1
|
Myers RA, Ortel TL, Waldrop A, Cornwell M, Newman JD, Levy NK, Barrett TJ, Ruggles K, Sowa MA, Dave S, Ginsburg GS, Berger JS, Voora D. Platelet RNA Biomarker of Ticagrelor-Responsive Genes Is Associated With Platelet Function and Cardiovascular Events. Arterioscler Thromb Vasc Biol 2024; 44:423-434. [PMID: 38059352 PMCID: PMC10843550 DOI: 10.1161/atvbaha.123.319759] [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: 06/19/2023] [Accepted: 11/10/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Identifying patients with the optimal risk:benefit for ticagrelor is challenging. The aim was to identify ticagrelor-responsive platelet transcripts as biomarkers of platelet function and cardiovascular risk. METHODS Healthy volunteers (n=58, discovery; n=49, validation) were exposed to 4 weeks of ticagrelor with platelet RNA data, platelet function, and self-reported bleeding measured pre-/post-ticagrelor. RNA sequencing was used to discover platelet genes affected by ticagrelor, and a subset of the most informative was summarized into a composite score and tested for validation. This score was further analyzed (1) in CD34+ megakaryocytes exposed to an P2Y12 inhibitor in vitro, (2) with baseline platelet function in healthy controls, (3) in peripheral artery disease patients (n=139) versus patient controls (n=30) without atherosclerosis, and (4) in patients with peripheral artery disease for correlation with atherosclerosis severity and risk of incident major adverse cardiovascular and limb events. RESULTS Ticagrelor exposure differentially expressed 3409 platelet transcripts. Of these, 111 were prioritized to calculate a Ticagrelor Exposure Signature score, which ticagrelor reproducibly increased in discovery and validation cohorts. Ticagrelor's effects on platelets transcripts positively correlated with effects of P2Y12 inhibition in primary megakaryocytes. In healthy controls, higher baseline scores correlated with lower baseline platelet function and with minor bleeding while receiving ticagrelor. In patients, lower scores independently associated with both the presence and extent of atherosclerosis and incident ischemic events. CONCLUSIONS Ticagrelor-responsive platelet transcripts are a biomarker for platelet function and cardiovascular risk and may have clinical utility for selecting patients with optimal risk:benefit for ticagrelor use.
Collapse
Affiliation(s)
- Rachel A Myers
- Duke Clinical Research Unit, Duke University School of Medicine, Durham NC
| | - Thomas L Ortel
- Departments of Medicine, Duke University Medical Center, Durham NC
| | - Alexander Waldrop
- Departments of Medicine, Duke University Medical Center, Durham NC
- Center for Genomics and Computational Biology, Duke University, Durham, NC
| | - MacIntosh Cornwell
- NYU Grossman School of Medicine, Leon H. Charney Division of Cardiology, New York, NY
| | - Jonathan D. Newman
- NYU Grossman School of Medicine, Leon H. Charney Division of Cardiology, New York, NY
| | - Natalie K Levy
- NYU Grossman School of Medicine, Leon H. Charney Division of Cardiology, New York, NY
| | - Tessa J. Barrett
- NYU Grossman School of Medicine, Leon H. Charney Division of Cardiology, New York, NY
| | - Kelly Ruggles
- NYU Grossman School of Medicine, Leon H. Charney Division of Cardiology, New York, NY
| | - Marcin A Sowa
- NYU Grossman School of Medicine, Leon H. Charney Division of Cardiology, New York, NY
| | - Sandeep Dave
- Departments of Medicine, Duke University Medical Center, Durham NC
- Center for Genomics and Computational Biology, Duke University, Durham, NC
| | | | - Jeffrey S. Berger
- NYU Grossman School of Medicine, Leon H. Charney Division of Cardiology, New York, NY
| | - Deepak Voora
- Departments of Medicine, Duke University Medical Center, Durham NC
| |
Collapse
|
2
|
Friede KA, Myers RA, Gales J, Zhbannikov I, Ortel TL, Shah SH, Kraus WE, Ginsburg GS, Voora D. OUP accepted manuscript. Cardiovasc Res 2022; 119:551-560. [PMID: 35576481 DOI: 10.1093/cvr/cvac079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 04/10/2022] [Accepted: 04/25/2022] [Indexed: 11/14/2022] Open
Abstract
AIMS Gene expression biosignatures may hold promise to individualize antiplatelet therapy in conjunction with current guidelines and risk scores. The Aspirin Response Signature (ARS) score is comprised of a weighted sum of correlated, pro-thrombotic gene transcripts measured in whole blood. In prior work where volunteers were exposed to aspirin 325 mg daily, higher ARS score was associated with lower platelet function; separately, in a clinical cohort of patients, higher ARS scores were associated with increased risk of adverse cardiovascular events. To better understand this apparent paradox, we measured ARS gene expression and score in volunteers to determine aspirin dose-response and ticagrelor relationships with ARS score and separately in patients to assess whether ARS is associated with incident bleeding. METHODS AND RESULTS Blood samples were collected from volunteers (N = 188) who were exposed to 4 weeks of daily aspirin 81 mg, daily aspirin 325 mg, and/or twice-daily ticagrelor 90 mg. ARS scores were calculated from whole blood RNA qPCR, and platelet function and protein expression were assessed in platelet-rich plasma. In mixed linear regression models, aspirin 81 mg exposure was not associated with changes in ARS gene expression or score. Aspirin 325 mg exposure resulted in a 6.0% increase in ARS gene expression (P = 7.5 × 10-9 vs. baseline, P = 2.1 × 10-4 vs. aspirin 81 mg) and an increase in expression of platelet proteins corresponding to ARS genes. Ticagrelor exposure resulted in a 30.7% increase in ARS gene expression (P < 1 × 10-10 vs. baseline and each aspirin dose) and ARS score (P = 7.0 × 10-7 vs. baseline, P = 3.6 × 10-6 and 5.59 × 10-4 vs. aspirin 81 and 325 mg, respectively). Increases in ARS gene expression or score were associated with the magnitude of platelet inhibition across agents. To assess the association between ARS scores and incident bleeding, ARS scores were calculated in patients undergoing cardiac catheterization (N = 1421), of whom 25.4% experienced bleeding events over a median 6.2 years of follow-up. In a Cox model adjusting for demographics and baseline antithrombotic medication use, patients with ARS scores above the median had a higher risk of incident bleeding [hazard ratio 1.26 (95% CI 1.01-1.56), P = 0.038]. CONCLUSIONS The ARS is an Antiplatelet Response Signature that increases in response to greater platelet inhibition due to antiplatelet therapy and may represent a homeostatic mechanism to prevent bleeding. ARS scores could inform future strategies to prevent bleeding while maintaining antiplatelet therapy's benefit of ischaemic cardiovascular event protection.
Collapse
Affiliation(s)
- Kevin A Friede
- Center for Applied Genomics & Precision Medicine, Duke University, 101 Science Dr, DUMC 3382, Durham, NC, USA
- Division of Cardiology, Duke University, Durham, NC, USA
| | - Rachel A Myers
- Center for Applied Genomics & Precision Medicine, Duke University, 101 Science Dr, DUMC 3382, Durham, NC, USA
- Division of Cardiology, Duke University, Durham, NC, USA
| | - Jordan Gales
- Department of Cardiology, Texas Heart Institute, Houston, TX, USA
| | - Ilya Zhbannikov
- Center for Applied Genomics & Precision Medicine, Duke University, 101 Science Dr, DUMC 3382, Durham, NC, USA
- Division of Cardiology, Duke University, Durham, NC, USA
| | - Thomas L Ortel
- Division of Hematology, Duke University, Durham, NC, USA
| | - Svati H Shah
- Division of Cardiology, Duke University, Durham, NC, USA
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - William E Kraus
- Division of Cardiology, Duke University, Durham, NC, USA
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - Geoffrey S Ginsburg
- All of Us Research Program, National Institutes of Health, Bethesda, MD, USA
| | - Deepak Voora
- Center for Applied Genomics & Precision Medicine, Duke University, 101 Science Dr, DUMC 3382, Durham, NC, USA
- Division of Cardiology, Duke University, Durham, NC, USA
| |
Collapse
|
3
|
Myers RA, Ortel TL, Waldrop A, Dave S, Ginsburg GS, Voora D. Aspirin effects on platelet gene expression are associated with a paradoxical, increase in platelet function. Br J Clin Pharmacol 2021; 88:2074-2083. [PMID: 34705291 PMCID: PMC9007832 DOI: 10.1111/bcp.15127] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/10/2021] [Accepted: 10/18/2021] [Indexed: 01/04/2023] Open
Abstract
Aspirin has known effects beyond inhibiting platelet cyclooxygenase-1 (COX-1) that have been incompletely characterized. Transcriptomics can comprehensively characterize the on- and off-target effects of medications. We used a systems pharmacogenomics approach of aspirin exposure in volunteers coupled with serial platelet function and purified platelet mRNA sequencing to test the hypothesis that aspirin's effects on the platelet transcriptome are associated with platelet function. We prospectively recruited 74 adult volunteers for a randomized crossover study of 81- vs. 325 mg/day, each for 4 weeks. Using mRNA sequencing of purified platelets collected before and after each 4-week exposure, we identified 208 aspirin-responsive genes with no evidence for dosage effects. In independent cohorts of healthy volunteers and patients with diabetes, we validated aspirin's effects on five genes: EIF2S3, CHRNB1, EPAS1, SLC9A3R2 and HLA-DRA. Functional characterization of the effects of aspirin on mRNA as well as platelet ribosomal RNA demonstrated that aspirin may act as an inhibitor of protein synthesis. Database searches for small molecules that mimicked the effects of aspirin on platelet gene expression in vitro identified aspirin but no other molecules that share aspirin's known mechanisms of action. The effects of aspirin on platelet mRNA were correlated with higher levels of platelet function both at baseline and after aspirin exposure-an effect that counteracts aspirin's known antiplatelet effect. In summary, this work collectively demonstrates a dose-independent effect of aspirin on the platelet transcriptome that counteracts the well-known antiplatelet effects of aspirin.
Collapse
Affiliation(s)
- Rachel A Myers
- Center for Applied Genomics & Precision Medicine, Duke University School of Medicine, Durham, NC, United States
| | - Thomas L Ortel
- Division of Hematology, Department of Medicine, Duke University Medical Center, Durham, NC, United States
| | - Alexander Waldrop
- Center for Genomics and Computational Biology, Duke University, Durham, NC, United States
| | - Sandeep Dave
- Center for Genomics and Computational Biology, Duke University, Durham, NC, United States
| | - Geoffrey S Ginsburg
- Center for Applied Genomics & Precision Medicine, Duke University School of Medicine, Durham, NC, United States
| | - Deepak Voora
- Center for Applied Genomics & Precision Medicine, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
4
|
Infeld M, Friede KA, San TR, Knickerbocker HJ, Ginsburg GS, Ortel TL, Voora D. Platelet reactivity in response to aspirin and ticagrelor in African-Americans and European-Americans. J Thromb Thrombolysis 2021; 51:249-259. [PMID: 33159252 PMCID: PMC7889728 DOI: 10.1007/s11239-020-02327-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/30/2020] [Indexed: 12/19/2022]
Abstract
Platelet gene polymorphisms are associated with variable on-treatment platelet reactivity and vary by race. Whether differences in platelet reactivity and aspirin or ticagrelor exist between African-American and European-Americans remains poorly understood. Biological samples from three prior prospective antiplatelet challenge studies at the Duke Clinical Research Unit were used to compare platelet reactivity between African-American and European-American subjects. Platelet reactivity at baseline, on-aspirin, on-ticagrelor, and the treatment effect of aspirin or ticagrelor were compared between groups using an adjusted mixed effects model. Compared with European-Americans (n = 282; 50% female; mean ± standard deviation age, 50 ± 16), African-Americans (n = 209; 67% female; age 48 ± 12) had lower baseline platelet reactivity with platelet function analyzer-100 (PFA-100) (p < 0.01) and with light transmission aggregometry (LTA) in response to arachidonic acid (AA), adenosine diphosphate (ADP), and epinephrine agonists (p < 0.05). African-Americans had lower platelet reactivity on aspirin in response to ADP, epinephrine, and collagen (p < 0.05) and on ticagrelor in response to AA, ADP, and collagen (p < 0.05). The treatment effect of aspirin was greater in European-Americans with an AA agonist (p = 0.002). Between-race differences with in vitro aspirin mirrored those seen in vivo. The treatment effect of ticagrelor was greater in European-Americans in response to ADP (p < 0.05) but with collagen, the treatment effect was greater for African-Americans (p < 0.05). Platelet reactivity was overall lower in African-Americans off-treatment, on aspirin, and on ticagrelor. European-Americans experienced greater platelet suppression on aspirin and on ticagrelor. The aspirin response difference in vivo and in vitro suggests a mechanism intrinsic to the platelet. Whether the absolute level of platelet reactivity or the degree of platelet suppression after treatment is more important for clinical outcomes is uncertain.
Collapse
Affiliation(s)
- Margaret Infeld
- Division of Cardiology, Department of Medicine, Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Kevin A Friede
- Division of Cardiology, Department of Medicine, Duke University, Durham, NC, USA
| | - Tan Ru San
- Department of Cardiology, National Heart Centre, Singapore, Singapore
| | - Holly J Knickerbocker
- Center for Applied Genomics & Precision Medicine, Duke University, 2187 CIEMAS, Campus Box 3382, Durham, NC, 27708, USA
| | - Geoffrey S Ginsburg
- Center for Applied Genomics & Precision Medicine, Duke University, 2187 CIEMAS, Campus Box 3382, Durham, NC, 27708, USA
- Division of Cardiology, Department of Medicine, Duke University, Durham, NC, USA
| | - Thomas L Ortel
- Division of Hematology, Duke University, Durham, NC, USA
| | - Deepak Voora
- Center for Applied Genomics & Precision Medicine, Duke University, 2187 CIEMAS, Campus Box 3382, Durham, NC, 27708, USA.
- Division of Cardiology, Department of Medicine, Duke University, Durham, NC, USA.
| |
Collapse
|
5
|
Zhou B, Wang Q, Zhao Z, Feng X. A Systematic Review of the Efficacy and Safety of Aspirin When Delivered at Different Medication Times for the Primary and Secondary Prevention of Cardiovascular and Cerebrovascular Diseases. Ther Innov Regul Sci 2020; 54:1339-1348. [PMID: 33258099 DOI: 10.1007/s43441-020-00156-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/19/2019] [Indexed: 10/23/2022]
Abstract
PURPOSE Effective antiplatelet therapy can significantly reduce the incidence and mortality rate of cardiovascular and cerebrovascular diseases. Aspirin is widely used in the secondary prevention of cardiovascular and cerebrovascular diseases; however, there is widespread debate as to when patients should take an enteric-coated aspirin tablet on a daily basis. In the present study, we evaluated the efficacy and safety of different aspirin medication times (morning or before bedtime) in terms of the primary and secondary prevention of cardiovascular and cerebrovascular diseases using meta-analysis. METHODS Studies with randomized control trials (RCT) or crossover trials regarding to the usage of aspirin (morning or before bedtime) for the primary or secondary prevention of cardiovascular and cerebrovascular diseases were searched in Medline, EMbase, Cochrane Library, CNKI, Wanfang Data, VIP Database and CBM. Review Manager 5 (RevMan 5, v5.3), a Cochrane systematic reviews software, was used to perform meta-analysis based on the recommendation of the Cochrane Handbook for risk assessment tools. RESULTS Meta-analysis showed that taking low-dose aspirin tablets before bed reduced systolic and diastolic blood pressure compared with taking it in the morning. At the same time, the number of studies on platelet aggregation rate, C-reactive protein (CRP), serum nitric oxide (NO) or thromboxane B2 (TXB2) is too small to be reliable. However, there was a large heterogeneity across the studies. The quality of some studies was not high enough. CONCLUSION Additional blood pressure benefits can be achieved by taking aspirin before bedtime, but it does not affect its antiplatelet effect and does not pose a higher risk of bleeding.
Collapse
Affiliation(s)
- Boya Zhou
- Pharmacy Department, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, No. 17, Qihelou Street, Dongcheng District, Beijing, 100006, People's Republic of China
| | - Qiaoyu Wang
- Pharmacy Department, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, People's Republic of China
| | - Zhigang Zhao
- Pharmacy Department, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, People's Republic of China
| | - Xin Feng
- Pharmacy Department, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, No. 17, Qihelou Street, Dongcheng District, Beijing, 100006, People's Republic of China.
| |
Collapse
|
6
|
Friede KA, Infeld MM, Tan RS, Knickerbocker HJ, Myers RA, Dubois LG, Thompson JW, Kaddurah‐Daouk R, Ginsburg GS, Ortel TL, Voora D. Influence of Sex on Platelet Reactivity in Response to Aspirin. J Am Heart Assoc 2020; 9:e014726. [PMID: 32654613 PMCID: PMC7660714 DOI: 10.1161/jaha.119.014726] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 05/06/2020] [Indexed: 01/29/2023]
Abstract
Background There are sex differences in the efficacy and safety of aspirin for the prevention of myocardial infarction and stroke. Whether this is explained by underlying differences in platelet reactivity and aspirin response remains poorly understood. Methods and Results Healthy volunteers (n=378 208 women) and patients with coronary artery disease or coronary artery disease risk factors (n=217 112 women) took aspirin for 4 weeks. Light transmittance aggregometry using platelet-rich plasma was used to measure platelet reactivity in response to epinephrine, collagen, and ADP at baseline, 3 hours after the first aspirin dose, and after 4 weeks of daily aspirin therapy. A subset of patients underwent pharmacokinetic and pharmacodynamic assessment with levels of salicylate and cyclooxygenase-1-derived prostaglandin metabolites and light transmittance aggregometry in response to arachidonic acid and after ex vivo exposure to aspirin. At baseline, women had increased platelet aggregation in response to ADP and collagen. Innate platelet response to aspirin, assessed with ex vivo aspirin exposure of baseline platelets, did not differ by sex. Three hours after the first oral aspirin dose, platelet aggregation was inhibited in women to a greater degree in response to epinephrine and to a lesser degree with collagen. After 4 weeks of daily therapy, despite higher salicylate concentrations and greater cyclooxygenase-1 inhibition, women exhibited an attenuation of platelet inhibition in response to epinephrine and ADP. Conclusions We observed agonist-dependent sex differences in platelet responses to aspirin. Despite higher cyclooxygenase-1 inhibition, daily aspirin exposure resulted in a paradoxical attenuation of platelet inhibition in response to epinephrine and ADP over time in women but not in men.
Collapse
Affiliation(s)
- Kevin A. Friede
- Division of CardiologyDuke UniversityDurhamNC
- Center for Applied Genomics & Precision MedicineDuke UniversityDurhamNC
| | - Margaret M. Infeld
- Division of CardiologyLarner College of Medicine at the University of VermontBurlingtonVT
| | - Ru San Tan
- Department of CardiologyNational Heart CentreSingapore
| | | | - Rachel A. Myers
- Center for Applied Genomics & Precision MedicineDuke UniversityDurhamNC
| | - Laura G. Dubois
- Center for Genomic and Computational BiologyDuke UniversityDurhamNC
| | - J. Will Thompson
- Center for Genomic and Computational BiologyDuke UniversityDurhamNC
| | | | - Geoffrey S. Ginsburg
- Division of CardiologyDuke UniversityDurhamNC
- Center for Applied Genomics & Precision MedicineDuke UniversityDurhamNC
| | | | - Deepak Voora
- Division of CardiologyDuke UniversityDurhamNC
- Center for Applied Genomics & Precision MedicineDuke UniversityDurhamNC
| |
Collapse
|
7
|
Furtado RHM, Giugliano RP, Dalcoquio TF, Arantes FBB, Barbosa CJDG, Genestreti PRR, Franci A, Menezes FR, Nakashima CAK, Scanavini Filho MA, Ferrari AG, Salsoso R, Baracioli LM, Nicolau JC. Increased bodyweight and inadequate response to aspirin in individuals with coronary artery disease. J Thromb Thrombolysis 2019; 48:217-224. [PMID: 30879169 DOI: 10.1007/s11239-019-01830-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Recent reports have suggested that aspirin effect might be influenced by bodyweight, with decreased efficacy in heavier individuals. We investigated the influence of bodyweight on aspirin pharmacodynamics in two independent datasets of patients taking non-enteric coated aspirin 100 mg QD for coronary artery disease (CAD). In the first dataset, 368 patients had their platelet aggregation assessed using VerifyNow Aspirin and measured in Aspirin Reaction Units (ARU). In the second dataset, 70 patients had serum thromboxane B2 (TXB2) dosage assessed by an ELISA assay and measured in pg/mL. Platelet aggregation was independently associated with bodyweight, with 8.41 (95% CI 1.86-14.97; adjusted p-value = 0.012) increase in ARU for every 10 kg. Furthermore, the rate of non-response to aspirin (defined as ARU ≥ 550) was significantly associated with increased bodyweight (adjusted p-value = 0.007), with OR = 1.23 (95% CI 1.06-1.42) for every 10 kg. Similar results were found considering body mass index (in kg/m2), with 15.5 (95% CI 5.0 to 25.9; adjusted p-value = 0.004) increase in ARU for every 10 kg and non-response OR = 1.43 (95% CI 1.13 to 1.81, adjusted p-value = 0.003) for every 5 kg/m2. Moreover, serum TXB2 was higher in patients weighting more than 70 kg (222.6 ± 62.9 versus 194.9 ± 61.9 pg/mL; adjusted p-value = 0.018). In two different datasets of patients with CAD on non-enteric coated aspirin 100 mg QD, increased bodyweight was independently associated with impaired response to aspirin.
Collapse
Affiliation(s)
- Remo H M Furtado
- Instituto do Coracao (InCor), Unidade de Coronariopatia Aguda, Hospital das Clinicas, Faculdade de Medicina (HCFMUSP), Universidade de Sao Paulo, São Paulo, Brazil.,TIMI Study Group - Brigham and Women's Hospital, Boston, MA, USA
| | | | - Talia F Dalcoquio
- Instituto do Coracao (InCor), Unidade de Coronariopatia Aguda, Hospital das Clinicas, Faculdade de Medicina (HCFMUSP), Universidade de Sao Paulo, São Paulo, Brazil
| | - Flavia B B Arantes
- Instituto do Coracao (InCor), Unidade de Coronariopatia Aguda, Hospital das Clinicas, Faculdade de Medicina (HCFMUSP), Universidade de Sao Paulo, São Paulo, Brazil.,Universidade Federal de Uberlandia, Uberlandia, Minas Gerais, Brazil
| | - Carlos J D G Barbosa
- Instituto do Coracao (InCor), Unidade de Coronariopatia Aguda, Hospital das Clinicas, Faculdade de Medicina (HCFMUSP), Universidade de Sao Paulo, São Paulo, Brazil.,Hospital do Coracao do Brasil, Brasília, Distrito Federal, Brazil
| | - Paulo R R Genestreti
- Instituto do Coracao (InCor), Unidade de Coronariopatia Aguda, Hospital das Clinicas, Faculdade de Medicina (HCFMUSP), Universidade de Sao Paulo, São Paulo, Brazil
| | - André Franci
- Instituto do Coracao (InCor), Unidade de Coronariopatia Aguda, Hospital das Clinicas, Faculdade de Medicina (HCFMUSP), Universidade de Sao Paulo, São Paulo, Brazil
| | - Fernando R Menezes
- Instituto do Coracao (InCor), Unidade de Coronariopatia Aguda, Hospital das Clinicas, Faculdade de Medicina (HCFMUSP), Universidade de Sao Paulo, São Paulo, Brazil
| | - Carlos A K Nakashima
- Instituto do Coracao (InCor), Unidade de Coronariopatia Aguda, Hospital das Clinicas, Faculdade de Medicina (HCFMUSP), Universidade de Sao Paulo, São Paulo, Brazil
| | - Marco A Scanavini Filho
- Instituto do Coracao (InCor), Unidade de Coronariopatia Aguda, Hospital das Clinicas, Faculdade de Medicina (HCFMUSP), Universidade de Sao Paulo, São Paulo, Brazil
| | - Aline G Ferrari
- Instituto do Coracao (InCor), Unidade de Coronariopatia Aguda, Hospital das Clinicas, Faculdade de Medicina (HCFMUSP), Universidade de Sao Paulo, São Paulo, Brazil
| | - Rocio Salsoso
- Instituto do Coracao (InCor), Unidade de Coronariopatia Aguda, Hospital das Clinicas, Faculdade de Medicina (HCFMUSP), Universidade de Sao Paulo, São Paulo, Brazil
| | - Luciano M Baracioli
- Instituto do Coracao (InCor), Unidade de Coronariopatia Aguda, Hospital das Clinicas, Faculdade de Medicina (HCFMUSP), Universidade de Sao Paulo, São Paulo, Brazil
| | - Jose C Nicolau
- Instituto do Coracao (InCor), Unidade de Coronariopatia Aguda, Hospital das Clinicas, Faculdade de Medicina (HCFMUSP), Universidade de Sao Paulo, São Paulo, Brazil.
| |
Collapse
|
8
|
Lowenstern A, Storey RF, Neely M, Sun JL, Angiolillo DJ, Cannon CP, Himmelmann A, Huber K, James SK, Katus HA, Morais J, Siegbahn A, Steg PG, Wallentin L, Becker RC. Platelet-related biomarkers and their response to inhibition with aspirin and p2y 12-receptor antagonists in patients with acute coronary syndrome. J Thromb Thrombolysis 2018; 44:145-153. [PMID: 28608165 DOI: 10.1007/s11239-017-1516-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The PLATelet inhibition and patient Outcomes (PLATO) trial showed that treatment with ticagrelor reduced the rate of death due to vascular causes, myocardial infarction and stroke when compared to clopidogrel in patients with ST-elevation or non-ST-elevation acute coronary syndrome (ACS). While the comparative benefit of ticagrelor over clopidogrel increased over time, event rates accrued in both groups during the study period. The purpose of our biomarker-based exploratory analysis was to determine whether long-term platelet inhibition may be associated with platelet adaptation. A sample of 4000 participants from the PLATO trial also consented to participate in a prospectively designed biomarker substudy. Blood samples were procured at baseline, immediately prior to hospital discharge and at 1 and 6 months. Markers of platelet activity, including platelet count, serum CD40-ligand and soluble P-selectin were analyzed. Mean levels were compared at discharge, 1 and 6 months following study drug initiation-first for all patients and subsequently stratified by treatment group. A linear mixed model was used to estimate the short-term change rate (baseline to 1 month) and long-term change rate (1-6 months) for each biomarker. A Cox proportional hazards model was used to calculate hazard ratios for each change in biomarker over the two time periods examined: baseline to 1 month and 1 to 6 months. Prior to randomized treatment (baseline), sCD40 ligand and sP-selectin levels were elevated above the normal range of the assay (0.39 and 33.5 µg/L, respectively). The mean level of each biomarker was significantly different at 1 month compared to baseline (p < 0.0001). When stratified by treatment group, at 1 month patients treated with ticagrelor had a larger increase in platelet count compared to those treated with clopidogrel (p < 0.0001). Similarly, when comparing biomarker levels for all patients at 6 months with those at 1 month, each differed significantly (p < 0.05). There was no significant difference between treatment groups during this time period. The rate of change for both platelet count and sP-selectin were significantly different between baseline and 1 month when compared to the 1 to 6-month time period (p < 0.0001). When comparing treatment groups, the rate of increase in platelets from baseline to 1 month was greater for patients treated with ticagrelor (p < 0.0001). This was no longer observed in the 1 to 6-month interval. Using a Cox proportional hazard model, the increase in platelet count from 1 to 6 months was associated with ischemic-thrombotic events, while sCD40 ligand decrease from 1 to 6 months was associated with hemorrhagic events. There were no differences between treatment groups for the associations with clinical endpoints. Dynamic changes in platelet count, sCD-40 ligand and sP-selectin occur over time among patients with ACS. Platelet-directed therapy with a P2Y12 receptor inhibitor in combination with aspirin modestly impacts the expression of these biomarkers. Platelet count and sCD40 ligand may offer modest overall predictive value for future ischemic-thrombotic or hemorrhagic clinical events, respectively. The existence of a platelet adaptome and its overall clinical significance among patients at risk for thrombotic events will require a more in-depth and platelet-biology specific investigation.
Collapse
Affiliation(s)
- Angela Lowenstern
- Duke Clinical Research Institute, Duke University, Medical Center, 2301 Erwin Road, DUMC Box 2845, Durham, NC, 27710, USA.
| | - Robert F Storey
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Megan Neely
- Duke Clinical Research Institute, Duke University, Medical Center, 2301 Erwin Road, DUMC Box 2845, Durham, NC, 27710, USA
| | - Jie-Lena Sun
- Duke Clinical Research Institute, Duke University, Medical Center, 2301 Erwin Road, DUMC Box 2845, Durham, NC, 27710, USA
| | - Dominick J Angiolillo
- Division of Cardiology, University of Florida College of Medicine-Jacksonville, Jacksonville, FL, USA
| | | | | | - Kurt Huber
- 3rd Department of Medicine, Cardiology and Intensive Care, Wilhelminen Hospital, Vienna, Austria.,Sigmund Freud Private University, Medical School, Vienna, Austria
| | - Stefan K James
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden.,Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Hugo A Katus
- Medizinishe Klinik, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | | | - Agneta Siegbahn
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden.,Department of Medical Sciences, Clinical Chemistry, Uppsala University, Uppsala, Sweden
| | - Phillippe Gabriel Steg
- Département Hospitalo-Universitaire FIRE, AP-HP, Hôpital Bichat, Paris, France.,Paris Diderot University, Sorbonne Paris Cité, Paris, France.,NHLI Imperial College, ICMS, Royal Brompton Hospital, London, UK.,FACT (French Alliance for Cardiovascular Trials), an F-CRIN network, INSERM U1148, Paris, France
| | - Lars Wallentin
- Department of Medical Sciences, Cardiology, Uppsala University, Uppsala, Sweden.,Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Richard C Becker
- Division of Cardiovascular Health and Disease, Heart, Lung and Vascular Institute, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | | |
Collapse
|
9
|
Platelet Carbonic Anhydrase II, a Forgotten Enzyme, May Be Responsible for Aspirin Resistance. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:3132063. [PMID: 29090039 PMCID: PMC5635279 DOI: 10.1155/2017/3132063] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 08/22/2017] [Accepted: 09/06/2017] [Indexed: 12/11/2022]
Abstract
Background Thromboembolic events constitute a major health problem, despite the steadily expanding arsenal of antiplatelet drugs. Hence, there is still a need to optimize the antiplatelet therapy. Objectives The aim of our study was to verify a hypothesis that there are no differences in platelet proteome between two groups of healthy people representing different acetylsalicylic acid (aspirin) responses as assessed by the liquid chromatography/mass spectrometry (LC/MS) technique. Patients/Methods A total of 61 healthy volunteers were recruited for the study. Physical examination and blood collection were followed by platelet-rich plasma aggregation assays and platelet separation for proteomic LC/MS analysis. Arachidonic acid- (AA-) induced aggregation (in the presence of aspirin) allowed to divide study participants into two groups aspirin-resistant (AR) and aspirin-sensitive (AS) ones. Subsequently, platelet proteome was compared in groups using the LC/MS analysis. Results The LC/MS analysis of platelet proteome between groups revealed that out of all identified proteins, the only discriminatory protein, affecting aspirin responsiveness, is platelet carbonic anhydrase II (CA II). Conclusions CA II is a platelet function modulator and should be taken into consideration as a cardiovascular event risk factor or therapeutic target.
Collapse
|
10
|
Systems Pharmacogenomics Finds RUNX1 Is an Aspirin-Responsive Transcription Factor Linked to Cardiovascular Disease and Colon Cancer. EBioMedicine 2016; 11:157-164. [PMID: 27566955 PMCID: PMC5049978 DOI: 10.1016/j.ebiom.2016.08.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 08/10/2016] [Accepted: 08/12/2016] [Indexed: 12/21/2022] Open
Abstract
Aspirin prevents cardiovascular disease and colon cancer; however aspirin's inhibition of platelet COX-1 only partially explains its diverse effects. We previously identified an aspirin response signature (ARS) in blood consisting of 62 co-expressed transcripts that correlated with aspirin's effects on platelets and myocardial infarction (MI). Here we report that 60% of ARS transcripts are regulated by RUNX1 - a hematopoietic transcription factor - and 48% of ARS gene promoters contain a RUNX1 binding site. Megakaryocytic cells exposed to aspirin and its metabolite (salicylic acid, a weak COX-1 inhibitor) showed up regulation in the RUNX1 P1 isoform and MYL9, which is transcriptionally regulated by RUNX1. In human subjects, RUNX1 P1 expression in blood and RUNX1-regulated platelet proteins, including MYL9, were aspirin-responsive and associated with platelet function. In cardiovascular disease patients RUNX1 P1 expression was associated with death or MI. RUNX1 acts as a tumor suppressor gene in gastrointestinal malignancies. We show that RUNX1 P1 expression is associated with colon cancer free survival suggesting a role for RUNX1 in aspirin's protective effect in colon cancer. Our studies reveal an effect of aspirin on RUNX1 and gene expression that may additionally explain aspirin's effects in cardiovascular disease and cancer.
Collapse
|
11
|
Fanaroff AC, Roe MT. Contemporary Reflections on the Safety of Long-Term Aspirin Treatment for the Secondary Prevention of Cardiovascular Disease. Drug Saf 2016; 39:715-27. [PMID: 27028617 PMCID: PMC5778440 DOI: 10.1007/s40264-016-0421-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Aspirin has been the cornerstone of therapy for the secondary prevention treatment of patients with cardiovascular disease since landmark trials were completed in the late 1970s and early 1980s that demonstrated the efficacy of aspirin for reducing the risk of ischemic events. Notwithstanding the consistent benefits demonstrated with aspirin for both acute and chronic cardiovascular disease, there are a number of toxicities associated with aspirin that have been showcased by recent long-term clinical trials that have included an aspirin monotherapy arm. As an inhibitor of cyclooxygenase (COX), aspirin impairs gastric mucosal protective mechanisms. Previous trials have shown that up to 15-20 % of patients developed gastrointestinal symptoms with aspirin monotherapy, and approximately 1 % of patients per year had a clinically significant bleeding event, including 1 in 1000 patients who suffered an intracranial or fatal bleed. These risks have been shown to be compounded for patients with acute coronary syndromes (ACS) and those undergoing percutaneous coronary intervention (PCI) who are also treated with other antithrombotic agents during the acute care/procedural period, as well as for an extended time period afterwards. Given observations of substantial increases in bleeding rates from many prior long-term clinical trials that have evaluated aspirin together with other oral platelet inhibitors or oral anticoagulants, the focus of contemporary research has pivoted towards tailored antithrombotic regimens that attempt to either shorten the duration of exposure to aspirin or replace aspirin with an alternative antithrombotic agent. While these shifts are occurring, the safety profile of aspirin when used for the secondary prevention treatment of patients with established cardiovascular disease deserves further consideration.
Collapse
Affiliation(s)
- Alexander C Fanaroff
- Duke Clinical Research Institute, Duke University Medical Center, 2400 Pratt Street, Room 7035, Durham, NC, 27705, USA
| | - Matthew T Roe
- Duke Clinical Research Institute, Duke University Medical Center, 2400 Pratt Street, Room 7035, Durham, NC, 27705, USA.
| |
Collapse
|
12
|
Pretreatment with low-dose gadolinium chloride attenuates myocardial ischemia/reperfusion injury in rats. Acta Pharmacol Sin 2016; 37:453-62. [PMID: 26948086 DOI: 10.1038/aps.2015.156] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 12/25/2015] [Indexed: 12/14/2022]
Abstract
AIM We have shown that low-dose gadolinium chloride (GdCl3) abolishes arachidonic acid (AA)-induced increase of cytoplasmic Ca(2+), which is known to play a crucial role in myocardial ischemia/reperfusion (I/R) injury. The present study sought to determine whether low-dose GdCl3 pretreatment protected rat myocardium against I/R injury in vitro and in vivo. METHODS Cultured neonatal rat ventricular myocytes (NRVMs) were treated with GdCl3 or nifedipine, followed by exposure to anoxia/reoxygenation (A/R). Cell apoptosis was detected; the levels of related signaling molecules were assessed. SD rats were intravenously injected with GdCl3 or nifedipine. Thirty min after the administration the rats were subjected to LAD coronary artery ligation followed by reperfusion. Infarction size, the release of serum myocardial injury markers and AA were measured; cell apoptosis and related molecules were assessed. RESULTS In A/R-treated NRVMs, pretreatment with GdCl3 (2.5, 5, 10 μmol/L) dose-dependently inhibited caspase-3 activation, death receptor-related molecules DR5/Fas/FADD/caspase-8 expression, cytochrome c release, AA release and sustained cytoplasmic Ca(2+) increases induced by exogenous AA. In I/R-treated rats, pre-administration of GdCl3 (10 mg/kg) significantly reduced the infarct size, and the serum levels of CK-MB, cardiac troponin-I, LDH and AA. Pre-administration of GdCl3 also significantly decreased the number of apoptotic cells, caspase-3 activity, death receptor-related molecules (DR5/Fas/FADD) expression and cytochrome c release in heart tissues. The positive control drug nifedipine produced comparable cardioprotective effects in vitro and in vivo. CONCLUSION Pretreatment with low-dose GdCl3 significantly attenuates I/R-induced myocardial apoptosis in rats by suppressing activation of both death receptor and mitochondria-mediated pathways.
Collapse
|
13
|
Yang Y, Lewis JP, Hulot JS, Scott SA. The pharmacogenetic control of antiplatelet response: candidate genes and CYP2C19. Expert Opin Drug Metab Toxicol 2015; 11:1599-617. [PMID: 26173871 DOI: 10.1517/17425255.2015.1068757] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Aspirin, clopidogrel, prasugrel and ticagrelor are antiplatelet agents for the prevention of ischemic events in patients with acute coronary syndromes (ACS), percutaneous coronary intervention (PCI) and other indications. Variability in response is observed to different degrees with these agents, which can translate to increased risks for adverse cardiovascular events. As such, potential pharmacogenetic determinants of antiplatelet pharmacokinetics, pharmacodynamics and clinical outcomes have been actively studied. AREAS COVERED This article provides an overview of the available antiplatelet pharmacogenetics literature. Evidence supporting the significance of candidate genes and their potential influence on antiplatelet response and clinical outcomes are summarized and evaluated. Additional focus is directed at CYP2C19 and clopidogrel response, including the availability of clinical testing and genotype-directed antiplatelet therapy. EXPERT OPINION The reported aspirin response candidate genes have not been adequately replicated and few candidate genes have thus far been implicated in prasugrel or ticagrelor response. However, abundant data support the clinical validity of CYP2C19 and clopidogrel response variability among ACS/PCI patients. Although limited prospective trial data are available to support the utility of routine CYP2C19 testing, the increased risks for reduced clopidogrel efficacy among ACS/PCI patients that carry CYP2C19 loss-of-function alleles should be considered when genotype results are available.
Collapse
Affiliation(s)
- Yao Yang
- a 1 Icahn School of Medicine at Mount Sinai, Department of Genetics and Genomic Sciences , New York, NY, USA +1 212 241 3780 ; +1 212 241 0139 ;
| | - Joshua P Lewis
- b 2 University of Maryland School of Medicine, Division of Endocrinology, Diabetes and Nutrition, and Program for Personalized and Genomic Medicine , Baltimore, MD, USA
| | - Jean-Sébastien Hulot
- c 3 Icahn School of Medicine at Mount Sinai, Cardiovascular Research Center , New York, NY, USA.,d 4 Sorbonne Universités, UPMC Univ Paris 06, INSERM , UMR_S 1166 ICAN, F-75005 Paris, France
| | - Stuart A Scott
- a 1 Icahn School of Medicine at Mount Sinai, Department of Genetics and Genomic Sciences , New York, NY, USA +1 212 241 3780 ; +1 212 241 0139 ;
| |
Collapse
|
14
|
Jiang W, Boyle SH, Ortel TL, Samad Z, Velazquez EJ, Harrison RW, Wilson J, Kuhn C, Williams RB, O’Connor CM, Becker RC. Platelet aggregation and mental stress induced myocardial ischemia: Results from the Responses of Myocardial Ischemia to Escitalopram Treatment (REMIT) study. Am Heart J 2015; 169:496-507.e1. [PMID: 25819856 PMCID: PMC4382806 DOI: 10.1016/j.ahj.2014.12.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Accepted: 12/15/2014] [Indexed: 12/23/2022]
Abstract
BACKGROUND Mental stress-induced myocardial ischemia (MSIMI) is common in patients with ischemic heart disease (IHD) and associated with a poorer cardiovascular prognosis. Platelet hyperactivity is an important factor in acute coronary syndrome. This study examined associations between MSIMI and resting and mental stress-induced platelet activity. METHODS Eligible patients with clinically stable IHD underwent a battery of 3 mental stress tests during the recruitment phase of REMIT study. MSIMI was assessed by echocardiography and electrocardiography. Ex vivo platelet aggregation in response to ADP, epinephrine, collagen, serotonin, and combinations of serotonin plus ADP, epinephrine, and collagen were evaluated as was platelet serotonin transporter expression. RESULTS Of the 270 participants who completed mental stress testing, and had both resting and post-stress platelet aggregation evaluation , 43.33% (n=117) met criteria for MSIMI and 18.15% (n=49) had normal left ventricular response to stress (NLVR). The MSIMI group, relative to the NLVR groups, demonstrated heightened mental stress-induced aggregation responses, as measured by area under the curve, to collagen 10μM (6.95[5.54] vs. -14.23[8.75].; P=0.045), epinephrine 10μM (12.84[4.84] vs. -6.40[7.61].; P=0.037) and to serotonin 10 μM plus ADP 1 μM (6.64[5.29] vs. -27.34[8.34]; P<.001). The resting platelet aggregation and serotonin transporter expression, however, were not different between the two groups. CONCLUSIONS These findings suggest that the dynamic change of platelet aggregation caused by mental stress may underlie MSIMI. While the importance of these findings requires additional investigation, they raise concern given the recognized relationship between mental stress-induced platelet hyperactivity and cardiovascular events in patients with IHD.
Collapse
|
15
|
Wang Z, Maity A, Hsiao CK, Voora D, Kaddurah-Daouk R, Tzeng JY. Module-based association analysis for omics data with network structure. PLoS One 2015; 10:e0122309. [PMID: 25822417 PMCID: PMC4378989 DOI: 10.1371/journal.pone.0122309] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 02/20/2015] [Indexed: 02/06/2023] Open
Abstract
Module-based analysis (MBA) aims to evaluate the effect of a group of biological elements sharing common features, such as SNPs in the same gene or metabolites in the same pathways, and has become an attractive alternative to traditional single bio-element approaches. Because bio-elements regulate and interact with each other as part of network, incorporating network structure information can more precisely model the biological effects, enhance the ability to detect true associations, and facilitate our understanding of the underlying biological mechanisms. However, most MBA methods ignore the network structure information, which depicts the interaction and regulation relationship among basic functional units in biology system. We construct the connectivity kernel and the topology kernel to capture the relationship among bio-elements in a module, and use a kernel machine framework to evaluate the joint effect of bio-elements. Our proposed kernel machine approach directly incorporates network structure so to enhance the study efficiency; it can assess interactions among modules, account covariates, and is computational efficient. Through simulation studies and real data application, we demonstrate that the proposed network-based methods can have markedly better power than the approaches ignoring network information under a range of scenarios.
Collapse
Affiliation(s)
- Zhi Wang
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina, 27695, United States of America
| | - Arnab Maity
- Department of Statistics, North Carolina State University, Raleigh, North Carolina, 27695, United States of America
| | - Chuhsing Kate Hsiao
- Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Deepak Voora
- Institute for Genome Sciences and Policy, Duke University, Durham, North Carolina, United States of America
| | - Rima Kaddurah-Daouk
- Department of Psychiatry and Behavioral Sciences, Duke University, Durham, North Carolina, United States of America
| | - Jung-Ying Tzeng
- Bioinformatics Research Center, North Carolina State University, Raleigh, North Carolina, 27695, United States of America
- Department of Statistics, North Carolina State University, Raleigh, North Carolina, 27695, United States of America
- Department of Statistics, National Cheng-Kung University, Taiwan, R.O.C
| |
Collapse
|
16
|
Beitelshees AL, Voora D, Lewis JP. Personalized antiplatelet and anticoagulation therapy: applications and significance of pharmacogenomics. Pharmgenomics Pers Med 2015; 8:43-61. [PMID: 25897256 PMCID: PMC4397717 DOI: 10.2147/pgpm.s52900] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In recent years, substantial effort has been made to better understand the influence of genetic factors on the efficacy and safety of numerous medications. These investigations suggest that the use of pharmacogenetic data to inform physician decision-making has great potential to enhance patient care by reducing on-treatment clinical events, adverse drug reactions, and health care-related costs. In fact, integration of such information into the clinical setting may be particularly applicable for antiplatelet and anticoagulation therapeutics, given the increasing body of evidence implicating genetic variation in variable drug response. In this review, we summarize currently available pharmacogenetic information for the most commonly used antiplatelet (ie, clopidogrel and aspirin) and anticoagulation (ie, warfarin) medications. Furthermore, we highlight the currently known role of genetic variability in response to next-generation antiplatelet (prasugrel and ticagrelor) and anticoagulant (dabigatran) agents. While compelling evidence suggests that genetic variants are important determinants of antiplatelet and anticoagulation therapy response, significant barriers to clinical implementation of pharmacogenetic testing exist and are described herein. In addition, we briefly discuss development of new diagnostic targets and therapeutic strategies as well as implications for enhanced patient care. In conclusion, pharmacogenetic testing can provide important information to assist clinicians with prescribing the most personalized and effective antiplatelet and anticoagulation therapy. However, several factors may limit its usefulness and should be considered.
Collapse
Affiliation(s)
- Amber L Beitelshees
- Program for Personalized and Genomic Medicine and Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Deepak Voora
- Center for Applied Genomics & Precision Medicine, Department of Medicine, Duke School of Medicine, Durham, NC, USA
| | - Joshua P Lewis
- Program for Personalized and Genomic Medicine and Division of Endocrinology, Diabetes, and Nutrition, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
17
|
Voora D, Ginsburg GS, Akerblom A. Platelet RNA as a novel biomarker for the response to antiplatelet therapy. Future Cardiol 2014; 10:9-12. [PMID: 24344654 DOI: 10.2217/fca.13.90] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- Deepak Voora
- Duke Clinical Research Institute, Durham, NC, USA
| | | | | |
Collapse
|
18
|
Affiliation(s)
- Deepak Voora
- From the Department of Medicine, Center for Personalized and Precision Medicine, Duke University, Durham, NC (D.V.); and Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH (R.C.B.).
| | - Richard C Becker
- From the Department of Medicine, Center for Personalized and Precision Medicine, Duke University, Durham, NC (D.V.); and Division of Cardiovascular Health and Disease, University of Cincinnati College of Medicine, Cincinnati, OH (R.C.B.)
| |
Collapse
|
19
|
Zaas AK, Burke T, Chen M, McClain M, Nicholson B, Veldman T, Tsalik EL, Fowler V, Rivers EP, Otero R, Kingsmore SF, Voora D, Lucas J, Hero AO, Carin L, Woods CW, Ginsburg GS. A host-based RT-PCR gene expression signature to identify acute respiratory viral infection. Sci Transl Med 2014; 5:203ra126. [PMID: 24048524 DOI: 10.1126/scitranslmed.3006280] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Improved ways to diagnose acute respiratory viral infections could decrease inappropriate antibacterial use and serve as a vital triage mechanism in the event of a potential viral pandemic. Measurement of the host response to infection is an alternative to pathogen-based diagnostic testing and may improve diagnostic accuracy. We have developed a host-based assay with a reverse transcription polymerase chain reaction (RT-PCR) TaqMan low-density array (TLDA) platform for classifying respiratory viral infection. We developed the assay using two cohorts experimentally infected with influenza A H3N2/Wisconsin or influenza A H1N1/Brisbane, and validated the assay in a sample of adults presenting to the emergency department with fever (n = 102) and in healthy volunteers (n = 41). Peripheral blood RNA samples were obtained from individuals who underwent experimental viral challenge or who presented to the emergency department and had microbiologically proven viral respiratory infection or systemic bacterial infection. The selected gene set on the RT-PCR TLDA assay classified participants with experimentally induced influenza H3N2 and H1N1 infection with 100 and 87% accuracy, respectively. We validated this host gene expression signature in a cohort of 102 individuals arriving at the emergency department. The sensitivity of the RT-PCR test was 89% [95% confidence interval (CI), 72 to 98%], and the specificity was 94% (95% CI, 86 to 99%). These results show that RT-PCR-based detection of a host gene expression signature can classify individuals with respiratory viral infection and sets the stage for prospective evaluation of this diagnostic approach in a clinical setting.
Collapse
Affiliation(s)
- Aimee K Zaas
- Institute for Genome Sciences and Policy, Duke University School of Medicine, Durham, NC 27710, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Aspirin exposure reveals novel genes associated with platelet function and cardiovascular events. J Am Coll Cardiol 2013; 62:1267-1276. [PMID: 23831034 DOI: 10.1016/j.jacc.2013.05.073] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2013] [Revised: 04/23/2013] [Accepted: 05/05/2013] [Indexed: 12/17/2022]
Abstract
OBJECTIVES The aim of this study was to develop ribonucleic acid (RNA) profiles that could serve as novel biomarkers for the response to aspirin. BACKGROUND Aspirin reduces death and myocardial infarction (MI), suggesting that aspirin interacts with biological pathways that may underlie these events. METHODS Aspirin was administered, followed by whole-blood RNA microarray profiling, in a discovery cohort of healthy volunteers (HV1) (n = 50) and 2 validation cohorts of healthy volunteers (HV2) (n = 53) and outpatient cardiology patients (OPC) (n = 25). Platelet function was assessed using the platelet function score (PFS) in HV1 and HV2 and the VerifyNow Aspirin Test (Accumetrics, Inc., San Diego, California) in OPC. Bayesian sparse factor analysis identified sets of coexpressed transcripts, which were examined for associations with PFS in HV1 and validated in HV2 and OPC. Proteomic analysis confirmed the association of validated transcripts in platelet proteins. Validated gene sets were tested for association with death or MI in 2 patient cohorts (n = 587 total) from RNA samples collected at cardiac catheterization. RESULTS A set of 60 coexpressed genes named the "aspirin response signature" (ARS) was associated with PFS in HV1 (r = -0.31, p = 0.03), HV2 (r = -0.34, Bonferroni p = 0.03), and OPC (p = 0.046). Corresponding proteins for the 17 ARS genes were identified in the platelet proteome, of which 6 were associated with PFS. The ARS was associated with death or MI in both patient cohorts (odds ratio: 1.2 [p = 0.01]; hazard ratio: 1.5 [p = 0.001]), independent of cardiovascular risk factors. Compared with traditional risk factors, reclassification (net reclassification index = 31% to 37%, p ≤ 0.0002) was improved by including the ARS or 1 of its genes, ITGA2B. CONCLUSIONS RNA profiles of platelet-specific genes are novel biomarkers for identifying patients who do not respond adequately to aspirin and who are at risk for death or MI.
Collapse
|