, Ohio, 43210, USA
| | - Danxin Wang
- Center for Pharmacogenomics and Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, 1005 BRT, 460 West 12th Ave, Columbus, Ohio, 43210, USA.
- Center for Pharmacogenomics and Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, 1005 BRT, 460 W 12th Avenue, Columbus, Ohio, 43210, USA.
| |
Collapse
|
18
|
β1-Adrenergic receptor Arg389Gly polymorphism affects the antiarrhythmic efficacy of flecainide in patients with coadministration of β-blockers. Pharmacogenet Genomics 2017; 26:481-5. [PMID: 27500822 DOI: 10.1097/fpc.0000000000000239] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
OBJECTIVE β1-Adrenergic receptor (β1-AR) stimulation modulates the antiarrhythmic activities of sodium channel blockers. The β1-AR Gly389 variant shows a marked decrease in agonist-stimulated cyclic AMP production compared with that of the wild-type Arg389 in vitro. We investigated whether the Arg389Gly polymorphism affects the efficacy of flecainide, a typical sodium channel blocker, in patients with or without coadministration of β-blockers. METHODS The effects of the β1-AR Arg389Gly polymorphism on the antiarrhythmic efficacy of flecainide were compared between with and without coadministered β-blockers in 159 patients with supraventricular tachyarrhythmia. The antiarrhythmic efficacy of flecainide was assessed for at least 2 months by evaluating symptomatology, 12-lead ECGs, and Holter monitoring results. RESULTS Genetic differences in the antiarrhythmic efficacy of flecainide were observed in patients with coadministration of β-blockers. Tachyarrhythmia was well controlled in 60% of Arg389-homozygotes, 30% of Gly389-heterozygotes, and 0% of Gly389-homozygotes (P=0.001). In contrast, no difference in the antiarrhythmic efficacy was observed among the three genotypes in the patients without coadministration of β-blockers (64, 70, and 60%, respectively). Heart rate in tachyarrhythmia in patients treated with flecainide was significantly higher in Gly389 carriers than in Arg389-homozygotes (P=0.013). CONCLUSION The Gly389 polymorphism decreased the antiarrhythmic efficacy of flecainide when coadministered with β-blockers. The results indicate that the Arg389Gly polymorphism may play an important role in predicting the efficacy of flecainide in patients with coadministration of β-blockers.
Collapse
|
19
|
Zaiou M, El Amri H. Cardiovascular pharmacogenetics: a promise for genomically-guided therapy and personalized medicine. Clin Genet 2017; 91:355-370. [PMID: 27714756 DOI: 10.1111/cge.12881] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 09/30/2016] [Accepted: 10/03/2016] [Indexed: 12/28/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide. The basic causes of CVD are not fully understood yet. Substantial evidence suggests that genetic predisposition plays a vital role in the physiopathology of this complex disease. Hence, identification of genetic contributors to CVD will likely add diagnostic accuracy and better prediction of an individual's risk. With high-throughput genetics and genomics technology and newer genome-wide study approaches, a number of genetic variations across the human genome were uncovered. Evidence suggests that genetic defects could influence CVD development and inter-individual responses to widely used cardiovascular drugs like clopidogrel, aspirin, warfarin, and statins, and therefore, they may be integrated into clinical practice. If clinically validated, better understanding of these genetic variations may provide new opportunities for personalized diagnostic, pharmacogenetic-based drug selection and best treatment in personalized medicine. However, numerous gaps remain unsolved due to the lack of underlying pathological mechanisms for how genetic predisposition could contribute to CVD. This review provides an overview of the extraordinary scientific progress in our understanding of genetic and genomic basis of CVD as well as the development of relevant genetic biomarkers for this disease. Some of the actual limitations to the promise of these markers and their translation for the benefit of patients will be discussed.
Collapse
Affiliation(s)
- M Zaiou
- Faculté de Pharmacie, Université de Lorraine, Nancy, France
| | - H El Amri
- Laboratoire de Génétique de la Gendarmerie Royale, Avenue Ibn Sina, Rabat, Maroc
| |
Collapse
|
20
|
Mottet F, Vardeny O, de Denus S. Pharmacogenomics of heart failure: a systematic review. Pharmacogenomics 2016; 17:1817-1858. [PMID: 27813451 DOI: 10.2217/pgs-2016-0118] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Heart failure (HF) and multiple HF-related phenotypes are heritable. Genes implicated in the HF pathophysiology would be expected to influence the response to treatment. METHODS We conducted a series of systematic literature searches on the pharmacogenetics of HF therapy to assess the current knowledge on this field. RESULTS Existing data related to HF pharmacogenomics are still limited. The ADRB1 gene is a likely candidate to predict response to β-blockers. Moreover, the cytochrome P450 2D6 coding gene (CYP2D6) clearly affects the pharmacokinetics of metoprolol, although the clinical impact of this association remains to be established. CONCLUSION Given the rising prevalence of HF and related costs, a more personalized use of HF drugs could have a remarkable benefit for patients, caregivers and healthcare systems.
Collapse
Affiliation(s)
- Fannie Mottet
- Faculty of Pharmacy, Université de Montréal, Montreal, Canada.,Montreal Heart Institute, Montreal, Canada
| | - Orly Vardeny
- Associate Professor of Pharmacy & Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Simon de Denus
- Faculty of Pharmacy, Université de Montréal, Montreal, Canada.,Montreal Heart Institute, Montreal, Canada
| |
Collapse
|
21
|
Van Gelder IC, Rienstra M, Crijns HJGM, Olshansky B. Rate control in atrial fibrillation. Lancet 2016; 388:818-28. [PMID: 27560277 DOI: 10.1016/s0140-6736(16)31258-2] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 07/12/2016] [Accepted: 07/13/2016] [Indexed: 12/14/2022]
Abstract
Control of the heart rate (rate control) is central to atrial fibrillation management, even for patients who ultimately require control of the rhythm. We review heart rate control in patients with atrial fibrillation, including the rationale for the intervention, patient selection, and the treatments available. The choice of rate control depends on the symptoms and clinical characteristics of the patient, but for all patients with atrial fibrillation, rate control is part of the management. Choice of drugs is patient-dependent. β blockers, alone or in combination with digoxin, or non-dihydropyridine calcium-channel blockers (not in heart failure) effectively lower the heart rate. Digoxin is least effective, but a reasonable choice for physically inactive patients aged 80 years or older, in whom other treatments are ineffective or are contraindicated, and as an additional drug to other rate-controlling drugs, especially in heart failure when instituted cautiously. Atrioventricular node ablation with pacemaker insertion for rate control should be used as an approach of last resort but is also an option early in the management of patients with atrial fibrillation treated with cardiac resynchronisation therapy. However, catheter ablation of atrial fibrillation should be considered before atrioventricular node ablation. Although rate control is a top priority and one of the first management issues for all patients with atrial fibrillation, many issues remain.
Collapse
Affiliation(s)
- Isabelle C Van Gelder
- Department of Cardiology, Thoraxcenter, University of Groningen, University Medical Center Groningen, Groningen, Netherlands.
| | - Michiel Rienstra
- Department of Cardiology, Thoraxcenter, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Harry J G M Crijns
- Maastricht University Medical Center and Cardiovascular Research Institute Maastricht (CARIM), Maastricht, Netherlands
| | - Brian Olshansky
- Mercy Heart and Vascular Institute, Mercy Medical Center North Iowa, Mason City, IA, USA
| |
Collapse
|
22
|
Cavallari LH, Mason DL. Cardiovascular Pharmacogenomics--Implications for Patients With CKD. Adv Chronic Kidney Dis 2016; 23:82-90. [PMID: 26979147 DOI: 10.1053/j.ackd.2015.12.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 12/04/2015] [Accepted: 12/08/2015] [Indexed: 01/20/2023]
Abstract
CKD is an independent risk factor for cardiovascular disease (CVD). Thus, patients with CKD often require treatment with cardiovascular drugs, such as antiplatelet, antihypertensive, anticoagulant, and lipid-lowering agents. There is significant interpatient variability in response to cardiovascular therapies, which contributes to risk for treatment failure or adverse drug effects. Pharmacogenomics offers the potential to optimize cardiovascular pharmacotherapy and improve outcomes in patients with CVD, although data in patients with concomitant CKD are limited. The drugs with the most pharmacogenomic evidence are warfarin, clopidogrel, and statins. There are also accumulating data for genetic contributions to β-blocker response. Guidelines are now available to assist with applying pharmacogenetic test results to optimize warfarin dosing, selection of antiplatelet therapy after percutaneous coronary intervention, and prediction of risk for statin-induced myopathy. Clinical data, such as age, body size, and kidney function have long been used to optimize drug prescribing. An increasing number of institutions are also implementing genetic testing to be considered in the context of important clinical factors to further personalize drug therapy for patients with CVD.
Collapse
|
23
|
Eadon MT, Chapman AB. A Physiologic Approach to the Pharmacogenomics of Hypertension. Adv Chronic Kidney Dis 2016; 23:91-105. [PMID: 26979148 DOI: 10.1053/j.ackd.2016.02.003] [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] [Indexed: 12/26/2022]
Abstract
Hypertension is a multifactorial condition with diverse physiological systems contributing to its pathogenesis. Individuals exhibit significant variation in their response to antihypertensive agents. Traditional markers, such as age, gender, diet, plasma renin level, and ethnicity, aid in drug selection. However, this review explores the contribution of genetics to facilitate antihypertensive agent selection and predict treatment efficacy. The findings, reproducibility, and limitations of published studies are examined, with emphasis placed on candidate genetic variants affecting drug metabolism, the renin-angiotensin system, adrenergic signalling, and renal sodium reabsorption. Single-nucleotide polymorphisms identified and replicated in unbiased genome-wide association studies of hypertension treatment are reviewed to illustrate the evolving understanding of the disease's complex and polygenic pathophysiology. Implementation efforts at academic centers seek to overcome barriers to the broad adoption of pharmacogenomics in the treatment of hypertension. The level of evidence required to support the implementation of pharmacogenomics in clinical practice is considered.
Collapse
|
24
|
Iyngkaran P, Thomas MC, Johnson R, French J, Ilton M, McDonald P, Hare DL, Fatkin D. Contextualizing Genetics for Regional Heart Failure Care. Curr Cardiol Rev 2016; 12:231-242. [PMID: 27280306 PMCID: PMC5011192 DOI: 10.2174/1573403x12666160606123103] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 12/18/2015] [Accepted: 01/11/2016] [Indexed: 12/21/2022] Open
Abstract
Congestive heart failure (CHF) is a chronic and often devastating cardiovascular disorder with no cure. There has been much advancement in the last two decades that has seen improvements in morbidity and mortality. Clinicians have also noted variations in the responses to therapies. More detailed observations also point to clusters of diseases, phenotypic groupings, unusual severity and the rates at which CHF occurs. Medical genetics is playing an increasingly important role in answering some of these observations. This developing field in many respects provides more information than is currently clinically applicable. This includes making sense of the established single gene mutations or uncommon private mutations. In this thematic series which discusses the many factors that could be relevant for CHF care, once established treatments are available in the communities; this section addresses a contextual role for medical genetics.
Collapse
|
25
|
Flenner F, Friedrich FW, Ungeheuer N, Christ T, Geertz B, Reischmann S, Wagner S, Stathopoulou K, Söhren KD, Weinberger F, Schwedhelm E, Cuello F, Maier LS, Eschenhagen T, Carrier L. Ranolazine antagonizes catecholamine-induced dysfunction in isolated cardiomyocytes, but lacks long-term therapeutic effects in vivo in a mouse model of hypertrophic cardiomyopathy. Cardiovasc Res 2015; 109:90-102. [PMID: 26531128 DOI: 10.1093/cvr/cvv247] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 10/27/2015] [Indexed: 12/19/2022] Open
Abstract
AIMS Hypertrophic cardiomyopathy (HCM) is often accompanied by increased myofilament Ca(2+) sensitivity and diastolic dysfunction. Recent findings indicate increased late Na(+) current density in human HCM cardiomyocytes. Since ranolazine has the potential to decrease myofilament Ca(2+) sensitivity and late Na(+) current, we investigated its effects in an Mybpc3-targeted knock-in (KI) mouse model of HCM. METHODS AND RESULTS Unloaded sarcomere shortening and Ca(2+) transients were measured in KI and wild-type (WT) cardiomyocytes. Measurements were performed at baseline (1 Hz) and under increased workload (30 nM isoprenaline (ISO), 5 Hz) in the absence or presence of 10 µM ranolazine. KI myocytes showed shorter diastolic sarcomere length at baseline, stronger inotropic response to ISO, and drastic drop of diastolic sarcomere length under increased workload. Ranolazine attenuated ISO responses in WT and KI cells and prevented workload-induced diastolic failure in KI. Late Na(+) current density was diminished and insensitive to ranolazine in KI cardiomyocytes. Ca(2+) sensitivity of skinned KI trabeculae was slightly decreased by ranolazine. Phosphorylation analysis of cAMP-dependent protein kinase A-target proteins and ISO concentration-response measurements on muscle strips indicated antagonism at β-adrenoceptors with 10 µM ranolazine shifting the ISO response by 0.6 log units. Six-month treatment with ranolazine (plasma level >20 µM) demonstrated a β-blocking effect, but did not reverse cardiac hypertrophy or dysfunction in KI mice. CONCLUSION Ranolazine improved tolerance to high workload in mouse HCM cardiomyocytes, not by blocking late Na(+) current, but by antagonizing β-adrenergic stimulation and slightly desensitizing myofilaments to Ca(2+). This effect did not translate in therapeutic efficacy in vivo.
Collapse
Affiliation(s)
- Frederik Flenner
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Felix W Friedrich
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Nele Ungeheuer
- Clinic for Cardiology and Pneumology, Georg-August-University Göttingen, Göttingen, Germany
| | - Torsten Christ
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Birgit Geertz
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Silke Reischmann
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Stefan Wagner
- Clinic for Cardiology and Pneumology, Georg-August-University Göttingen, Göttingen, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany Department for Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Konstantina Stathopoulou
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Klaus-Dieter Söhren
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany
| | - Florian Weinberger
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Edzard Schwedhelm
- DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany Department of Clinical Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friederike Cuello
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Lars S Maier
- Clinic for Cardiology and Pneumology, Georg-August-University Göttingen, Göttingen, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany Department for Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Thomas Eschenhagen
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Lucie Carrier
- Department of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Martinistraße 52, D-20246 Hamburg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| |
Collapse
|
26
|
Association of common polymorphisms in β1-adrenergic receptor with antihypertensive response to carvedilol. J Cardiovasc Pharmacol 2015; 64:306-9. [PMID: 25291495 DOI: 10.1097/fjc.0000000000000119] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
OBJECTIVES Marked interpatient variability exists in the blood pressure response to carvedilol, a nonselective β-blocker. Here we evaluated the influence of 4 common polymorphisms in genes of the β-adrenergic receptor on the antihypertensive efficacy of carvedilol in patients in a double-blinded monotherapy study. METHODS Eighty-seven subjects with uncomplicated essential hypertensive (49% men; age = 52.2 ± 11.1 years) from Jilin province of China were enrolled in the study, and 5 of them discontinued the treatment due to adverse effects. Both systolic and diastolic blood pressures (DBPs) were measured before and after 7 days of treatment with carvedilol (10 mg/d). Genotypes of the β1-adrenergic receptor (ADRB1 Ser49Gly and Arg389Gly) and β2-adrenergic receptor (ADRB2 Gly16Arg and Glu27Gln) were determined by polymerase chain reaction with restriction fragment length polymorphism. RESULTS Patients homozygous for ADRB1 Arg389 had an approximately 4-fold greater reduction in DBPs than those homozygous for ADRB1 Gly389 (10.61 vs. 2.62 mm Hg, P = 0.013). The ADRB1 haplotype was also a significant predictor of response, as patients with the Gly49Arg389/Ser49Arg389 haplotype pair had a 5.7-fold greater reduction in DBPs than those homozygous for the Ser49Gly389 haplotype (16.11 vs. 2.83 mm Hg, P = 0.0055). An association was not found between ADRB2 polymorphism and carvedilol responsiveness in antihypertensive therapy. CONCLUSIONS This study provides the first evidence to support that ADRB1 polymorphisms play an important role in the DBPs response to carvedilol treatment in patients with essential hypertension.
Collapse
|
27
|
Abstract
Pharmacological treatment of atrial fibrillation in the context of heart failure poses numerous challenges. Management decisions are limited by contraindications to several drugs and the paucity of robust clinical trials that provide evidence-based guidance. This review proposes a structured action plan for managing atrial fibrillation coexisting with heart failure that considers published clinical guidelines and integrates recent data derived from substudies of randomized trials, including the atrial fibrillation and congestive heart failure (AF-CHF) trial. Areas of uncertainty, such as target heart rates in atrial fibrillation and upstream therapies, are also discussed.
Collapse
|
28
|
Abstract
Heart failure is becoming increasingly prevalent in the United States and is a significant cause of morbidity and mortality. Several therapies are currently available to treat this chronic illness; however, clinical response to these treatment options exhibit significant interpatient variation. It is now clearly understood that genetics is a key contributor to diversity in therapeutic response, and evidence that genetic polymorphisms alter the pharmacokinetics, pharmacodynamics, and clinical response of heart failure drugs continues to accumulate. This suggests that pharmacogenomics has the potential to help clinicians improve the management of heart failure by choosing the safest and most effective medications and doses. Unfortunately, despite much supportive data, pharmacogenetic optimization of heart failure treatment regimens is not yet a reality. In order to attenuate the rising burden of heart failure, particularly in the context of the recent paucity of new effective interventions, there is an urgent need to extend pharmacogenetic knowledge and leverage these associations in order to enhance the effectiveness of existing heart failure therapies. This review focuses on the current state of pharmacogenomics in heart failure and provides a glimpse of the aforementioned future needs.
Collapse
Affiliation(s)
- Akinyemi Oni-Orisan
- University of North Carolina at Chapel Hill, UNC Eshelman School of Pharmacy, Center for Pharmacogenomics and Individualized Therapy
| | - David Lanfear
- Section Head, Advanced Heart Failure and Cardiac Transplantation, Research Scientist, Center for Health Services Research, Henry Ford Hospital, 2799 W. Grand Boulevard Detroit, MI 48202, Phone: 313-916-6375, Fax: 313-916-8799
| |
Collapse
|
29
|
Ahles A, Engelhardt S. Polymorphic variants of adrenoceptors: pharmacology, physiology, and role in disease. Pharmacol Rev 2014; 66:598-637. [PMID: 24928328 DOI: 10.1124/pr.113.008219] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2025] Open
Abstract
The human genome encodes nine different adrenoceptor genes. These are grouped into three families, namely, the α1-, α2-, and β-adrenoceptors, with three family members each. Adrenoceptors are expressed by most cell types of the human body and are primary targets of the catecholamines epinephrine and norepinephrine that are released from the sympathetic nervous system during its activation. Upon catecholamine binding, adrenoceptors change conformation, couple to and activate G proteins, and thereby initiate various intracellular signaling cascades. As the primary receivers and transducers of sympathetic activation, adrenoceptors have a central role in human physiology and disease and are important targets for widely used drugs. All nine adrenoceptor subtypes display substantial genetic variation, both in their coding sequence as well as in adjacent regions. Despite the fact that some of the adrenoceptor variants range among the most frequently studied genetic variants assessed in pharmacogenetics to date, their functional relevance remains ill defined in many cases. A substantial fraction of the associations reported from early candidate gene approaches have not subsequently been confirmed in different cohorts or in genome-wide association studies, which have increasingly been conducted in recent years. This review aims to provide a comprehensive overview of all adrenoceptor variants that have reproducibly been detected in the larger genome sequencing efforts. We evaluate these variants with respect to the modulation of receptor function and expression and discuss their role in physiology and disease.
Collapse
Affiliation(s)
- Andrea Ahles
- Institut für Pharmakologie und Toxikologie, Technische Universität München, Munich, Germany (A.A., S.E.); and DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (S.E.)
| | - Stefan Engelhardt
- Institut für Pharmakologie und Toxikologie, Technische Universität München, Munich, Germany (A.A., S.E.); and DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany (S.E.)
| |
Collapse
|
30
|
Baker JG, Proudman RGW, Hill SJ. Impact of polymorphic variants on the molecular pharmacology of the two-agonist conformations of the human β1-adrenoceptor. PLoS One 2013; 8:e77582. [PMID: 24250787 PMCID: PMC3826719 DOI: 10.1371/journal.pone.0077582] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 09/04/2013] [Indexed: 11/18/2022] Open
Abstract
β-blockers are widely used to improve symptoms and prolong life in heart disease primarily by inhibiting the actions of endogenous catecholamines at the β1-adrenoceptor. There are two common naturally occurring polymorphisms within the human β1-adrenoceptor sequence: Ser or Gly at position 49 in the N-terminus and Gly or Arg at position 389 in the C-terminus and some clinical studies have suggested that expression of certain variants may be associated with disease and affect response to treatment with β-blockers. The β1-adrenoceptor also exists in two agonist conformations - a high affinity catecholamine conformation and a low affinity secondary agonist conformation. Receptor-effector coupling and intracellular signalling from the different conformations may be affected by the polymorphic variants. Here, we examine in detail the molecular pharmacology of the β1-adrenoceptor polymorphic variants with respect to ligand affinity, efficacy, activation of the different agonist conformations and signal transduction and determine whether the polymorphic variants do indeed affect this secondary conformation. Stable cell lines expressing the wildtype and polymorphic variants were constructed and receptor pharmacology examined using whole cell binding and intracellular secondary messenger techniques. There was no difference in affinity for agonists and antagonists at the human wildtype β1-adrenoceptor (Ser49/Gly389) and the polymorphic variants Gly49/Gly389 and Ser49/Arg389. Furthermore, the polymorphic variant receptors both have two active agonist conformations with pharmacological properties similar to the wildtype receptor. Although the polymorphism at position 389 is thought to occur in an intracellular domain important for Gs-coupling, the two agonist conformations of the polymorphic variants stimulate intracellular signalling pathways, including Gs-cAMP intracellular signalling, in a manner very similar to that of the wildtype receptor.
Collapse
Affiliation(s)
- Jillian G. Baker
- Cell Signalling, School of Life Sciences, University of Nottingham, Nottingham, Nottinghamshire, United Kingdom
| | - Richard G. W. Proudman
- Cell Signalling, School of Life Sciences, University of Nottingham, Nottingham, Nottinghamshire, United Kingdom
| | - Stephen J. Hill
- Cell Signalling, School of Life Sciences, University of Nottingham, Nottingham, Nottinghamshire, United Kingdom
| |
Collapse
|
31
|
Abstract
In the last few years, there has been a major shift in the treatment of atrial fibrillation (AF) in the setting of hear failure (HF), from rhythm to ventricular rate control in most patients with both conditions. In this article, the authors focus on ventricular rate control and discuss the indications; the optimal ventricular rate-control target, including detailed results of the Rate Control Efficacy in Permanent Atrial Fibrillation: a Comparison Between Lenient versus Strict Rate Control II (RACE II) study; and the pharmacologic and nonpharmacologic options to control the ventricular rate during AF in the setting of HF.
Collapse
Affiliation(s)
- Michiel Rienstra
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, Hanzeplein 1, PO Box 30.001, Groningen 9700 RB, The Netherlands
| | | |
Collapse
|
32
|
[Personalized therapy in cardiology. Biomarkers, pharmacogenetics and therapy of monogenic diseases]. Internist (Berl) 2013; 54:147-8, 150-2, 154. [PMID: 23371262 DOI: 10.1007/s00108-012-3157-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Improved therapy and prophylaxis of cardiovascular diseases have contributed to an increase in life expectancy like no other field of medicine. However, many cardiological diseases remain untreatable and standard therapies often work only in a minority of patients or cause more harm than benefit. Personalized approaches appear to be a promising solution. Monogenic heart diseases are paradigmatic for this approach and can in rare cases be treated mutation specifically. Overall, however, success remains limited. Next generation sequencing will facilitate the identification of mutations causing diseases. Cell culture models based on induced pluripotent stem cells open the perspective of individualized testing of disease severity and pharmacological or genetic therapy. In contrast to monogenic diseases genetic testing plays no practical role yet in the management of multifactorial cardiovascular diseases. Biomarkers can identify individuals with increased cardiovascular risk. Furthermore, biomarker-guided therapy represents an attractive option with troponin-guided therapy of acute coronary syndromes as a successful example. Individual responses to drugs vary and are partly determined by genes. Simple genetic analyses can improve response prediction and minimize side effects in cases such as warfarin and high doses of simvastatin. Taken together personalized approaches will gain importance in the cardiovascular field but this requires the development of better methods and research that quantifies the true value of the new knowledge.
Collapse
|
33
|
Eschenhagen T. A frequent gene polymorphism affecting the heart-rate response to carvedilol. Pharmacogenomics 2013; 14:115-8. [PMID: 23327570 DOI: 10.2217/pgs.12.198] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
34
|
Filigheddu F. Genetic prediction of heart failure incidence, prognosis and beta-blocker response. Mol Diagn Ther 2013; 17:205-19. [PMID: 23592012 DOI: 10.1007/s40291-013-0035-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Heart failure (HF) is a widespread syndrome due to left ventricular dysfunction with high mortality, morbidity and health-care costs. Beta-blockers, together with diuretics and ACE-inhibitors or angiotensin receptor blockers, are a cornerstone of HF therapy, as they reduce mortality and morbidity. Nevertheless, their efficacy varies among patients, and genetics is likely to be one of the modifying factors. In this article, literature on the role of candidate genes on the development of HF, its prognosis and pharmacogenomics of β-blockers in patients with HF is reviewed. The available findings do not support, at the present time, a role for genetic tests in the treatment of HF. More large-scale genome-wide studies with adequate methodology and statistical analysis are required before considering genetic tailoring of HF therapy in patients with systolic HF.
Collapse
Affiliation(s)
- Fabiana Filigheddu
- Department of Clinical and Experimental Medicine, University of Sassari, Viale S.Pietro 8, 07100 Sassari, Italy.
| |
Collapse
|
35
|
Kao DP, Davis G, Aleong R, O'Connor CM, Fiuzat M, Carson PE, Anand IS, Plehn JF, Gottlieb SS, Silver MA, Lindenfeld J, Miller AB, White M, Murphy GA, Sauer W, Bristow MR. Effect of bucindolol on heart failure outcomes and heart rate response in patients with reduced ejection fraction heart failure and atrial fibrillation. Eur J Heart Fail 2012; 15:324-33. [PMID: 23223178 DOI: 10.1093/eurjhf/hfs181] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AIMS There is little evidence of beta-blocker treatment benefit in patients with heart failure and reduced left ventricular ejection fraction (HFREF) and atrial fibrillation (AF). We investigated the effects of bucindolol in HFREF patients with AF enrolled in the Beta-blocker Evaluation of Survival Trial (BEST). METHODS AND RESULTS A post-hoc analysis of patients in BEST with and without AF was performed to estimate the effect of bucindolol on mortality and hospitalization. Patients were also evaluated for treatment effects on heart rate and the influence of beta1-adrenergic receptor position 389 (β(1)389) arginine (Arg) vs. glycine (Gly) genotypes. In the 303/2708 patients in AF, patients receiving bucindolol were more likely to achieve a resting heart rate ≤ 80 b.p.m. at 3 months (P < 0.005) in the absence of treatment-limiting bradycardia. In AF patients and sinus rhythm (SR) patients who achieved a resting heart rate ≤ 80 b.p.m., there were beneficial treatment effects on cardiovascular mortality/cardiovascular hospitalization [hazard ratio (HR) 0.61, P = 0.025, and 0.79, P = 0.002]. Without achieving a resting heart rate ≤ 80 b.p.m., there were no treatment effects on events in either group. β(1)389-Arg/Arg AF patients had nominally significant reductions in all-cause mortality/HF hospitalization and cardiovascular mortality/hospitalization with bucindolol (HR 0.23, P = 0.037 and 0.28, P = 0.039), whereas Gly carriers did not. There was no evidence of diminished heart rate response in β(1)389-Arg homozygotes. CONCLUSION In HFREF patients with AF, bucindolol was associated with reductions in composite HF endpoints in those who achieved a resting heart rate ≤ 80 b.p.m. and nominally in those with the β(1)389-Arg homozygous genotype.
Collapse
Affiliation(s)
- David P Kao
- University of Colorado School of Medicine, Aurora, CO 80045, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|