1
|
Woodworth CF, Yee RC, Harris S, Young PM, Araoz PA, Collins JD. Coronary Artery Vasculitis and Encasement: Multimodality Imaging Findings and Mimics. Radiographics 2024; 44:e240009. [PMID: 39388372 DOI: 10.1148/rg.240009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Coronary artery vasculitis (CAV) and coronary artery encasement are rarely diagnosed conditions that are important diagnostic considerations, particularly in patients with acute coronary syndrome without traditional cardiovascular risk factors or systemic illness. Vasculitis refers to inflammation of the blood vessel walls, which can be primary or secondary. This process should be distinguished from neoplastic involvement of the coronary arteries, termed coronary artery encasement. Prospective diagnosis of these diseases is challenging, often requiring multidisciplinary workup with careful attention to clinical presentation and multiorgan findings. While CAV and coronary artery encasement can be indistinguishable at coronary CT angiography, certain imaging features help order the differential diagnosis. CAV should be considered when there is smooth wall thickening that is circumferential and/or continuous. A diagnosis of coronary artery encasement is favored when there is irregular or nodular wall thickening that is eccentric to the vessel lumen. Epicardial fat stranding may also appear more extensive compared with CAV. Potential mimics of CAV include atherosclerosis, acute plaque rupture, coronary artery aneurysm, and spontaneous coronary artery dissection. Detection and diagnosis of CAV may help avoid complications related to accelerated atherosclerosis and infarction. Radiologists should be familiar with the range of pathologic conditions that can affect the coronary arteries beyond atherosclerosis as they may be the first to raise such diagnostic possibilities, guiding next steps in patient workup and management. ©RSNA, 2024 Supplemental material is available for this article.
Collapse
Affiliation(s)
- Claire F Woodworth
- From the Department of Radiology, Memorial University of Newfoundland Faculty of Medicine, Health Sciences Centre, 300 Prince Philip Dr, St. John's, NL, Canada A1B 3V6 (C.F.W., R.C.Y., S.H.); and Department of Radiology, Mayo Clinic, Rochester, Minn (P.M.Y., P.A.A., J.D.C.)
| | - Ryan C Yee
- From the Department of Radiology, Memorial University of Newfoundland Faculty of Medicine, Health Sciences Centre, 300 Prince Philip Dr, St. John's, NL, Canada A1B 3V6 (C.F.W., R.C.Y., S.H.); and Department of Radiology, Mayo Clinic, Rochester, Minn (P.M.Y., P.A.A., J.D.C.)
| | - Scott Harris
- From the Department of Radiology, Memorial University of Newfoundland Faculty of Medicine, Health Sciences Centre, 300 Prince Philip Dr, St. John's, NL, Canada A1B 3V6 (C.F.W., R.C.Y., S.H.); and Department of Radiology, Mayo Clinic, Rochester, Minn (P.M.Y., P.A.A., J.D.C.)
| | - Phillip M Young
- From the Department of Radiology, Memorial University of Newfoundland Faculty of Medicine, Health Sciences Centre, 300 Prince Philip Dr, St. John's, NL, Canada A1B 3V6 (C.F.W., R.C.Y., S.H.); and Department of Radiology, Mayo Clinic, Rochester, Minn (P.M.Y., P.A.A., J.D.C.)
| | - Philip A Araoz
- From the Department of Radiology, Memorial University of Newfoundland Faculty of Medicine, Health Sciences Centre, 300 Prince Philip Dr, St. John's, NL, Canada A1B 3V6 (C.F.W., R.C.Y., S.H.); and Department of Radiology, Mayo Clinic, Rochester, Minn (P.M.Y., P.A.A., J.D.C.)
| | - Jeremy D Collins
- From the Department of Radiology, Memorial University of Newfoundland Faculty of Medicine, Health Sciences Centre, 300 Prince Philip Dr, St. John's, NL, Canada A1B 3V6 (C.F.W., R.C.Y., S.H.); and Department of Radiology, Mayo Clinic, Rochester, Minn (P.M.Y., P.A.A., J.D.C.)
| |
Collapse
|
2
|
García-Abellán J, García JA, Padilla S, Fernández-González M, Agulló V, Mascarell P, Botella Á, Gutiérrez F, Masiá M. No accelerated progression of subclinical atherosclerosis with integrase strand transfer inhibitors compared to non-nucleoside reverse transcriptase inhibitors. J Antimicrob Chemother 2024:dkae383. [PMID: 39450853 DOI: 10.1093/jac/dkae383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND The role of integrase strand transfer inhibitors (INSTI) in the cardiovascular risk of people with HIV is controversial. OBJECTIVES To assess the association of INSTI to subclinical atherosclerosis progression measured with the carotid intima-media thickness (cIMT). METHODS Prospective study in virologically suppressed people with HIV receiving INSTI- or NNRTI-based regimens. cIMT was measured at baseline, 48 and 96 weeks. cIMT progression was analysed both as a continuous and categorical variable, defined as cIMT increase ≥ 10% and/or new carotid plaque. Adjustments through Cox proportional hazard regression and linear mixed models, and propensity score matching were conducted. RESULTS 190 participants were recruited and 173 completed the 96 week follow-up. 107 (56.3%) were receiving an INSTI-containing, 128 (67.4%) a NNRTI-containing and 45 (23.7%) a NNRTI plus an INSTI-containing regimen. The overall median (IQR) 2-year change of cIMT was 0.029 (-0.041 to 0.124) mm; 87 (45.8%) participants experienced a cIMT increase ≥ 10%, of whom 54 (28.4%) developed a new carotid plaque. Adjusted Cox regression showed no differences between INSTI and NNRTI groups in the categorical 2-year progression of cIMT, both including or excluding participants receiving INSTI + NNRTI. Similar results were observed for the continuous cIMT increase through adjusted linear mixed models. Propensity score matching showed no significant differences in the 2 year cIMT change between treatment groups [0.049 mm (-0.031-0.103) in the INSTI group versus 0.047 mm (-0.023-0.115) in the NNRTI group; P = 0.647]. cIMT progression was associated with traditional cardiovascular risk factors. CONCLUSIONS INSTI-based regimens are not associated with increased progression of subclinical atherosclerosis when compared to NNRTI.
Collapse
Affiliation(s)
- Javier García-Abellán
- Infectious Diseases Unit, Hospital General Universitario de Elche and Universidad Miguel Hernández de Elche, Alicante, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - José A García
- Infectious Diseases Unit, Hospital General Universitario de Elche and Universidad Miguel Hernández de Elche, Alicante, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Sergio Padilla
- Infectious Diseases Unit, Hospital General Universitario de Elche and Universidad Miguel Hernández de Elche, Alicante, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Fernández-González
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicante, Spain
| | - Vanesa Agulló
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicante, Spain
| | - Paula Mascarell
- Infectious Diseases Unit, Hospital General Universitario de Elche and Universidad Miguel Hernández de Elche, Alicante, Spain
| | - Ángela Botella
- Infectious Diseases Unit, Hospital General Universitario de Elche, Alicante, Spain
| | - Félix Gutiérrez
- Infectious Diseases Unit, Hospital General Universitario de Elche and Universidad Miguel Hernández de Elche, Alicante, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Mar Masiá
- Infectious Diseases Unit, Hospital General Universitario de Elche and Universidad Miguel Hernández de Elche, Alicante, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
3
|
Bilgiç Z, Kayaaslan B, Köksal M, Kaya-Kalem A, Eser F, Hasanoğlu İ, Güner R. The Relationship of Carotid and Brachial Artery Wall Thickness Measurement with Cardiovascular Risk Scoring in Individuals Living with HIV. INFECTIOUS DISEASES & CLINICAL MICROBIOLOGY 2024; 6:164-173. [PMID: 39399750 PMCID: PMC11465432 DOI: 10.36519/idcm.2024.323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 07/04/2024] [Indexed: 10/15/2024]
Abstract
Objective This study aimed to investigate whether human immunodeficiency virus (HIV) infection affects carotid and brachial artery wall thickness and whether measurement of this thickness contributes to traditional cardiovascular risk scoring in individuals living with HIV. Materials and Methods The patient group included people living with HIV who were followed up in the infectious disease clinic, and the control group included patients without HIV. In both groups, carotid artery intima-media thickness (cIMT) was measured with B-mode ultrasonography (B-USG). cIMT 0.9 mm and above was considered subclinical atherosclerosis. Results The patient group consisted of 66, and the control group consisted of 40 participants. The median cIMT of the patient and control groups was 0.92 (0.45-1.45) mm and 0.55 (0.35-1.25) mm, respectively (p<0.001). Brachial artery IMT was significantly higher in the patient group with 0.45 (0.30-0.76) mm, while it was 0.35 (0.17-0.50) mm in the control group (p<0.001). Although the difference between the cIMT and brachial artery IMT results of the patient and control groups was significant between 18-59 years of age, this difference disappeared in older ages. In the patient group, subclinical atherosclerosis was detected with cIMT in individuals under 30 years of age for whom FRS (Framingham risk score) could not be calculated and in low-risk groups according to FRS (20% and 62.9%, respectively). Conclusion cIMT and brachial artery IMT were found to be significantly higher in people living with HIV. The cIMT measurement seems to be very useful in calculating the CVD risk in individuals living with HIV, especially at young ages, in catching patients who are overlooked by traditional scoring systems.
Collapse
Affiliation(s)
- Zeynep Bilgiç
- Department of Infectious Diseases and Clinical Microbiology, Ankara Yıldırım Beyazıt University Ankara City Hospital, Ankara, Türkiye
| | - Bircan Kayaaslan
- Department of Infectious Diseases and Clinical Microbiology, Ankara Yıldırım Beyazıt University Ankara City Hospital, Ankara, Türkiye
| | | | - Ayşe Kaya-Kalem
- Department of Infectious Diseases and Clinical Microbiology, Ankara Yıldırım Beyazıt University Ankara City Hospital, Ankara, Türkiye
| | - Fatma Eser
- Department of Infectious Diseases and Clinical Microbiology, Ankara Yıldırım Beyazıt University Ankara City Hospital, Ankara, Türkiye
| | - İmran Hasanoğlu
- Department of Infectious Diseases and Clinical Microbiology, Ankara Yıldırım Beyazıt University Ankara City Hospital, Ankara, Türkiye
| | - Rahmet Güner
- Department of Infectious Diseases and Clinical Microbiology, Ankara Yıldırım Beyazıt University Ankara City Hospital, Ankara, Türkiye
| |
Collapse
|
4
|
Karady J, Lu MT, Bergström G, Mayrhofer T, Taron J, Foldyna B, Paradis K, McCallum S, Aberg JA, Currier JS, Fitch KV, Fulda ES, Bloomfield GS, Overton ET, Lind L, Östgren CJ, Elvstam O, Söderberg S, Jernberg T, Pepe R, Dubé MP, Mushatt D, Fichtenbaum CJ, Malvestutto C, Zanni MV, Hoffmann U, Ribaudo H, Grinspoon SK, Douglas PS. Coronary Plaque in People With HIV vs Non-HIV Asymptomatic Community and Symptomatic Higher-Risk Populations. JACC. ADVANCES 2024; 3:100968. [PMID: 38938873 PMCID: PMC11198107 DOI: 10.1016/j.jacadv.2024.100968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/27/2024] [Accepted: 03/13/2024] [Indexed: 06/29/2024]
Abstract
Background People with HIV (PWH) have a high burden of coronary plaques; however, the comparison to people without known HIV (PwoH) needs clarification. Objectives The purpose of this study was to determine coronary plaque burden/phenotype in PWH vs PwoH. Methods Nonstatin using participants from 3 contemporary populations without known coronary plaques with coronary CT were compared: the REPRIEVE (Randomized Trial to Prevent Vascular Events in HIV) studying PWH without cardiovascular symptoms at low-to-moderate risk (n = 755); the SCAPIS (Swedish Cardiopulmonary Bioimage Study) of asymptomatic community PwoH at low-to-intermediate cardiovascular risk (n = 23,558); and the PROMISE (Prospective Multicenter Imaging Study for Evaluation of Chest Pain) of stable chest pain PwoH (n = 2,291). The coronary plaque prevalence on coronary CT was compared, and comparisons were stratified by 10-year atherosclerotic cardiovascular disease (ASCVD) risk, age, and coronary artery calcium (CAC) presence. Results Compared to SCAPIS and PROMISE PwoH, REPRIEVE PWH were younger (50.8 ± 5.8 vs 57.3 ± 4.3 and 60.0 ± 8.0 years; P < 0.001) and had lower ASCVD risk (5.0% ± 3.2% vs 6.0% ± 5.3% and 13.5% ± 11.0%; P < 0.001). More PWH had plaque compared to the asymptomatic cohort (48.5% vs 40.3%; P < 0.001). When stratified by ASCVD risk, PWH had more plaque compared to SCAPIS and a similar prevalence of plaque compared to PROMISE. CAC = 0 was more prevalent in PWH (REPRIEVE 65.2%; SCAPIS 61.6%; PROMISE 49.6%); among CAC = 0, plaque was more prevalent in PWH compared to the PwoH cohorts (REPRIEVE 20.8%; SCAPIS 5.4%; PROMISE 12.3%, P < 0.001). Conclusions Asymptomatic PWH in REPRIEVE had more plaque than asymptomatic PwoH in SCAPIS but had similar prevalence to a higher-risk stable chest pain cohort in PROMISE. In PWH, CAC = 0 does not reliably exclude plaque.
Collapse
Affiliation(s)
- Julia Karady
- Cardiovascular Imaging Research Center, Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts, USA
- Cardiovascular Imaging Research Group, Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Michael T. Lu
- Cardiovascular Imaging Research Center, Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts, USA
| | - Göran Bergström
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Physiology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Thomas Mayrhofer
- Cardiovascular Imaging Research Center, Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts, USA
- School of Business Studies, Stralsund University of Applied Sciences, Stralsund, Germany
| | - Jana Taron
- Cardiovascular Imaging Research Center, Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts, USA
- Faculty of Medicine, Department of Radiology, Medical Center-University of Freiburg, University of Freiburg, Freiburg im Breisgau, Germany
| | - Borek Foldyna
- Cardiovascular Imaging Research Center, Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts, USA
| | - Kayla Paradis
- Cardiovascular Imaging Research Center, Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts, USA
| | - Sara McCallum
- Metabolism Unit, Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts, USA
| | - Judith A. Aberg
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Judith S. Currier
- Department of Medicine, University of California at Los Angeles, Los Angeles, California, USA
| | - Kathleen V. Fitch
- Metabolism Unit, Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts, USA
| | - Evelynne S. Fulda
- Metabolism Unit, Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts, USA
| | - Gerald S. Bloomfield
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Edgar T. Overton
- Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Lars Lind
- Division of Clinical Epidemiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Carl Johan Östgren
- Faculty of Medicine and Health Sciences, Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Center of Medical Image Science and Visualization, Linköping University, Linköping, Sweden
| | - Olof Elvstam
- Department of Translational Medicine, Lund University, Malmö, Sweden
- Department of Infectious Diseases, Växjö Central Hospital, Växjö, Sweden
| | - Stefan Söderberg
- Section of Medicine, Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Tomas Jernberg
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Danderyd Hospital, Karolinska Institute, Stockholm, Sweden
| | - Rosalie Pepe
- Cooper University Hospital, Camden, New Jersey, USA
| | - Michael P. Dubé
- Division of Infectious Diseases, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - David Mushatt
- Section of Infectious Disease, Tulane School of Medicine, New Orleans, Louisiana, USA
| | - Carl J. Fichtenbaum
- Division of Infectious Diseases, University of Cincinnati, Cincinnati, Ohio, USA
| | - Carlos Malvestutto
- Division of Infectious Diseases, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Markella V. Zanni
- Metabolism Unit, Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts, USA
| | - Udo Hoffmann
- Cardiovascular Imaging Research Center, Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts, USA
- Innovative Imaging Consulting LLC, Boston, Massachusetts, USA
| | - Heather Ribaudo
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Steven K. Grinspoon
- Metabolism Unit, Massachusetts General Hospital & Harvard Medical School, Boston, Massachusetts, USA
| | - Pamela S. Douglas
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, USA
| |
Collapse
|
5
|
Prakash P, Swami Vetha BS, Chakraborty R, Wenegieme TY, Masenga SK, Muthian G, Balasubramaniam M, Wanjalla CN, Hinton AO, Kirabo A, Williams CR, Aileru A, Dash C. HIV-Associated Hypertension: Risks, Mechanisms, and Knowledge Gaps. Circ Res 2024; 134:e150-e175. [PMID: 38781298 PMCID: PMC11126208 DOI: 10.1161/circresaha.124.323979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
HIV type 1 (HIV-1) is the causative agent of AIDS. Since the start of the epidemic, HIV/AIDS has been responsible for ≈40 million deaths. Additionally, an estimated 39 million people are currently infected with the virus. HIV-1 primarily infects immune cells, such as CD4+ (cluster of differentiation 4+) T lymphocytes (T cells), and as a consequence, the number of CD4+ T cells progressively declines in people living with HIV. Within a span of ≈10 years, HIV-1 infection leads to the systemic failure of the immune system and progression to AIDS. Fortunately, potent antiviral therapy effectively controls HIV-1 infection and prevents AIDS-related deaths. The efficacy of the current antiviral therapy regimens has transformed the outcome of HIV/AIDS from a death sentence to a chronic disease with a prolonged lifespan of people living with HIV. However, antiviral therapy is not curative, is challenged by virus resistance, can be toxic, and, most importantly, requires lifelong adherence. Furthermore, the improved lifespan has resulted in an increased incidence of non-AIDS-related morbidities in people living with HIV including cardiovascular diseases, renal disease, liver disease, bone disease, cancer, and neurological conditions. In this review, we summarize the current state of knowledge of the cardiovascular comorbidities associated with HIV-1 infection, with a particular focus on hypertension. We also discuss the potential mechanisms known to drive HIV-1-associated hypertension and the knowledge gaps in our understanding of this comorbid condition. Finally, we suggest several directions of future research to better understand the factors, pathways, and mechanisms underlying HIV-1-associated hypertension in the post-antiviral therapy era.
Collapse
Affiliation(s)
- Prem Prakash
- The Center for AIDS Health Disparities Research
- Department of Microbiology, Immunology, and Physiology
- Department of Biochemistry, Cancer Biology, Pharmacology and Neuroscience Meharry Medical College, Nashville, Tennessee, 37208, USA
| | - Berwin Singh Swami Vetha
- Department of Foundational Sciences and Research, School of Dental Medicine, East Carolina University, 1851 MacGregor Downs Road, MS 701, Greenville, NC 27834
| | - Rajasree Chakraborty
- The Center for AIDS Health Disparities Research
- Department of Microbiology, Immunology, and Physiology
- Department of Biochemistry, Cancer Biology, Pharmacology and Neuroscience Meharry Medical College, Nashville, Tennessee, 37208, USA
| | - Tara-Yesomi Wenegieme
- Department of Neuroscience, Cell Biology and Physiology; Boonshoft School of Medicine and the College of Science and Mathematics; Wright State University, Dayton, OH 45435, USA
| | - Sepiso K. Masenga
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Kabwe, Central Province, 10101, Zambia
- HAND Research Group, School of Medicine and Health Sciences, Mulungushi University, Kabwe, Central Province, 10101, Zambia
| | - Gladson Muthian
- The Center for AIDS Health Disparities Research
- Department of Microbiology, Immunology, and Physiology
- Department of Biochemistry, Cancer Biology, Pharmacology and Neuroscience Meharry Medical College, Nashville, Tennessee, 37208, USA
| | - Muthukumar Balasubramaniam
- The Center for AIDS Health Disparities Research
- Department of Microbiology, Immunology, and Physiology
- Department of Biochemistry, Cancer Biology, Pharmacology and Neuroscience Meharry Medical College, Nashville, Tennessee, 37208, USA
| | | | - Antentor O Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine
- Vanderbilt Center for Immunobiology
- Vanderbilt Institute for Infection, Immunology and Inflammation
- Vanderbilt Institute for Global Health, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Clintoria R. Williams
- Department of Neuroscience, Cell Biology and Physiology; Boonshoft School of Medicine and the College of Science and Mathematics; Wright State University, Dayton, OH 45435, USA
| | - Azeez Aileru
- Department of Foundational Sciences and Research, School of Dental Medicine, East Carolina University, 1851 MacGregor Downs Road, MS 701, Greenville, NC 27834
| | - Chandravanu Dash
- The Center for AIDS Health Disparities Research
- Department of Microbiology, Immunology, and Physiology
- Department of Biochemistry, Cancer Biology, Pharmacology and Neuroscience Meharry Medical College, Nashville, Tennessee, 37208, USA
| |
Collapse
|
6
|
Lu MT, Ribaudo H, Foldyna B, Zanni MV, Mayrhofer T, Karady J, Taron J, Fitch KV, McCallum S, Burdo TH, Paradis K, Hedgire SS, Meyersohn NM, DeFilippi C, Malvestutto CD, Sturniolo A, Diggs M, Siminski S, Bloomfield GS, Alston-Smith B, Desvigne-Nickens P, Overton ET, Currier JS, Aberg JA, Fichtenbaum CJ, Hoffmann U, Douglas PS, Grinspoon SK. Effects of Pitavastatin on Coronary Artery Disease and Inflammatory Biomarkers in HIV: Mechanistic Substudy of the REPRIEVE Randomized Clinical Trial. JAMA Cardiol 2024; 9:323-334. [PMID: 38381407 PMCID: PMC10882511 DOI: 10.1001/jamacardio.2023.5661] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/15/2023] [Indexed: 02/22/2024]
Abstract
Importance Cardiovascular disease (CVD) is increased in people with HIV (PWH) and is characterized by premature noncalcified coronary plaque. In the Randomized Trial to Prevent Vascular Events in HIV (REPRIEVE), pitavastatin reduced major adverse cardiovascular events (MACE) by 35% over a median of 5.1 years. Objective To investigate the effects of pitavastatin on noncalcified coronary artery plaque by coronary computed tomography angiography (CTA) and on inflammatory biomarkers as potential mechanisms for MACE prevention. Design, Setting, and Participants This double-blind, placebo-controlled randomized clinical trial enrolled participants from April 2015 to February 2018 at 31 US clinical research sites. PWH without known CVD who were taking antiretroviral therapy and had low to moderate 10-year CVD risk were included. Data were analyzed from April to November 2023. Intervention Oral pitavastatin calcium, 4 mg per day. Main Outcomes and Measures Coronary CTA and inflammatory biomarkers at baseline and 24 months. The primary outcomes were change in noncalcified coronary plaque volume and progression of noncalcified plaque. Results Of 804 enrolled persons, 774 had at least 1 evaluable CTA. Plaque changes were assessed in 611 who completed both CT scans. Of 611 analyzed participants, 513 (84.0%) were male, the mean (SD) age was 51 (6) years, and the median (IQR) 10-year CVD risk was 4.5% (2.6-7.0). A total of 302 were included in the pitavastatin arm and 309 in the placebo arm. The mean noncalcified plaque volume decreased with pitavastatin compared with placebo (mean [SD] change, -1.7 [25.2] mm3 vs 2.6 [27.1] mm3; baseline adjusted difference, -4.3 mm3; 95% CI, -8.6 to -0.1; P = .04; 7% [95% CI, 1-12] greater reduction relative to placebo). A larger effect size was seen among the subgroup with plaque at baseline (-8.8 mm3 [95% CI, -17.9 to 0.4]). Progression of noncalcified plaque was 33% less likely with pitavastatin compared with placebo (relative risk, 0.67; 95% CI, 0.52-0.88; P = .003). Compared with placebo, the mean low-density lipoprotein cholesterol decreased with pitavastatin (mean change: pitavastatin, -28.5 mg/dL; 95% CI, -31.9 to -25.1; placebo, -0.8; 95% CI, -3.8 to 2.2). The pitavastatin arm had a reduction in both oxidized low-density lipoprotein (-29% [95% CI, -32 to -26] vs -13% [95% CI, -17 to -9]; P < .001) and lipoprotein-associated phospholipase A2 (-7% [95% CI, -11 to -4] vs 14% [95% CI, 10-18]; P < .001) compared with placebo at 24 months. Conclusions and Relevance In PWH at low to moderate CVD risk, 24 months of pitavastatin reduced noncalcified plaque volume and progression as well as markers of lipid oxidation and arterial inflammation. These changes may contribute to the observed MACE reduction in REPRIEVE. Trial Registration ClinicalTrials.gov Identifier: NCT02344290.
Collapse
Affiliation(s)
- Michael T. Lu
- Cardiovascular Imaging Research Center, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Heather Ribaudo
- Center for Biostatistics in AIDS Research, Harvard T. H. Chan School of Public Health, Boston, Massachusetts
| | - Borek Foldyna
- Cardiovascular Imaging Research Center, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Markella V. Zanni
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Thomas Mayrhofer
- Cardiovascular Imaging Research Center, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
- School of Business Studies, Stralsund University of Applied Sciences, Stralsund, Germany
| | - Julia Karady
- Cardiovascular Imaging Research Center, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
- Cardiovascular Imaging Research Group, Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Jana Taron
- Cardiovascular Imaging Research Center, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
- Department of Radiology, Medical Center University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kathleen V. Fitch
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Sara McCallum
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Tricia H. Burdo
- Department of Microbiology, Immunology, and Inflammation, Center for NeuroVirology and Gene Editing, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania
| | - Kayla Paradis
- Cardiovascular Imaging Research Center, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Sandeep S. Hedgire
- Cardiovascular Imaging Research Center, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Nandini M. Meyersohn
- Cardiovascular Imaging Research Center, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
| | | | | | - Audra Sturniolo
- Cardiovascular Imaging Research Center, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston
| | - Marissa Diggs
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston
| | | | - Gerald S. Bloomfield
- Department of Medicine, Duke Global Health Institute, Duke Clinical Research Institute, Duke University, Durham, North Carolina
| | - Beverly Alston-Smith
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Patrice Desvigne-Nickens
- Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Edgar T. Overton
- Division of Infectious Diseases, University of Alabama at Birmingham
- ViiV Healthcare, Research Triangle Park, North Carolina
| | - Judith S. Currier
- Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles
| | - Judith A. Aberg
- Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Carl J. Fichtenbaum
- Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | | | - Pamela S. Douglas
- Duke University Research Institute, Duke University School of Medicine, Durham, North Carolina
| | - Steven K. Grinspoon
- Metabolism Unit, Massachusetts General Hospital, Harvard Medical School, Boston
| |
Collapse
|
7
|
Nazari I, Feinstein MJ. Evolving mechanisms and presentations of cardiovascular disease in people with HIV: implications for management. Clin Microbiol Rev 2024; 37:e0009822. [PMID: 38299802 PMCID: PMC10938901 DOI: 10.1128/cmr.00098-22] [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] [Indexed: 02/02/2024] Open
Abstract
People with HIV (PWH) are at elevated risk for cardiovascular diseases (CVDs), including myocardial infarction, heart failure, and sudden cardiac death, among other CVD manifestations. Chronic immune dysregulation resulting in persistent inflammation is common among PWH, particularly those with sustained viremia and impaired CD4+ T cell recovery. This inflammatory milieu is a major contributor to CVDs among PWH, in concert with common comorbidities (such as dyslipidemia and smoking) and, to a lesser extent, off-target effects of antiretroviral therapy. In this review, we discuss the clinical and mechanistic evidence surrounding heightened CVD risks among PWH, implications for specific CVD manifestations, and practical guidance for management in the setting of evolving data.
Collapse
Affiliation(s)
- Ilana Nazari
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Matthew J. Feinstein
- Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Division of Cardiology in the Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
8
|
Suleman M, Khan SU, Hussain T, Khan MU, Shamsul Hassan S, Majid M, Khan SU, Shehzad Khan M, Shan Ahmad RU, Arif M, Ahmad Z, Crovella S, Anthony S. Cardiovascular challenges in the era of antiretroviral therapy for AIDS/ HIV: A comprehensive review of research advancements, pathophysiological insights, and future directions. Curr Probl Cardiol 2024; 49:102353. [PMID: 38128638 DOI: 10.1016/j.cpcardiol.2023.102353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023]
Abstract
Cardiovascular disease, particularly coronary heart disease, is becoming more common among those living with HIV. Individuals with HIV face an increased susceptibility to myocardial infarction, also known as a heart attack, as compared to the general population in developed countries. This heightened risk can be attributed mainly to the presence of effective antiretroviral drugs and the resulting longer lifespan. Some cardiac issues linked to non-antiretroviral medications, including myocarditis, endocarditis, cardiomyopathy with dilation, pulmonary hypertension, and oedema of the heart, may affect those not undergoing highly active antiretroviral therapy (ART). Impaired immune function and systemic inflammation are significant contributors to this phenomenon after initiating highly aggressive antiretroviral treatment ART. It is becoming more challenging to determine the best course of treatment for HIV-associated cardiomyopathy due to new research suggesting that protease inhibitors might have a negative impact on the development of HF. Currently, the primary focus of research on ART medications is centered on the cardiovascular adverse effects of nucleoside reverse transcriptase inhibitors and protease inhibitors. This review paper thoroughly evaluates the advancements achieved in cardiovascular disease research and explores the potential implications for prospects. Additionally, it considers the field's future prospects while examining how ART might be altered and its clinical applications.
Collapse
Affiliation(s)
- Muhammad Suleman
- Laboratory of Animal Research Center (LARC), Qatar University, Doha, Qatar; Center for Biotechnology and Microbiology, University of Swat, Swat, Pakistan
| | - Shahid Ullah Khan
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City and Southwest University, College of Agronomy and Biotechnology, Southwest University, Chongqing 400715, PR China; Department of Biochemistry, Women Medical and Dental College, Khyber Medical University, Abbottabad, Khyber Pakhtunkhwa 22080, Pakistan
| | - Talib Hussain
- Women Dental College Abbottabad, KPK 22020, Pakistan
| | - Munir Ullah Khan
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, International Research Center for X Polymers, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027 PR China
| | - Syed Shamsul Hassan
- Chinese Academy of Sciences, Institute of Basic Medicine and Cancer (IBMC),Hangzhou 310002, PR China
| | - Muhammad Majid
- Faculty of Pharmacy, Hamdard University, Islamabad 45550, Pakistan
| | - Safir Ullah Khan
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei 230027, PR China
| | - Muhammad Shehzad Khan
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering (COCHE), Shatin city, HKSAR, Hong Kong
| | - Rafi U Shan Ahmad
- Department of Biomedical Engineering, City university of Hong Kong, Kowloon City, HKSAR, Hong Kong
| | - Muhammad Arif
- College of Agriculture, Guizhou University, Guiyang, Guizhou, China
| | - Zubair Ahmad
- Applied College, Center of Bee Research and its Products, Unit of Bee Research and Honey Production, and Research Center for Advanced Materials Science (RCAMS), King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia
| | - Sergio Crovella
- Center for Biotechnology and Microbiology, University of Swat, Swat, Pakistan
| | - Stefan Anthony
- Chinese Academy of Sciences, Institute of Basic Medicine and Cancer (IBMC),Hangzhou 310002, PR China.
| |
Collapse
|
9
|
Foldyna B, Mayrhofer T, Zanni MV, Lyass A, Barve R, Karady J, McCallum S, Burdo TH, Fitch KV, Paradis K, Fulda ES, Diggs MR, Bloomfield GS, Malvestutto CD, Fichtenbaum CJ, Aberg JA, Currier JS, Ribaudo HJ, Hoffmann U, Lu MT, Douglas PS, Grinspoon SK. Pericoronary Adipose Tissue Density, Inflammation, and Subclinical Coronary Artery Disease Among People With HIV in the REPRIEVE Cohort. Clin Infect Dis 2023; 77:1676-1686. [PMID: 37439633 PMCID: PMC10724469 DOI: 10.1093/cid/ciad419] [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: 04/06/2023] [Revised: 06/30/2023] [Accepted: 07/10/2023] [Indexed: 07/14/2023] Open
Abstract
BACKGROUND Pericoronary adipose tissue (PCAT) may influence plaque development through inflammatory mechanisms. We assessed PCAT density, as a measure of pericoronary inflammation, in relationship to coronary plaque among people with human immunodeficiency virus (HIV [PWH]) and to a matched control population. METHODS In this baseline analysis of 727 participants of the Randomized Trial to Prevent Vascular Events in HIV (REPRIEVE) Mechanistic Substudy, we related computed tomography-derived PCAT density to presence and extent (Leaman score) of coronary artery disease (CAD), noncalcified plaque, coronary artery calcium (CAC), and vulnerable plaque features using multivariable logistic regression analyses. We further compared the PCAT density between PWH and age, sex, body mass index, CAC score, and statin use-matched controls from the community-based Framingham Heart Study (N = 464), adjusting for relevant clinical covariates. RESULTS Among 727 REPRIEVE participants (age 50.8 ± 5.8 years; 83.6% [608/727] male), PCAT density was higher in those with (vs without) coronary plaque, noncalcified plaque, CAC >0, vulnerable plaque, and high CAD burden (Leaman score >5) (P < .001 for each comparison). PCAT density related to prevalent coronary plaque (adjusted odds ratio [per 10 HU]: 1.44; 95% confidence interval, 1.22-1.70; P < .001), adjusted for clinical cardiovascular risk factors, body mass index, and systemic immune/inflammatory biomarkers. Similarly, PCAT density related to CAC >0, noncalcified plaque, vulnerable plaque, and Leaman score >5 (all P ≤ .002). PCAT density was greater among REPRIEVE participants versus Framingham Heart Study (-88.2 ± 0.5 HU versus -90.6 ± 0.4 HU; P < .001). CONCLUSIONS Among PWH in REPRIEVE, a large primary cardiovascular disease prevention cohort, increased PCAT density independently associated with prevalence and severity of coronary plaque, linking increased coronary inflammation to CAD in PWH.
Collapse
Affiliation(s)
- Borek Foldyna
- Department of Radiology, Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas Mayrhofer
- Department of Radiology, Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Health Economics, School of Business Studies, Stralsund University of Applied Sciences, Stralsund, Germany
| | - Markella V Zanni
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Asya Lyass
- Department of Mathematics and Statistics, Boston University, Boston, Massachusetts, USA
| | - Radhika Barve
- Department of Radiology, Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Julia Karady
- Department of Radiology, Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Sara McCallum
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Tricia H Burdo
- Department of Microbiology, Immunology, and Inflammation and Center for NeuroVirology and Gene Editing, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Kathleen V Fitch
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Kayla Paradis
- Department of Radiology, Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Evelynne S Fulda
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Marissa R Diggs
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Gerald S Bloomfield
- Department of Medicine, Duke Global Health Institute and Duke Clinical Research Institute, Duke University, Durham, North Carolina, USA
| | - Carlos D Malvestutto
- Division of Infectious Diseases, Ohio State University Medical Center, Columbus, Ohio, USA
| | - Carl J Fichtenbaum
- Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Judith A Aberg
- Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Judith S Currier
- Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Heather J Ribaudo
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Udo Hoffmann
- Innovative Imaging Consulting LLC, Waltham, Massachusetts, USA
| | - Michael T Lu
- Department of Radiology, Cardiovascular Imaging Research Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Pamela S Douglas
- Department of Medicine (Cardiology), Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Steven K Grinspoon
- Metabolism Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
10
|
Awamura T, Nakasone ES, Gangcuangco LM, Subia NT, Bali AJ, Chow DC, Shikuma CM, Park J. Platelet and HIV Interactions and Their Contribution to Non-AIDS Comorbidities. Biomolecules 2023; 13:1608. [PMID: 38002289 PMCID: PMC10669125 DOI: 10.3390/biom13111608] [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: 10/05/2023] [Revised: 10/29/2023] [Accepted: 10/31/2023] [Indexed: 11/26/2023] Open
Abstract
Platelets are anucleate cytoplasmic cell fragments that circulate in the blood, where they are involved in regulating hemostasis. Beyond their normal physiologic role, platelets have emerged as versatile effectors of immune response. During an infection, cell surface receptors enable platelets to recognize viruses, resulting in their activation. Activated platelets release biologically active molecules that further trigger host immune responses to protect the body against infection. Their impact on the immune response is also associated with the recruitment of circulating leukocytes to the site of infection. They can also aggregate with leukocytes, including lymphocytes, monocytes, and neutrophils, to immobilize pathogens and prevent viral dissemination. Despite their host protective role, platelets have also been shown to be associated with various pathophysiological processes. In this review, we will summarize platelet and HIV interactions during infection. We will also highlight and discuss platelet and platelet-derived mediators, how they interact with immune cells, and the multifaceted responsibilities of platelets in HIV infection. Furthermore, we will give an overview of non-AIDS comorbidities linked to platelet dysfunction and the impact of antiretroviral therapy on platelet function.
Collapse
Affiliation(s)
- Thomas Awamura
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (T.A.); (N.T.S.); (A.-J.B.)
| | - Elizabeth S. Nakasone
- University of Hawai‘i Cancer Center, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA;
- Department of Medicine, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA;
| | - Louie Mar Gangcuangco
- Hawai‘i Center for AIDS, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (L.M.G.); (C.M.S.)
| | - Natalie T. Subia
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (T.A.); (N.T.S.); (A.-J.B.)
| | - Aeron-Justin Bali
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (T.A.); (N.T.S.); (A.-J.B.)
| | - Dominic C. Chow
- Department of Medicine, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA;
- Hawai‘i Center for AIDS, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (L.M.G.); (C.M.S.)
| | - Cecilia M. Shikuma
- Hawai‘i Center for AIDS, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (L.M.G.); (C.M.S.)
| | - Juwon Park
- Department of Tropical Medicine, Medical Microbiology, and Pharmacology, John A. Burns School Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (T.A.); (N.T.S.); (A.-J.B.)
- Hawai‘i Center for AIDS, John A. Burns School of Medicine, University of Hawai‘i at Mānoa, Honolulu, HI 96813, USA; (L.M.G.); (C.M.S.)
| |
Collapse
|
11
|
Avagimyan A, Pogosova N, Kakturskiy L, Sheibani M, Urazova O, Trofimenko A, Navarsdyan G, Jndoyan Z, Abgaryan K, Fogacci F, Galli M, Agati L, Kobalava Z, Shafie D, Marzilli M, Gogiashvili L, Sarrafzadegan N. HIV-Related Atherosclerosis: State-of-the-Art-Review. Curr Probl Cardiol 2023; 48:101783. [PMID: 37172874 DOI: 10.1016/j.cpcardiol.2023.101783] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 05/07/2023] [Indexed: 05/15/2023]
Abstract
The infection caused by the Human Immunodeficiency Virus (HIV) has spread rapidly across the globe, assuming the characteristics of an epidemic in some regions. Thanks to the introduction of antiretroviral therapy into routine clinical practice, there was a considerable breakthrough in the treatment of HIV, that is now HIV is potentially well-controlled even in low-income countries. To date, HIV infection has moved from the group of life-threatening conditions to the group of chronic and well controlled ones and the quality of life and life expectancy of HIV+ people, with an undetectable viral load is closer to that of an HIV- people. However, unsolved issues still persist. For example: people living with HIV are more prone to the age-related diseases, especially atherosclerosis. For this reason, a better understanding of the mechanisms of HIV-associated destabilization of vascular homeostasis seems to be an urgent duty, that may lead to the development of new protocols, bringing the possibilities of pathogenetic therapies to a new level. The purpose of the article was to evaluate the pathological aspects of HIV-induced atherosclerosis.
Collapse
Affiliation(s)
- Ashot Avagimyan
- Assistant Professor, Anatomical Pathology and Clinical Morphology Department, Yerevan State Medical University after M. Heratsi, Yerevan, Armenia.
| | - Nana Pogosova
- Professor, Deputy of General Director for Science and Preventive Cardiology, National Medical Research Centre of Cardiology after E. Chazov, Moscow, Russia
| | - Lev Kakturskiy
- Professor, Scientific Director, Research Institute of Human Morphology FSBI «Petrovskiy NRCS, Moscow, Russia
| | - Mohammad Sheibani
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Razi Drug Research Centre, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Olga Urazova
- Professor, Head of Pathophysiology Department, Siberian State Medical University, Tomsk, Russia
| | - Artem Trofimenko
- Associate Professor, Pathophysiology Department, Kuban State Medical University, Krasnodar, Russia
| | - Grizelda Navarsdyan
- Professor, Pathophysiology Department, Yerevan State Medical University after M. Heratsi, Yerevan, Armenia
| | - Zinaida Jndoyan
- Professor, Head of Internal Diseases Propedeutics Department, Yerevan State Medical University after M. Heratsi, Armenia
| | - Kristina Abgaryan
- Associate Professor, Medical Microbiology Department, Yerevan State Medical University after M.Heratsi, Armenia
| | - Federica Fogacci
- Research Fellow, Atherosclerosis and Metabolic Disorders Research Unit, University of Bologna, Bologna, Italy
| | - Mattia Galli
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
| | - Luciano Agati
- Professor of Cardiology Department, Head of Cardiology Unit Azienda Policlinico Umberto II, Sapienza University, Rome, Italy
| | - Zhanna Kobalava
- Professor, Head of Internal Disease, Cardiology and Clinical Pharmacology Department, Peoples' Friendship University of Russia (RUDN), Moscow, Russia
| | - Davood Shafie
- Isfahan Cardiovascular Research Institute, Isfahan, Iran
| | - Mario Marzilli
- Professor, Head of Cardiovascular Medicine Division, University of Pisa, Pisa, Italy
| | - Liana Gogiashvili
- Professor, Head of Experimental and Clinical Pathology Department, Al. Natishvili Institute of Experimental Morphology, I. Javakhishvili Tbilisi State University, Tbilisi, Georgia
| | - Nizal Sarrafzadegan
- Professor, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
12
|
Grinspoon SK, Fitch KV, Zanni MV, Fichtenbaum CJ, Umbleja T, Aberg JA, Overton ET, Malvestutto CD, Bloomfield GS, Currier JS, Martinez E, Roa JC, Diggs MR, Fulda ES, Paradis K, Wiviott SD, Foldyna B, Looby SE, Desvigne-Nickens P, Alston-Smith B, Leon-Cruz J, McCallum S, Hoffmann U, Lu MT, Ribaudo HJ, Douglas PS. Pitavastatin to Prevent Cardiovascular Disease in HIV Infection. N Engl J Med 2023; 389:687-699. [PMID: 37486775 PMCID: PMC10564556 DOI: 10.1056/nejmoa2304146] [Citation(s) in RCA: 134] [Impact Index Per Article: 134.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
BACKGROUND The risk of cardiovascular disease is increased among persons with human immunodeficiency virus (HIV) infection, so data regarding primary prevention strategies in this population are needed. METHODS In this phase 3 trial, we randomly assigned 7769 participants with HIV infection with a low-to-moderate risk of cardiovascular disease who were receiving antiretroviral therapy to receive daily pitavastatin calcium (at a dose of 4 mg) or placebo. The primary outcome was the occurrence of a major adverse cardiovascular event, which was defined as a composite of cardiovascular death, myocardial infarction, hospitalization for unstable angina, stroke, transient ischemic attack, peripheral arterial ischemia, revascularization, or death from an undetermined cause. RESULTS The median age of the participants was 50 years (interquartile range, 45 to 55); the median CD4 count was 621 cells per cubic millimeter (interquartile range, 448 to 827), and the HIV RNA value was below quantification in 5250 of 5997 participants (87.5%) with available data. The trial was stopped early for efficacy after a median follow-up of 5.1 years (interquartile range, 4.3 to 5.9). The incidence of a major adverse cardiovascular event was 4.81 per 1000 person-years in the pitavastatin group and 7.32 per 1000 person-years in the placebo group (hazard ratio, 0.65; 95% confidence interval [CI], 0.48 to 0.90; P = 0.002). Muscle-related symptoms occurred in 91 participants (2.3%) in the pitavastatin group and in 53 (1.4%) in the placebo group; diabetes mellitus occurred in 206 participants (5.3%) and in 155 (4.0%), respectively. CONCLUSIONS Participants with HIV infection who received pitavastatin had a lower risk of a major adverse cardiovascular event than those who received placebo over a median follow-up of 5.1 years. (Funded by the National Institutes of Health and others; REPRIEVE ClinicalTrials.gov number, NCT02344290.).
Collapse
Affiliation(s)
- Steven K Grinspoon
- From the Metabolism Unit (S.K.G., K.V.F., M.V.Z., M.R.D., E.S.F., S.E.L., S.M.), the Cardiovascular Imaging Research Center, Department of Radiology (K.P., B.F., M.T.L.), and the Yvonne L. Munn Center for Nursing Research (S.E.L.), Massachusetts General Hospital and Harvard Medical School, the Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health (T.U., J.L.-C., H.J.R.), and the Thrombolysis in Myocardial Infarction Study Group, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School (S.D.W.) - all in Boston; the Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati (C.J.F.), and the Division of Infectious Diseases, Ohio State University Medical Center, Columbus (C.D.M.); the Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York (J.A.A.); the Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham (E.T.O.); the Department of Medicine, Duke Global Health Institute and Duke Clinical Research Institute, Duke University (G.S.B.), and Duke University Research Institute, Duke University School of Medicine (P.S.D.) - both in Durham, NC; the Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles (J.S.C.); the Infectious Diseases Service, Hospital Clinic and University of Barcelona, Barcelona, and CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid - both in Spain (E.M.); DLH, Silver Spring (J.C.R.), and the Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute (P.D.-N.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (B.A.-S.), National Institutes of Health, Bethesda - all in Maryland; and Cleerly, Denver (U.H.)
| | - Kathleen V Fitch
- From the Metabolism Unit (S.K.G., K.V.F., M.V.Z., M.R.D., E.S.F., S.E.L., S.M.), the Cardiovascular Imaging Research Center, Department of Radiology (K.P., B.F., M.T.L.), and the Yvonne L. Munn Center for Nursing Research (S.E.L.), Massachusetts General Hospital and Harvard Medical School, the Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health (T.U., J.L.-C., H.J.R.), and the Thrombolysis in Myocardial Infarction Study Group, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School (S.D.W.) - all in Boston; the Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati (C.J.F.), and the Division of Infectious Diseases, Ohio State University Medical Center, Columbus (C.D.M.); the Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York (J.A.A.); the Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham (E.T.O.); the Department of Medicine, Duke Global Health Institute and Duke Clinical Research Institute, Duke University (G.S.B.), and Duke University Research Institute, Duke University School of Medicine (P.S.D.) - both in Durham, NC; the Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles (J.S.C.); the Infectious Diseases Service, Hospital Clinic and University of Barcelona, Barcelona, and CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid - both in Spain (E.M.); DLH, Silver Spring (J.C.R.), and the Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute (P.D.-N.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (B.A.-S.), National Institutes of Health, Bethesda - all in Maryland; and Cleerly, Denver (U.H.)
| | - Markella V Zanni
- From the Metabolism Unit (S.K.G., K.V.F., M.V.Z., M.R.D., E.S.F., S.E.L., S.M.), the Cardiovascular Imaging Research Center, Department of Radiology (K.P., B.F., M.T.L.), and the Yvonne L. Munn Center for Nursing Research (S.E.L.), Massachusetts General Hospital and Harvard Medical School, the Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health (T.U., J.L.-C., H.J.R.), and the Thrombolysis in Myocardial Infarction Study Group, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School (S.D.W.) - all in Boston; the Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati (C.J.F.), and the Division of Infectious Diseases, Ohio State University Medical Center, Columbus (C.D.M.); the Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York (J.A.A.); the Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham (E.T.O.); the Department of Medicine, Duke Global Health Institute and Duke Clinical Research Institute, Duke University (G.S.B.), and Duke University Research Institute, Duke University School of Medicine (P.S.D.) - both in Durham, NC; the Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles (J.S.C.); the Infectious Diseases Service, Hospital Clinic and University of Barcelona, Barcelona, and CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid - both in Spain (E.M.); DLH, Silver Spring (J.C.R.), and the Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute (P.D.-N.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (B.A.-S.), National Institutes of Health, Bethesda - all in Maryland; and Cleerly, Denver (U.H.)
| | - Carl J Fichtenbaum
- From the Metabolism Unit (S.K.G., K.V.F., M.V.Z., M.R.D., E.S.F., S.E.L., S.M.), the Cardiovascular Imaging Research Center, Department of Radiology (K.P., B.F., M.T.L.), and the Yvonne L. Munn Center for Nursing Research (S.E.L.), Massachusetts General Hospital and Harvard Medical School, the Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health (T.U., J.L.-C., H.J.R.), and the Thrombolysis in Myocardial Infarction Study Group, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School (S.D.W.) - all in Boston; the Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati (C.J.F.), and the Division of Infectious Diseases, Ohio State University Medical Center, Columbus (C.D.M.); the Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York (J.A.A.); the Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham (E.T.O.); the Department of Medicine, Duke Global Health Institute and Duke Clinical Research Institute, Duke University (G.S.B.), and Duke University Research Institute, Duke University School of Medicine (P.S.D.) - both in Durham, NC; the Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles (J.S.C.); the Infectious Diseases Service, Hospital Clinic and University of Barcelona, Barcelona, and CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid - both in Spain (E.M.); DLH, Silver Spring (J.C.R.), and the Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute (P.D.-N.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (B.A.-S.), National Institutes of Health, Bethesda - all in Maryland; and Cleerly, Denver (U.H.)
| | - Triin Umbleja
- From the Metabolism Unit (S.K.G., K.V.F., M.V.Z., M.R.D., E.S.F., S.E.L., S.M.), the Cardiovascular Imaging Research Center, Department of Radiology (K.P., B.F., M.T.L.), and the Yvonne L. Munn Center for Nursing Research (S.E.L.), Massachusetts General Hospital and Harvard Medical School, the Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health (T.U., J.L.-C., H.J.R.), and the Thrombolysis in Myocardial Infarction Study Group, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School (S.D.W.) - all in Boston; the Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati (C.J.F.), and the Division of Infectious Diseases, Ohio State University Medical Center, Columbus (C.D.M.); the Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York (J.A.A.); the Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham (E.T.O.); the Department of Medicine, Duke Global Health Institute and Duke Clinical Research Institute, Duke University (G.S.B.), and Duke University Research Institute, Duke University School of Medicine (P.S.D.) - both in Durham, NC; the Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles (J.S.C.); the Infectious Diseases Service, Hospital Clinic and University of Barcelona, Barcelona, and CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid - both in Spain (E.M.); DLH, Silver Spring (J.C.R.), and the Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute (P.D.-N.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (B.A.-S.), National Institutes of Health, Bethesda - all in Maryland; and Cleerly, Denver (U.H.)
| | - Judith A Aberg
- From the Metabolism Unit (S.K.G., K.V.F., M.V.Z., M.R.D., E.S.F., S.E.L., S.M.), the Cardiovascular Imaging Research Center, Department of Radiology (K.P., B.F., M.T.L.), and the Yvonne L. Munn Center for Nursing Research (S.E.L.), Massachusetts General Hospital and Harvard Medical School, the Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health (T.U., J.L.-C., H.J.R.), and the Thrombolysis in Myocardial Infarction Study Group, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School (S.D.W.) - all in Boston; the Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati (C.J.F.), and the Division of Infectious Diseases, Ohio State University Medical Center, Columbus (C.D.M.); the Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York (J.A.A.); the Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham (E.T.O.); the Department of Medicine, Duke Global Health Institute and Duke Clinical Research Institute, Duke University (G.S.B.), and Duke University Research Institute, Duke University School of Medicine (P.S.D.) - both in Durham, NC; the Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles (J.S.C.); the Infectious Diseases Service, Hospital Clinic and University of Barcelona, Barcelona, and CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid - both in Spain (E.M.); DLH, Silver Spring (J.C.R.), and the Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute (P.D.-N.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (B.A.-S.), National Institutes of Health, Bethesda - all in Maryland; and Cleerly, Denver (U.H.)
| | - Edgar T Overton
- From the Metabolism Unit (S.K.G., K.V.F., M.V.Z., M.R.D., E.S.F., S.E.L., S.M.), the Cardiovascular Imaging Research Center, Department of Radiology (K.P., B.F., M.T.L.), and the Yvonne L. Munn Center for Nursing Research (S.E.L.), Massachusetts General Hospital and Harvard Medical School, the Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health (T.U., J.L.-C., H.J.R.), and the Thrombolysis in Myocardial Infarction Study Group, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School (S.D.W.) - all in Boston; the Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati (C.J.F.), and the Division of Infectious Diseases, Ohio State University Medical Center, Columbus (C.D.M.); the Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York (J.A.A.); the Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham (E.T.O.); the Department of Medicine, Duke Global Health Institute and Duke Clinical Research Institute, Duke University (G.S.B.), and Duke University Research Institute, Duke University School of Medicine (P.S.D.) - both in Durham, NC; the Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles (J.S.C.); the Infectious Diseases Service, Hospital Clinic and University of Barcelona, Barcelona, and CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid - both in Spain (E.M.); DLH, Silver Spring (J.C.R.), and the Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute (P.D.-N.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (B.A.-S.), National Institutes of Health, Bethesda - all in Maryland; and Cleerly, Denver (U.H.)
| | - Carlos D Malvestutto
- From the Metabolism Unit (S.K.G., K.V.F., M.V.Z., M.R.D., E.S.F., S.E.L., S.M.), the Cardiovascular Imaging Research Center, Department of Radiology (K.P., B.F., M.T.L.), and the Yvonne L. Munn Center for Nursing Research (S.E.L.), Massachusetts General Hospital and Harvard Medical School, the Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health (T.U., J.L.-C., H.J.R.), and the Thrombolysis in Myocardial Infarction Study Group, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School (S.D.W.) - all in Boston; the Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati (C.J.F.), and the Division of Infectious Diseases, Ohio State University Medical Center, Columbus (C.D.M.); the Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York (J.A.A.); the Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham (E.T.O.); the Department of Medicine, Duke Global Health Institute and Duke Clinical Research Institute, Duke University (G.S.B.), and Duke University Research Institute, Duke University School of Medicine (P.S.D.) - both in Durham, NC; the Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles (J.S.C.); the Infectious Diseases Service, Hospital Clinic and University of Barcelona, Barcelona, and CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid - both in Spain (E.M.); DLH, Silver Spring (J.C.R.), and the Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute (P.D.-N.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (B.A.-S.), National Institutes of Health, Bethesda - all in Maryland; and Cleerly, Denver (U.H.)
| | - Gerald S Bloomfield
- From the Metabolism Unit (S.K.G., K.V.F., M.V.Z., M.R.D., E.S.F., S.E.L., S.M.), the Cardiovascular Imaging Research Center, Department of Radiology (K.P., B.F., M.T.L.), and the Yvonne L. Munn Center for Nursing Research (S.E.L.), Massachusetts General Hospital and Harvard Medical School, the Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health (T.U., J.L.-C., H.J.R.), and the Thrombolysis in Myocardial Infarction Study Group, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School (S.D.W.) - all in Boston; the Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati (C.J.F.), and the Division of Infectious Diseases, Ohio State University Medical Center, Columbus (C.D.M.); the Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York (J.A.A.); the Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham (E.T.O.); the Department of Medicine, Duke Global Health Institute and Duke Clinical Research Institute, Duke University (G.S.B.), and Duke University Research Institute, Duke University School of Medicine (P.S.D.) - both in Durham, NC; the Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles (J.S.C.); the Infectious Diseases Service, Hospital Clinic and University of Barcelona, Barcelona, and CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid - both in Spain (E.M.); DLH, Silver Spring (J.C.R.), and the Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute (P.D.-N.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (B.A.-S.), National Institutes of Health, Bethesda - all in Maryland; and Cleerly, Denver (U.H.)
| | - Judith S Currier
- From the Metabolism Unit (S.K.G., K.V.F., M.V.Z., M.R.D., E.S.F., S.E.L., S.M.), the Cardiovascular Imaging Research Center, Department of Radiology (K.P., B.F., M.T.L.), and the Yvonne L. Munn Center for Nursing Research (S.E.L.), Massachusetts General Hospital and Harvard Medical School, the Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health (T.U., J.L.-C., H.J.R.), and the Thrombolysis in Myocardial Infarction Study Group, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School (S.D.W.) - all in Boston; the Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati (C.J.F.), and the Division of Infectious Diseases, Ohio State University Medical Center, Columbus (C.D.M.); the Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York (J.A.A.); the Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham (E.T.O.); the Department of Medicine, Duke Global Health Institute and Duke Clinical Research Institute, Duke University (G.S.B.), and Duke University Research Institute, Duke University School of Medicine (P.S.D.) - both in Durham, NC; the Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles (J.S.C.); the Infectious Diseases Service, Hospital Clinic and University of Barcelona, Barcelona, and CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid - both in Spain (E.M.); DLH, Silver Spring (J.C.R.), and the Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute (P.D.-N.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (B.A.-S.), National Institutes of Health, Bethesda - all in Maryland; and Cleerly, Denver (U.H.)
| | - Esteban Martinez
- From the Metabolism Unit (S.K.G., K.V.F., M.V.Z., M.R.D., E.S.F., S.E.L., S.M.), the Cardiovascular Imaging Research Center, Department of Radiology (K.P., B.F., M.T.L.), and the Yvonne L. Munn Center for Nursing Research (S.E.L.), Massachusetts General Hospital and Harvard Medical School, the Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health (T.U., J.L.-C., H.J.R.), and the Thrombolysis in Myocardial Infarction Study Group, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School (S.D.W.) - all in Boston; the Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati (C.J.F.), and the Division of Infectious Diseases, Ohio State University Medical Center, Columbus (C.D.M.); the Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York (J.A.A.); the Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham (E.T.O.); the Department of Medicine, Duke Global Health Institute and Duke Clinical Research Institute, Duke University (G.S.B.), and Duke University Research Institute, Duke University School of Medicine (P.S.D.) - both in Durham, NC; the Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles (J.S.C.); the Infectious Diseases Service, Hospital Clinic and University of Barcelona, Barcelona, and CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid - both in Spain (E.M.); DLH, Silver Spring (J.C.R.), and the Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute (P.D.-N.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (B.A.-S.), National Institutes of Health, Bethesda - all in Maryland; and Cleerly, Denver (U.H.)
| | - Jhoanna C Roa
- From the Metabolism Unit (S.K.G., K.V.F., M.V.Z., M.R.D., E.S.F., S.E.L., S.M.), the Cardiovascular Imaging Research Center, Department of Radiology (K.P., B.F., M.T.L.), and the Yvonne L. Munn Center for Nursing Research (S.E.L.), Massachusetts General Hospital and Harvard Medical School, the Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health (T.U., J.L.-C., H.J.R.), and the Thrombolysis in Myocardial Infarction Study Group, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School (S.D.W.) - all in Boston; the Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati (C.J.F.), and the Division of Infectious Diseases, Ohio State University Medical Center, Columbus (C.D.M.); the Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York (J.A.A.); the Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham (E.T.O.); the Department of Medicine, Duke Global Health Institute and Duke Clinical Research Institute, Duke University (G.S.B.), and Duke University Research Institute, Duke University School of Medicine (P.S.D.) - both in Durham, NC; the Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles (J.S.C.); the Infectious Diseases Service, Hospital Clinic and University of Barcelona, Barcelona, and CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid - both in Spain (E.M.); DLH, Silver Spring (J.C.R.), and the Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute (P.D.-N.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (B.A.-S.), National Institutes of Health, Bethesda - all in Maryland; and Cleerly, Denver (U.H.)
| | - Marissa R Diggs
- From the Metabolism Unit (S.K.G., K.V.F., M.V.Z., M.R.D., E.S.F., S.E.L., S.M.), the Cardiovascular Imaging Research Center, Department of Radiology (K.P., B.F., M.T.L.), and the Yvonne L. Munn Center for Nursing Research (S.E.L.), Massachusetts General Hospital and Harvard Medical School, the Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health (T.U., J.L.-C., H.J.R.), and the Thrombolysis in Myocardial Infarction Study Group, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School (S.D.W.) - all in Boston; the Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati (C.J.F.), and the Division of Infectious Diseases, Ohio State University Medical Center, Columbus (C.D.M.); the Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York (J.A.A.); the Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham (E.T.O.); the Department of Medicine, Duke Global Health Institute and Duke Clinical Research Institute, Duke University (G.S.B.), and Duke University Research Institute, Duke University School of Medicine (P.S.D.) - both in Durham, NC; the Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles (J.S.C.); the Infectious Diseases Service, Hospital Clinic and University of Barcelona, Barcelona, and CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid - both in Spain (E.M.); DLH, Silver Spring (J.C.R.), and the Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute (P.D.-N.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (B.A.-S.), National Institutes of Health, Bethesda - all in Maryland; and Cleerly, Denver (U.H.)
| | - Evelynne S Fulda
- From the Metabolism Unit (S.K.G., K.V.F., M.V.Z., M.R.D., E.S.F., S.E.L., S.M.), the Cardiovascular Imaging Research Center, Department of Radiology (K.P., B.F., M.T.L.), and the Yvonne L. Munn Center for Nursing Research (S.E.L.), Massachusetts General Hospital and Harvard Medical School, the Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health (T.U., J.L.-C., H.J.R.), and the Thrombolysis in Myocardial Infarction Study Group, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School (S.D.W.) - all in Boston; the Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati (C.J.F.), and the Division of Infectious Diseases, Ohio State University Medical Center, Columbus (C.D.M.); the Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York (J.A.A.); the Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham (E.T.O.); the Department of Medicine, Duke Global Health Institute and Duke Clinical Research Institute, Duke University (G.S.B.), and Duke University Research Institute, Duke University School of Medicine (P.S.D.) - both in Durham, NC; the Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles (J.S.C.); the Infectious Diseases Service, Hospital Clinic and University of Barcelona, Barcelona, and CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid - both in Spain (E.M.); DLH, Silver Spring (J.C.R.), and the Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute (P.D.-N.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (B.A.-S.), National Institutes of Health, Bethesda - all in Maryland; and Cleerly, Denver (U.H.)
| | - Kayla Paradis
- From the Metabolism Unit (S.K.G., K.V.F., M.V.Z., M.R.D., E.S.F., S.E.L., S.M.), the Cardiovascular Imaging Research Center, Department of Radiology (K.P., B.F., M.T.L.), and the Yvonne L. Munn Center for Nursing Research (S.E.L.), Massachusetts General Hospital and Harvard Medical School, the Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health (T.U., J.L.-C., H.J.R.), and the Thrombolysis in Myocardial Infarction Study Group, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School (S.D.W.) - all in Boston; the Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati (C.J.F.), and the Division of Infectious Diseases, Ohio State University Medical Center, Columbus (C.D.M.); the Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York (J.A.A.); the Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham (E.T.O.); the Department of Medicine, Duke Global Health Institute and Duke Clinical Research Institute, Duke University (G.S.B.), and Duke University Research Institute, Duke University School of Medicine (P.S.D.) - both in Durham, NC; the Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles (J.S.C.); the Infectious Diseases Service, Hospital Clinic and University of Barcelona, Barcelona, and CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid - both in Spain (E.M.); DLH, Silver Spring (J.C.R.), and the Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute (P.D.-N.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (B.A.-S.), National Institutes of Health, Bethesda - all in Maryland; and Cleerly, Denver (U.H.)
| | - Stephen D Wiviott
- From the Metabolism Unit (S.K.G., K.V.F., M.V.Z., M.R.D., E.S.F., S.E.L., S.M.), the Cardiovascular Imaging Research Center, Department of Radiology (K.P., B.F., M.T.L.), and the Yvonne L. Munn Center for Nursing Research (S.E.L.), Massachusetts General Hospital and Harvard Medical School, the Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health (T.U., J.L.-C., H.J.R.), and the Thrombolysis in Myocardial Infarction Study Group, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School (S.D.W.) - all in Boston; the Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati (C.J.F.), and the Division of Infectious Diseases, Ohio State University Medical Center, Columbus (C.D.M.); the Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York (J.A.A.); the Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham (E.T.O.); the Department of Medicine, Duke Global Health Institute and Duke Clinical Research Institute, Duke University (G.S.B.), and Duke University Research Institute, Duke University School of Medicine (P.S.D.) - both in Durham, NC; the Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles (J.S.C.); the Infectious Diseases Service, Hospital Clinic and University of Barcelona, Barcelona, and CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid - both in Spain (E.M.); DLH, Silver Spring (J.C.R.), and the Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute (P.D.-N.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (B.A.-S.), National Institutes of Health, Bethesda - all in Maryland; and Cleerly, Denver (U.H.)
| | - Borek Foldyna
- From the Metabolism Unit (S.K.G., K.V.F., M.V.Z., M.R.D., E.S.F., S.E.L., S.M.), the Cardiovascular Imaging Research Center, Department of Radiology (K.P., B.F., M.T.L.), and the Yvonne L. Munn Center for Nursing Research (S.E.L.), Massachusetts General Hospital and Harvard Medical School, the Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health (T.U., J.L.-C., H.J.R.), and the Thrombolysis in Myocardial Infarction Study Group, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School (S.D.W.) - all in Boston; the Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati (C.J.F.), and the Division of Infectious Diseases, Ohio State University Medical Center, Columbus (C.D.M.); the Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York (J.A.A.); the Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham (E.T.O.); the Department of Medicine, Duke Global Health Institute and Duke Clinical Research Institute, Duke University (G.S.B.), and Duke University Research Institute, Duke University School of Medicine (P.S.D.) - both in Durham, NC; the Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles (J.S.C.); the Infectious Diseases Service, Hospital Clinic and University of Barcelona, Barcelona, and CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid - both in Spain (E.M.); DLH, Silver Spring (J.C.R.), and the Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute (P.D.-N.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (B.A.-S.), National Institutes of Health, Bethesda - all in Maryland; and Cleerly, Denver (U.H.)
| | - Sara E Looby
- From the Metabolism Unit (S.K.G., K.V.F., M.V.Z., M.R.D., E.S.F., S.E.L., S.M.), the Cardiovascular Imaging Research Center, Department of Radiology (K.P., B.F., M.T.L.), and the Yvonne L. Munn Center for Nursing Research (S.E.L.), Massachusetts General Hospital and Harvard Medical School, the Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health (T.U., J.L.-C., H.J.R.), and the Thrombolysis in Myocardial Infarction Study Group, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School (S.D.W.) - all in Boston; the Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati (C.J.F.), and the Division of Infectious Diseases, Ohio State University Medical Center, Columbus (C.D.M.); the Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York (J.A.A.); the Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham (E.T.O.); the Department of Medicine, Duke Global Health Institute and Duke Clinical Research Institute, Duke University (G.S.B.), and Duke University Research Institute, Duke University School of Medicine (P.S.D.) - both in Durham, NC; the Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles (J.S.C.); the Infectious Diseases Service, Hospital Clinic and University of Barcelona, Barcelona, and CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid - both in Spain (E.M.); DLH, Silver Spring (J.C.R.), and the Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute (P.D.-N.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (B.A.-S.), National Institutes of Health, Bethesda - all in Maryland; and Cleerly, Denver (U.H.)
| | - Patrice Desvigne-Nickens
- From the Metabolism Unit (S.K.G., K.V.F., M.V.Z., M.R.D., E.S.F., S.E.L., S.M.), the Cardiovascular Imaging Research Center, Department of Radiology (K.P., B.F., M.T.L.), and the Yvonne L. Munn Center for Nursing Research (S.E.L.), Massachusetts General Hospital and Harvard Medical School, the Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health (T.U., J.L.-C., H.J.R.), and the Thrombolysis in Myocardial Infarction Study Group, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School (S.D.W.) - all in Boston; the Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati (C.J.F.), and the Division of Infectious Diseases, Ohio State University Medical Center, Columbus (C.D.M.); the Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York (J.A.A.); the Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham (E.T.O.); the Department of Medicine, Duke Global Health Institute and Duke Clinical Research Institute, Duke University (G.S.B.), and Duke University Research Institute, Duke University School of Medicine (P.S.D.) - both in Durham, NC; the Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles (J.S.C.); the Infectious Diseases Service, Hospital Clinic and University of Barcelona, Barcelona, and CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid - both in Spain (E.M.); DLH, Silver Spring (J.C.R.), and the Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute (P.D.-N.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (B.A.-S.), National Institutes of Health, Bethesda - all in Maryland; and Cleerly, Denver (U.H.)
| | - Beverly Alston-Smith
- From the Metabolism Unit (S.K.G., K.V.F., M.V.Z., M.R.D., E.S.F., S.E.L., S.M.), the Cardiovascular Imaging Research Center, Department of Radiology (K.P., B.F., M.T.L.), and the Yvonne L. Munn Center for Nursing Research (S.E.L.), Massachusetts General Hospital and Harvard Medical School, the Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health (T.U., J.L.-C., H.J.R.), and the Thrombolysis in Myocardial Infarction Study Group, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School (S.D.W.) - all in Boston; the Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati (C.J.F.), and the Division of Infectious Diseases, Ohio State University Medical Center, Columbus (C.D.M.); the Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York (J.A.A.); the Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham (E.T.O.); the Department of Medicine, Duke Global Health Institute and Duke Clinical Research Institute, Duke University (G.S.B.), and Duke University Research Institute, Duke University School of Medicine (P.S.D.) - both in Durham, NC; the Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles (J.S.C.); the Infectious Diseases Service, Hospital Clinic and University of Barcelona, Barcelona, and CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid - both in Spain (E.M.); DLH, Silver Spring (J.C.R.), and the Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute (P.D.-N.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (B.A.-S.), National Institutes of Health, Bethesda - all in Maryland; and Cleerly, Denver (U.H.)
| | - Jorge Leon-Cruz
- From the Metabolism Unit (S.K.G., K.V.F., M.V.Z., M.R.D., E.S.F., S.E.L., S.M.), the Cardiovascular Imaging Research Center, Department of Radiology (K.P., B.F., M.T.L.), and the Yvonne L. Munn Center for Nursing Research (S.E.L.), Massachusetts General Hospital and Harvard Medical School, the Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health (T.U., J.L.-C., H.J.R.), and the Thrombolysis in Myocardial Infarction Study Group, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School (S.D.W.) - all in Boston; the Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati (C.J.F.), and the Division of Infectious Diseases, Ohio State University Medical Center, Columbus (C.D.M.); the Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York (J.A.A.); the Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham (E.T.O.); the Department of Medicine, Duke Global Health Institute and Duke Clinical Research Institute, Duke University (G.S.B.), and Duke University Research Institute, Duke University School of Medicine (P.S.D.) - both in Durham, NC; the Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles (J.S.C.); the Infectious Diseases Service, Hospital Clinic and University of Barcelona, Barcelona, and CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid - both in Spain (E.M.); DLH, Silver Spring (J.C.R.), and the Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute (P.D.-N.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (B.A.-S.), National Institutes of Health, Bethesda - all in Maryland; and Cleerly, Denver (U.H.)
| | - Sara McCallum
- From the Metabolism Unit (S.K.G., K.V.F., M.V.Z., M.R.D., E.S.F., S.E.L., S.M.), the Cardiovascular Imaging Research Center, Department of Radiology (K.P., B.F., M.T.L.), and the Yvonne L. Munn Center for Nursing Research (S.E.L.), Massachusetts General Hospital and Harvard Medical School, the Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health (T.U., J.L.-C., H.J.R.), and the Thrombolysis in Myocardial Infarction Study Group, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School (S.D.W.) - all in Boston; the Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati (C.J.F.), and the Division of Infectious Diseases, Ohio State University Medical Center, Columbus (C.D.M.); the Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York (J.A.A.); the Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham (E.T.O.); the Department of Medicine, Duke Global Health Institute and Duke Clinical Research Institute, Duke University (G.S.B.), and Duke University Research Institute, Duke University School of Medicine (P.S.D.) - both in Durham, NC; the Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles (J.S.C.); the Infectious Diseases Service, Hospital Clinic and University of Barcelona, Barcelona, and CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid - both in Spain (E.M.); DLH, Silver Spring (J.C.R.), and the Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute (P.D.-N.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (B.A.-S.), National Institutes of Health, Bethesda - all in Maryland; and Cleerly, Denver (U.H.)
| | - Udo Hoffmann
- From the Metabolism Unit (S.K.G., K.V.F., M.V.Z., M.R.D., E.S.F., S.E.L., S.M.), the Cardiovascular Imaging Research Center, Department of Radiology (K.P., B.F., M.T.L.), and the Yvonne L. Munn Center for Nursing Research (S.E.L.), Massachusetts General Hospital and Harvard Medical School, the Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health (T.U., J.L.-C., H.J.R.), and the Thrombolysis in Myocardial Infarction Study Group, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School (S.D.W.) - all in Boston; the Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati (C.J.F.), and the Division of Infectious Diseases, Ohio State University Medical Center, Columbus (C.D.M.); the Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York (J.A.A.); the Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham (E.T.O.); the Department of Medicine, Duke Global Health Institute and Duke Clinical Research Institute, Duke University (G.S.B.), and Duke University Research Institute, Duke University School of Medicine (P.S.D.) - both in Durham, NC; the Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles (J.S.C.); the Infectious Diseases Service, Hospital Clinic and University of Barcelona, Barcelona, and CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid - both in Spain (E.M.); DLH, Silver Spring (J.C.R.), and the Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute (P.D.-N.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (B.A.-S.), National Institutes of Health, Bethesda - all in Maryland; and Cleerly, Denver (U.H.)
| | - Michael T Lu
- From the Metabolism Unit (S.K.G., K.V.F., M.V.Z., M.R.D., E.S.F., S.E.L., S.M.), the Cardiovascular Imaging Research Center, Department of Radiology (K.P., B.F., M.T.L.), and the Yvonne L. Munn Center for Nursing Research (S.E.L.), Massachusetts General Hospital and Harvard Medical School, the Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health (T.U., J.L.-C., H.J.R.), and the Thrombolysis in Myocardial Infarction Study Group, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School (S.D.W.) - all in Boston; the Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati (C.J.F.), and the Division of Infectious Diseases, Ohio State University Medical Center, Columbus (C.D.M.); the Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York (J.A.A.); the Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham (E.T.O.); the Department of Medicine, Duke Global Health Institute and Duke Clinical Research Institute, Duke University (G.S.B.), and Duke University Research Institute, Duke University School of Medicine (P.S.D.) - both in Durham, NC; the Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles (J.S.C.); the Infectious Diseases Service, Hospital Clinic and University of Barcelona, Barcelona, and CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid - both in Spain (E.M.); DLH, Silver Spring (J.C.R.), and the Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute (P.D.-N.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (B.A.-S.), National Institutes of Health, Bethesda - all in Maryland; and Cleerly, Denver (U.H.)
| | - Heather J Ribaudo
- From the Metabolism Unit (S.K.G., K.V.F., M.V.Z., M.R.D., E.S.F., S.E.L., S.M.), the Cardiovascular Imaging Research Center, Department of Radiology (K.P., B.F., M.T.L.), and the Yvonne L. Munn Center for Nursing Research (S.E.L.), Massachusetts General Hospital and Harvard Medical School, the Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health (T.U., J.L.-C., H.J.R.), and the Thrombolysis in Myocardial Infarction Study Group, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School (S.D.W.) - all in Boston; the Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati (C.J.F.), and the Division of Infectious Diseases, Ohio State University Medical Center, Columbus (C.D.M.); the Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York (J.A.A.); the Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham (E.T.O.); the Department of Medicine, Duke Global Health Institute and Duke Clinical Research Institute, Duke University (G.S.B.), and Duke University Research Institute, Duke University School of Medicine (P.S.D.) - both in Durham, NC; the Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles (J.S.C.); the Infectious Diseases Service, Hospital Clinic and University of Barcelona, Barcelona, and CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid - both in Spain (E.M.); DLH, Silver Spring (J.C.R.), and the Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute (P.D.-N.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (B.A.-S.), National Institutes of Health, Bethesda - all in Maryland; and Cleerly, Denver (U.H.)
| | - Pamela S Douglas
- From the Metabolism Unit (S.K.G., K.V.F., M.V.Z., M.R.D., E.S.F., S.E.L., S.M.), the Cardiovascular Imaging Research Center, Department of Radiology (K.P., B.F., M.T.L.), and the Yvonne L. Munn Center for Nursing Research (S.E.L.), Massachusetts General Hospital and Harvard Medical School, the Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health (T.U., J.L.-C., H.J.R.), and the Thrombolysis in Myocardial Infarction Study Group, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School (S.D.W.) - all in Boston; the Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati (C.J.F.), and the Division of Infectious Diseases, Ohio State University Medical Center, Columbus (C.D.M.); the Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York (J.A.A.); the Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham (E.T.O.); the Department of Medicine, Duke Global Health Institute and Duke Clinical Research Institute, Duke University (G.S.B.), and Duke University Research Institute, Duke University School of Medicine (P.S.D.) - both in Durham, NC; the Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles (J.S.C.); the Infectious Diseases Service, Hospital Clinic and University of Barcelona, Barcelona, and CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid - both in Spain (E.M.); DLH, Silver Spring (J.C.R.), and the Division of Cardiovascular Sciences, National Heart, Lung, and Blood Institute (P.D.-N.), and the Division of AIDS, National Institute of Allergy and Infectious Diseases (B.A.-S.), National Institutes of Health, Bethesda - all in Maryland; and Cleerly, Denver (U.H.)
| |
Collapse
|
13
|
Schnittman SR, Lu MT, Mayrhofer T, Burdo TH, Fitch KV, McCallum S, Fulda ES, Zanni MV, Foldyna B, Malvestutto C, Fichtenbaum CJ, Aberg JA, Bloomfield GS, Overton ET, Currier J, Tebas P, Sha BE, Ribaudo HJ, Flynn JM, Douglas PS, Erlandson KM, Grinspoon SK. Cytomegalovirus Immunoglobulin G (IgG) Titer and Coronary Artery Disease in People With Human Immunodeficiency Virus (HIV). Clin Infect Dis 2023; 76:e613-e621. [PMID: 35975297 PMCID: PMC10169419 DOI: 10.1093/cid/ciac662] [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: 06/19/2022] [Revised: 08/05/2022] [Accepted: 08/12/2022] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Cytomegalovirus (CMV) infection is thought to result in increased immune activation in people with human immunodeficiency virus (HIV, PWH). Although some data have linked asymptomatic CMV infection to cardiovascular disease among PWH, it remains unknown whether CMV is associated with increased or high-risk coronary plaque. METHODS The Randomized Trial to Prevent Vascular Events in HIV (REPRIEVE) enrolled PWH aged 40-75 years on stable antiretroviral therapy (ART) with low-to-moderate atherosclerotic cardiovascular disease (ASCVD) risk. Among a subset of US REPRIEVE participants, coronary plaque was assessed by coronary computed tomography angiography. Here, we assessed the relationship between CMV immunoglobulin G (IgG) titer and (1) levels of immune activation, (2) inflammatory biomarkers, and (3) coronary plaque phenotypes at study entry. RESULTS Of 672 participants, mean age was 51 years, 83% were men, median ASCVD risk score was 4.5%, and 66% had current CD4+ T-cell count ≥500 cells/mm3. Higher CMV IgG quartile group was associated with older age and lower current and nadir CD4+ T-cell counts. CMV IgG titer was associated with specific inflammatory biomarkers (sCD163, MCP-1, interleukin [IL]-6, hsCRP) in univariate analysis, but not after controlling for HIV-specific factors. In contrast, CMV IgG titer was not associated with coronary artery disease indexes, including presence of plaque, coronary artery calcium (CAC) score >0, vulnerable plaque presence, or Leaman score >5. CONCLUSIONS No meaningful association was seen between CMV IgG titer and coronary artery disease indexes among ART-treated PWH at study enrollment. Longitudinal assessments in REPRIEVE will determine the relationship of CMV IgG titer to plaque progression and cardiovascular events. CLINICAL TRIALS REGISTRATION NCT02344290.
Collapse
Affiliation(s)
- Samuel R Schnittman
- Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Michael T Lu
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Thomas Mayrhofer
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- School of Business Studies, Stralsund University of Applied Sciences, Stralsund, Germany
| | - Tricia H Burdo
- Department of Microbiology, Immunology, and Inflammation and Center for Neuro-Virology and Gene Editing, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Kathleen V Fitch
- Metabolism Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sara McCallum
- Metabolism Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Evelynne S Fulda
- Metabolism Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Markella V Zanni
- Metabolism Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Borek Foldyna
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Carlos Malvestutto
- Division of Infectious Diseases, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Carl J Fichtenbaum
- Division of Infectious Diseases, Department of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
| | - Judith A Aberg
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Gerald S Bloomfield
- Division of Cardiology, Department of Medicine and Duke Clinical Research Institute, Duke University, Durham, North Carolina, USA
| | - Edgar T Overton
- Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Judith Currier
- Division of Infectious Diseases, University of California at Los Angeles, Los Angeles, California, USA
| | - Pablo Tebas
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Beverly E Sha
- Division of Infectious Diseases, Department of Internal Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Heather J Ribaudo
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Jacqueline M Flynn
- Department of Microbiology, Immunology, and Inflammation and Center for Neuro-Virology and Gene Editing, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Pamela S Douglas
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Kristine M Erlandson
- Division of Infectious Diseases, Department of Medicine, University of Colorado, Aurora, Colorado, USA
| | - Steven K Grinspoon
- Metabolism Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
14
|
Kumar P, Arendt C, Martin S, Al Soufi S, DeLeuw P, Nagel E, Puntmann VO. Multimodality Imaging in HIV-Associated Cardiovascular Complications: A Comprehensive Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:2201. [PMID: 36767567 PMCID: PMC9915416 DOI: 10.3390/ijerph20032201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/17/2023] [Accepted: 01/20/2023] [Indexed: 06/18/2023]
Abstract
Human immunodeficiency virus (HIV) infection is a leading cause of mortality and morbidity worldwide. The introduction of antiretroviral therapy (ART) has significantly reduced the risk of developing acquired immune deficiency syndrome and increased life expectancy, approaching that of the general population. However, people living with HIV have a substantially increased risk of cardiovascular diseases despite long-term viral suppression using ART. HIV-associated cardiovascular complications encompass a broad spectrum of diseases that involve the myocardium, pericardium, coronary arteries, valves, and systemic and pulmonary vasculature. Traditional risk stratification tools do not accurately predict cardiovascular risk in this population. Multimodality imaging plays an essential role in the evaluation of various HIV-related cardiovascular complications. Here, we emphasize the role of multimodality imaging in establishing the diagnosis and aetiopathogenesis of various cardiovascular manifestations related to chronic HIV disease. This review also provides a critical appraisal of contemporary data and illustrative cases.
Collapse
Affiliation(s)
- Parveen Kumar
- Institute of Experimental and Translational Cardiac Imaging, DZHK, Centre for Cardiovascular Imaging, University Hospital Frankfurt, 60590 Frankfurt am Main, Germany
| | - Christophe Arendt
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, 60590 Frankfurt am Main, Germany
| | - Simon Martin
- Institute of Experimental and Translational Cardiac Imaging, DZHK, Centre for Cardiovascular Imaging, University Hospital Frankfurt, 60590 Frankfurt am Main, Germany
| | - Safaa Al Soufi
- Institute of Experimental and Translational Cardiac Imaging, DZHK, Centre for Cardiovascular Imaging, University Hospital Frankfurt, 60590 Frankfurt am Main, Germany
| | | | - Eike Nagel
- Institute of Experimental and Translational Cardiac Imaging, DZHK, Centre for Cardiovascular Imaging, University Hospital Frankfurt, 60590 Frankfurt am Main, Germany
| | - Valentina O. Puntmann
- Institute of Experimental and Translational Cardiac Imaging, DZHK, Centre for Cardiovascular Imaging, University Hospital Frankfurt, 60590 Frankfurt am Main, Germany
| |
Collapse
|
15
|
Suero-Abreu GA, Zanni MV, Neilan TG. Atherosclerosis With Immune Checkpoint Inhibitor Therapy: Evidence, Diagnosis, and Management: JACC: CardioOncology State-of-the-Art Review. JACC CardioOncol 2022; 4:598-615. [PMID: 36636438 PMCID: PMC9830225 DOI: 10.1016/j.jaccao.2022.11.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 11/13/2022] [Indexed: 12/24/2022] Open
Abstract
As the clinical applications of immune checkpoint inhibitors (ICIs) expand, our knowledge of the potential adverse effects of these drugs continues to broaden. Emerging evidence supports the association between ICI therapy with accelerated atherosclerosis and atherosclerotic cardiovascular (CV) events. We discuss the biological plausibility and the clinical evidence supporting an effect of inhibition of these immune checkpoints on atherosclerotic CV disease. Further, we provide a perspective on potential diagnostic and pharmacological strategies to reduce atherosclerotic risk in ICI-treated patients. Our understanding of the pathophysiology of ICI-related atherosclerosis is in its early stages. Further research is needed to identify the mechanisms linking ICI therapy to atherosclerosis, leverage the insight that ICI therapy provides into CV biology, and develop robust approaches to manage the expanding cohort of patients who may be at risk for atherosclerotic CV disease.
Collapse
Affiliation(s)
| | - Markella V. Zanni
- Metabolism Unit, Division of Endocrinology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Tomas G. Neilan
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA,Cardiovascular Imaging Research Center, Department of Radiology and Department of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA,Address for correspondence: Dr Tomas G. Neilan, Cardio-Oncology Program and Cardiovascular Imaging Research Center (CIRC), Massachusetts General Hospital, 165 Cambridge Street, Suite 400, Boston, Massachusetts 02114, USA. @TomasNeilan
| |
Collapse
|
16
|
Werede AT, Terry JG, Nair S, Temu TM, Shepherd BE, Bailin SS, Mashayekhi M, Gabriel CL, Lima M, Woodward BO, Hannah L, Mallal SA, Beckman JA, Li JZ, Fajnzylber J, Harrison DG, Carr JJ, Koethe JR, Wanjalla CN. Mean Coronary Cross-Sectional Area as a Measure of Arterial Remodeling Using Noncontrast CT Imaging in Persons With HIV. J Am Heart Assoc 2022; 11:e025768. [PMID: 36382956 PMCID: PMC9851442 DOI: 10.1161/jaha.122.025768] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 08/02/2022] [Indexed: 11/17/2022]
Abstract
Background Persons with HIV have a higher prevalence of coronary artery disease compared with their HIV-negative counterparts. Earlier identification of subclinical atherosclerosis may provide a greater opportunity for cardiovascular disease risk reduction. We investigated coronary cross-sectional area (CorCSA) by noncontrasted computed tomography imaging as a noninvasive measure of arterial remodeling among virally suppressed persons with HIV. Methods and Results We assessed 105 persons with HIV with a spectrum of cardiometabolic health. All participants underwent computed tomography imaging to assess the mean corCSA of the proximal left anterior descending artery and 28 participants underwent additional coronary computed tomography angiography. Partial Spearman rank correlations adjusted for cardiovascular disease risk factors were used to assess relationships of corCSA with anthropometric measurements, HIV-related factors, and plasma cytokines. Mean corCSA measured by noncontrast computed tomography and coronary computed tomography angiography were strongly correlated (ρ=0.91, P<0.0001). Higher mean corCSA was present in those with coronary artery calcium (P=0.005) and it correlated with participants' atherosclerotic cardiovascular disease risk score (ρ=0.35, P=0.01). After adjusting for established cardiovascular disease risk factors, we observed an inverse relationship between corCSA and CD4+ T-cell count (ρ=-0.2, P=0.047). Removal of age from the model strengthened the relationships between corCSA and antiretroviral therapy duration (from ρ=0.19, P=0.08 to ρ=0.3, P=0.01). CorCSA was also inversely correlated with plasma IL-10 (ρ=-0.25, P=0.03) but had no relationship with IL-6 (ρ=0.11, P=0.4) or IL-1β (ρ=0.08, P=0.5). Conclusions Positive coronary arterial remodeling, an imaging marker of subclinical atherosclerosis, is associated with a lower CD4 T-cell count, lower circulating IL-10, and possibly a longer antiretroviral therapy duration in persons with HIV. Registration Clinicaltrials.gov; Unique identifier: NCT04451980.
Collapse
Affiliation(s)
- Ayoda T. Werede
- Division of Infectious DiseasesVanderbilt University Medical CenterNashvilleTN
| | - James G. Terry
- Department of RadiologyVanderbilt University Medical CenterNashvilleTN
| | - Sangeeta Nair
- Department of RadiologyVanderbilt University Medical CenterNashvilleTN
| | - Tecla M. Temu
- Departments of Global HealthUniversity of WashingtonSeattleWA
| | - Bryan E. Shepherd
- Department of BiostatisticsVanderbilt University Medical CenterNashvilleTN
| | - Samuel S. Bailin
- Division of Infectious DiseasesVanderbilt University Medical CenterNashvilleTN
| | - Mona Mashayekhi
- Division of Diabetes, Endocrinology and MetabolismVanderbilt University Medical CenterNashvilleTN
| | - Curtis L. Gabriel
- Division of GastroenterologyVanderbilt University Medical CenterNashvilleTN
| | - Morgan Lima
- Division of Infectious DiseasesVanderbilt University Medical CenterNashvilleTN
| | | | - LaToya Hannah
- Division of Infectious DiseasesVanderbilt University Medical CenterNashvilleTN
| | - Simon A. Mallal
- Division of Infectious DiseasesVanderbilt University Medical CenterNashvilleTN
- Department of Pathology, Microbiology and ImmunologyVanderbilt University Medical CenterNashvilleTN
- Department of Biomedical InformaticsVanderbilt University Medical CenterNashvilleTN
| | - Joshua A. Beckman
- Division of CardiologyVanderbilt University Medical CenterNashvilleTN
| | - Jonathan Z. Li
- Center for AIDS Research Clinical CoreBrigham and Women’s HospitalBostonMA
| | - Jesse Fajnzylber
- Center for AIDS Research Clinical CoreBrigham and Women’s HospitalBostonMA
| | - David G. Harrison
- Division of Clinical PharmacologyVanderbilt University Medical CenterNashvilleTN
| | - John Jeffrey Carr
- Department of RadiologyVanderbilt University Medical CenterNashvilleTN
| | - John R. Koethe
- Division of Infectious DiseasesVanderbilt University Medical CenterNashvilleTN
- Veterans Affairs Tennessee Valley Healthcare SystemNashvilleTN
| | | |
Collapse
|
17
|
Kolossváry M, deFilippi C, Lu MT, Zanni MV, Fulda ES, Foldyna B, Ribaudo H, Mayrhofer T, Collier AC, Bloomfield GS, Fichtenbaum C, Overton ET, Aberg JA, Currier J, Fitch KV, Douglas PS, Grinspoon SK. Proteomic Signature of Subclinical Coronary Artery Disease in People With HIV: Analysis of the REPRIEVE Mechanistic Substudy. J Infect Dis 2022; 226:1809-1822. [PMID: 35535576 PMCID: PMC10205625 DOI: 10.1093/infdis/jiac196] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/06/2022] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND People with HIV (PWH) have subclinical coronary artery disease (CAD) despite low traditional atherosclerotic cardiovascular disease (ASCVD) risk scores. Coronary plaque in PWH presents as a unique phenotype, but little is known about the contributions of specific inflammatory pathways to plaque phenotypes in PWH. METHODS The REPRIEVE Mechanistic Substudy enrolled PWH on ART without known cardiovascular disease. We used a targeted discovery proteomics approach to evaluate 246 unique proteins representing cardiovascular, inflammatory, and immune pathways. Proteomic signatures were determined for presence of coronary artery calcium (CAC > 0) and presence of coronary plaque. RESULTS Data were available for 662 participants (aged 51 [SD 6] years, ASCVD risk score 4.9% [SD 3.1%]). Among 12 proteins associated with both CAC and presence of coronary plaque, independent of ASCVD risk score, the odds ratios were highest for NRP1: 5.1 (95% confidence interval [CI], 2.3-11.4) for CAC and 2.9 (95% CI, 1.4-6.1) for presence of plaque. Proteins uniquely related to presence of plaque were CST3, LTBR, MEPE, PLC, SERPINA5, and TNFSF13B; in contrast, DCN, IL-6RA, OSMR, ST2, and VCAM1 were only related to CAC. CONCLUSIONS Distinct immune and inflammatory pathways are differentially associated with subclinical CAD phenotypes among PWH. This comprehensive set of targets should be further investigated to reduce atherosclerosis and ASCVD in PWH. CLINICAL TRIALS REGISTRATION NCT02344290.
Collapse
Affiliation(s)
- Márton Kolossváry
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Chris deFilippi
- Inova Heart and Vascular Institute, Falls Church, Virginia, USA
| | - Michael T Lu
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Markella V Zanni
- Metabolism Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Evelynne S Fulda
- Metabolism Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Borek Foldyna
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Heather Ribaudo
- Center for Biostatistics in AIDS Research, Harvard T. H. Chan School of Public Health, Boston Massachusetts, USA
| | - Thomas Mayrhofer
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, Massachusetts, USA
- School of Business Studies, Stralsund University of Applied Sciences, Stralsund, Germany
| | - Ann C Collier
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Gerald S Bloomfield
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Carl Fichtenbaum
- Department of Medicine for Translational Research, University of Cincinnati, Cincinnati, Ohio, USA
| | - Edgar T Overton
- Division of Infectious Diseases, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Judith A Aberg
- Division of Infectious Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Judith Currier
- Division of Infectious Diseases, University of California at Los Angeles, Los Angeles, California, USA
| | - Kathleen V Fitch
- Metabolism Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Pamela S Douglas
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina, USA
| | - Steven K Grinspoon
- Metabolism Unit, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
18
|
Fitch KV, Fulda ES, Grinspoon SK. Statins for primary cardiovascular disease prevention among people with HIV: emergent directions. Curr Opin HIV AIDS 2022; 17:293-300. [PMID: 35938463 PMCID: PMC9415230 DOI: 10.1097/coh.0000000000000752] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW While people with HIV (PWH) are living longer due to advances in antiretroviral therapy, recent data have demonstrated an increased risk of cardiovascular disease (CVD) among this population. This increased risk is thought to be due to both traditional (for example, smoking, diabetes) and HIV-specific (for example, inflammation, persistent immune activation) risk factors. This review focuses on the potential for statin therapy to mitigate this increased risk. RECENT FINDINGS Several randomized clinical trials have demonstrated that statins, a class of lipid-lowering medications, are effective as a primary CVD prevention strategy among people without HIV. Among PWH, statins have been shown to lower cholesterol, exert immunomodulatory effects, stabilize coronary atherosclerotic plaque, and even induce plaque regression. SUMMARY Prevention of CVD among the aging population of people with controlled, but chronic, HIV is vital. Data exploring primary prevention in this context are thus far limited. The Randomized Trial to Prevent Vascular Events in HIV (REPRIEVE) is ongoing; this trial will inform the field by investigating the effects of pitavastatin calcium as a primary prevention strategy for major adverse cardiovascular events among PWH on antiretroviral therapy (ART) at low-to-moderate traditional CVD risk.
Collapse
Affiliation(s)
- Kathleen V Fitch
- Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | | |
Collapse
|
19
|
Assessment of Clinical Features in HIV-Infected Patients with Acute Coronary Syndromes Undergoing Percutaneous Coronary Intervention in China. J Interv Cardiol 2022; 2022:8351304. [PMID: 35847237 PMCID: PMC9256447 DOI: 10.1155/2022/8351304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/01/2022] [Accepted: 06/15/2022] [Indexed: 11/24/2022] Open
Abstract
Objectives We aimed to compare coronary risk factors, burden of coronary artery disease (CAD), and 1-year prognosis of people living with HIV (PLWH) and HIV-negative controls who underwent percutaneous coronary intervention (PCI) for acute coronary syndromes (ACSs). Background Cardiovascular disease is drawing more and more attention in PLWH since effective antiretroviral therapy (ART) has been available. Clinical characteristics and outcomes of PLWH undergoing PCI for ACS in China remain unknown. Methods We compared demographic characteristics, angiographic features, and 1-year outcomes of 48 PLWH versus 48 HIV-negative controls matched for age (±2 years), sex, diabetes mellitus, and year of PCI (±2 years) in Beijing Ditan Hospital, Capital Medical University from January 2008 to November 2020. Results In PLWH (mean age: 53.6 ± 10.6 years, 95.8% male, and 79.2% on ART), high-density lipoprotein cholesterol was lower than in HIV-negative controls; however, the statin use was more common, the incidence of hypertension was lower, and low-density lipoprotein cholesterol, and the body mass index were significantly lower than in controls. Two groups had a similar extent of coronary atherosclerosis as measured by the presence of multivessel diseases and the median Gensini score; however, lesions of PLWH were longer and were more likely to locate at the proximal segment of the coronary artery. In addition, the risk of major adverse cardiac and cerebrovascular events at 1 year was similar in both groups. Conclusion PLWH undergoing PCI displayed similar CAD burden and 1-year prognosis compared with HIV-negative patients. Early detection of cardiovascular risk factors and appropriate secondary prevention of CAD in PLWH might alleviate the risk of severe adverse cardiovascular events.
Collapse
|
20
|
Kovacs L, Kress TC, Belin de Chantemèle EJ. HIV, Combination Antiretroviral Therapy, and Vascular Diseases in Men and Women. JACC Basic Transl Sci 2022; 7:410-421. [PMID: 35540101 PMCID: PMC9079796 DOI: 10.1016/j.jacbts.2021.10.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 10/22/2021] [Accepted: 10/26/2021] [Indexed: 12/01/2022]
Abstract
Thanks to the advent of combination antiretroviral therapy (cART), people living with human immunodeficiency virus (HIV) (PLWH) experienced a marked increase in life expectancy but are now at higher risk for cardiovascular disease (CVD), the current leading cause of death in PLWH on cART. Although HIV preponderantly affects men over women, manifestations of HIV-related CVD differ by sex with women experiencing greater risks than men. Despite extensive investigation, the etiopathology of CVD, notably the respective contribution of viral infection and cART, remain ill-defined. However, both viral infection and cART have been reported to contribute to endothelial dysfunction, the precursor and major cause of atherosclerosis-associated CVD, through mechanisms involving endothelial cell activation, inflammation, and oxidative stress, all leading to reduced nitric oxide bioavailability. Therefore, preserving endothelial function in PLWH on cART should be a main target to reduce CVD morbidity and mortality, notably in females.
Collapse
Key Words
- CVD, cardiovascular disease
- FMD, flow-mediated dilatation
- HF, heart failure
- HIV
- HIV, human immunodeficiency virus
- MI, myocardial infarction
- NO, nitric oxide
- PAD, peripheral artery disease
- PH, pulmonary hypertension
- PLWH, people living with HIV
- cART, combination antiretroviral therapy
- cIMT, carotid intima-media thickness
- combination antiretroviral therapy
- endothelial dysfunction
- sex differences
Collapse
Affiliation(s)
- Laszlo Kovacs
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Taylor C Kress
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| | - Eric J Belin de Chantemèle
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA.,Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta Georgia, USA
| |
Collapse
|
21
|
Longenecker CT, Bogorodskaya M, Margevicius S, Nazzinda R, Bittencourt MS, Erem G, Nalukwago S, Huaman MA, Ghoshhajra BB, Siedner MJ, Juchnowski SM, Zidar DA, McComsey GA, Kityo C. Sex modifies the association between HIV and coronary artery disease among older adults in Uganda. J Int AIDS Soc 2022; 25:e25868. [PMID: 34995413 PMCID: PMC8741262 DOI: 10.1002/jia2.25868] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 12/15/2021] [Indexed: 02/02/2023] Open
Abstract
INTRODUCTION Little is known about the epidemiology of coronary artery disease (CAD) in sub-Saharan Africa, where the majority of people living with HIV (PLHIV) live. We assessed the association of HIV with CAD and explored relationships with monocyte activation in sex-stratified analyses of older PLHIV and people without HIV (PWOH) in Uganda. METHODS The Ugandan Study of HIV effects on the Myocardium and Atherosclerosis (mUTIMA) follows 100 PLHIV on antiretroviral therapy (ART) and 100 age- and sex-matched PWOH controls in Kampala, Uganda; all >45 years of age with >1 cardiovascular disease risk factor. At the year 2 exam (2017-2019), 189 participants had available coronary calcium score and 165 had coronary CT angiography (CCTA) for this analysis. A subset of participants (n = 107) had both CCTA and fresh whole blood flow cytometry for monocyte phenotyping. RESULTS Median age was 57.8 years and 63% were females. Overall, 88% had hypertension, 37% had diabetes and 4% were smokers. Atherosclerotic cardiovascular disease (ASCVD) risk was modestly higher for PWOH, but not statistically significant (median 10-year ASCVD risk 7.2% for PLHIV vs. 8.6% for PWOH, p = 0.09). Median duration of ART was 12.7 years and 86% had suppressed viral load. Despite a high prevalence of risk factors, only 34/165 (21%, 95% CI 15-28%) had any coronary plaque. After adjustment for ASCVD risk score, HIV status was not associated with CAD (OR 0.55, 95% CI 0.23-1.30) but was associated with more severe CAD (segment severity score>3) among those with disease (OR 10.9, 95% CI 1.67-70.45). Females had a trend towards higher odds of CAD among PLHIV (OR 4.1, 95% CI 0.4-44.9), but a trend towards lower odds of CAD among PWOH (OR 0.30; 95% CI 0.07-1.3; HIV*sex interaction p = 0.019). CAD was positively correlated with classical monocytes (r = 0.3, p = 0.012) and negatively correlated with CX3CR1 expression (r = -0.31, p = 0.011) in PLHIV and negatively correlated with patrolling monocytes (r = -0.36, p = 0.031) and tissue factor expression (r = -0.39, p = 0.017) in PWOH. CONCLUSIONS Our results suggest that HIV may be associated more with severity rather than the presence of CAD in Uganda. Sex differences in the HIV effect suggest that tailored CAD prevention strategies may be required in this setting.
Collapse
Affiliation(s)
- Chris T. Longenecker
- University Hospitals of ClevelandClevelandOhioUSA
- Case Western Reserve UniversityClevelandOhioUSA
| | - Milana Bogorodskaya
- Case Western Reserve UniversityClevelandOhioUSA
- MetroHealth Medical CenterClevelandOhioUSA
| | | | | | | | - Geoffrey Erem
- St. Francis Hospital NsambyaKampalaUganda
- Makerere University School of MedicineKampalaUganda
| | | | | | | | | | | | - David A. Zidar
- Case Western Reserve UniversityClevelandOhioUSA
- Louis Stokes Cleveland Veterans Affairs Medical CenterClevelandOhioUSA
| | - Grace A. McComsey
- University Hospitals of ClevelandClevelandOhioUSA
- Case Western Reserve UniversityClevelandOhioUSA
| | | |
Collapse
|
22
|
SADOUNI M, DURAND M, BOLDEANU I, DANIELI C, BODSON-CLERMONT P, MANSOUR S, BARIL JG, TROTTIER B, TREMBLAY C, CHARTRAND-LEFEBVRE C. Association of epicardial fat with noncalcified coronary plaque volume and with low attenuation plaque in people with HIV. AIDS 2021; 35:1575-1584. [PMID: 33831908 PMCID: PMC8286299 DOI: 10.1097/qad.0000000000002911] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES People with HIV are exposed to a higher risk of coronary artery disease (CAD) compared with the general population. Epicardial fat may play a unique role in promoting coronary atherosclerosis. We measured epicardial fat in participants living with HIV and controls and investigated its association with coronary plaque volume and low attenuation plaque, a marker of plaque vulnerability. DESIGN This is a cross-sectional study, nested in the Canadian HIV and Aging Cohort Study, a large prospective cohort actively following participants with HIV and controls. Participants with low/intermediate cardiovascular risk without symptoms/history of CAD were invited to undergo cardiac computed tomography (CT). METHODS Volume of epicardial fat, coronary plaque and low attenuation component of the plaque were measured. Association between epicardial fat, coronary plaque volume and low attenuation component was tested using adjusted regression analysis. RESULTS A total of 169 participants with HIV and 81 controls underwent cardiac CT. Participants with HIV had a greater epicardial fat volume compared with controls (P = 0.019). In participants with HIV, epicardial fat volume was positively associated with duration of nonnucleoside reverse transcriptase inhibitors (NNRTI) (β=2.19, P = 0.004). After adjustment for cardiovascular risk factors, epicardial fat volume was positively associated to noncalcified plaque volume [odds ratio (OR) = 1.09, P = 0.028] and to the low-attenuation plaque component portion (β=0.38, P = 0.026). CONCLUSION The association of epicardial fat volume to noncalcified plaque volume and to low attenuation component plaque may suggest a potential mechanism by which epicardial fat could be a silent driver of CAD in the HIV population.
Collapse
Affiliation(s)
- Manel SADOUNI
- Centre hospitalier de l’Université de Montréal (CHUM) Research Center, Montreal, Canada
- Université de Montréal, Montreal, Canada
| | - Madeleine DURAND
- Centre hospitalier de l’Université de Montréal (CHUM) Research Center, Montreal, Canada
- Université de Montréal, Montreal, Canada
- Internal Medicine, CHUM, Montreal, Canada
| | | | | | - Paule BODSON-CLERMONT
- Centre hospitalier de l’Université de Montréal (CHUM) Research Center, Montreal, Canada
| | | | - Jean-Guy BARIL
- Centre hospitalier de l’Université de Montréal (CHUM) Research Center, Montreal, Canada
- Université de Montréal, Montreal, Canada
- Medical Clinic Quartier Latin, Montreal, Canada
| | | | - Cécile TREMBLAY
- Centre hospitalier de l’Université de Montréal (CHUM) Research Center, Montreal, Canada
- Université de Montréal, Montreal, Canada
- Microbiology, CHUM, Montreal, Canada
| | - Carl CHARTRAND-LEFEBVRE
- Centre hospitalier de l’Université de Montréal (CHUM) Research Center, Montreal, Canada
- Université de Montréal, Montreal, Canada
- Radiology, CHUM, Montreal, Canada
| | | |
Collapse
|
23
|
Hatleberg CI, Ryom L, Sabin C. Cardiovascular risks associated with protease inhibitors for the treatment of HIV. Expert Opin Drug Saf 2021; 20:1351-1366. [PMID: 34047238 DOI: 10.1080/14740338.2021.1935863] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Introduction: Cumulative use of some first-generation protease inhibitors has been associated with higher rates of dyslipidemia and increased risk of cardiovascular disease. The protease inhibitors most commonly in use are atazanavir and darunavir, which have fewer detrimental lipid effects and greater tolerability. This paper aims to review the evidence of a potential association of these contemporary protease inhibitors with the risk of ischemic CVD and atherosclerotic markers.Areas covered: We searched for publications of randomized trials and observational studies on PubMed from 1 January 2000 onwards, using search terms including: protease inhibitors; darunavir; atazanavir; cardiovascular disease; cardiovascular events; dyslipidemia; mortality; carotid intima media thickness; arterial elasticity; arterial stiffness and drug discontinuation. Ongoing studies registered on clinicaltrials.gov as well as conference abstracts from major HIV conferences from 2015-2020 were also searched.Expert opinion: Atazanavir and darunavir are no longer part of first-line HIV treatment, but continue to be recommended as alternative first line, second- and third-line regimens, as part of two drug regimens, and darunavir is used as salvage therapy. Although these drugs will likely remain in use globally for several years to come, baseline CVD risk should be considered when considering their use, especially as the population with HIV ages.
Collapse
Affiliation(s)
- Camilla Ingrid Hatleberg
- Department of Infectious Diseases, Centre of Excellence for Health, Immunity and Infections (CHIP), Section 2100, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Lene Ryom
- Department of Infectious Diseases, Centre of Excellence for Health, Immunity and Infections (CHIP), Section 2100, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Caroline Sabin
- Centre for Clinical Research, Epidemiology, Modelling and Evaluation (CREME), Institute for Global Health,University College London, London, UK
| |
Collapse
|
24
|
Patel AA, Budoff MJ. Coronary Artery Disease in Patients with HIV Infection: An Update. Am J Cardiovasc Drugs 2021; 21:411-417. [PMID: 33184766 DOI: 10.1007/s40256-020-00451-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/30/2020] [Indexed: 12/13/2022]
Abstract
Premature cardiovascular disease among the HIV-infected population is of great concern among clinicians. The increased life expectancy of HIV-infected individuals is mainly due to the early detection of infection and the advent of antiretroviral therapy. Once known as a deadly disease, HIV infection has transitioned into a chronic condition. Cardiovascular disease in this population is thought to progress early due to traditional and non-traditional risk factors. Early detection of subclinical atherosclerosis has become a center of focus in research as our complete understanding of this process it not yet well known. Advancements in cardiac computed tomography angiography has enabled the exploration of coronary artery disease by further evaluation of coronary stenosis and plaque analysis. An increase in cardiovascular event rates in this population is currently thought to be linked to antiretroviral therapy, Framingham risk factors, and HIV. We sought to present an updated comprehensive review of the available literature on HIV related to atherosclerosis and cardiovascular risk.
Collapse
Affiliation(s)
- Amish A Patel
- Division of Cardiology, Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA
- Division of Cardiology, University of California Riverside School of Medicine, Riverside, CA, USA
| | - Matthew J Budoff
- Division of Cardiology, Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, 90502, USA.
| |
Collapse
|
25
|
Douglas PS, Umbleja T, Bloomfield GS, Fichtenbaum CJ, Zanni MV, Overton ET, Fitch KV, Kileel EM, Aberg JA, Currier J, Sponseller CA, Melbourne K, Avihingsanon A, Bustorff F, Estrada V, Ruxrungtham K, Saumoy M, Navar AM, Hoffmann U, Ribaudo HJ, Grinspoon S. Cardiovascular Risk and Health Among People With HIV Eligible for Primary Prevention: Insights From the REPRIEVE Trial. Clin Infect Dis 2021; 73:2009-2022. [PMID: 34134131 PMCID: PMC8664454 DOI: 10.1093/cid/ciab552] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Indexed: 11/13/2022] Open
Abstract
Background In addition to traditional cardiovascular (CV) risk factors, antiretroviral therapy, lifestyle, and human immunodeficiency virus (HIV)-related factors may contribute to future CV events in persons with HIV (PWH). Methods Among participants in the global REPRIEVE randomized trial, we characterized demographics and HIV characteristics relative to ACC/AHA pooled cohort equations (PCE) for atherosclerotic CV disease predicted risk and CV health evaluated by Life’s Simple 7 (LS7; includes smoking, diet, physical activity, body mass index, blood pressure, total cholesterol, and glucose). Results Among 7382 REPRIEVE participants (31% women, 45% Black), the median PCE risk score was 4.5% (lower and upper quartiles Q1, Q3: 2.2, 7.2); 29% had a PCE score <2.5%, and 9% scored above 10%. PCE score was related closely to known CV risk factors and modestly (<1% difference in risk score) to immune function and HIV parameters. The median LS7 score was 9 (Q1, Q3: 7, 10) of a possible 14. Only 24 participants (0.3%) had 7/7 ideal components, and 36% had ≤2 ideal components; 90% had <5 ideal components. The distribution of LS7 did not vary by age or natal sex, although ideal health was more common in low sociodemographic index countries and among Asians. Poor dietary and physical activity patterns on LS7 were seen across all PCE scores, including the lowest risk categories. Conclusions Poor CV health by LS7 was common among REPRIEVE participants, regardless of PCE. This suggests a critical and independent role for lifestyle interventions in conjunction with conventional treatment to improve CV outcomes in PWH. Clinical Trials Registration: NCT02344290. AIDS Clinical Trials Group study number: A5332.
Collapse
Affiliation(s)
- Pamela S Douglas
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| | - Triin Umbleja
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Gerald S Bloomfield
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| | | | | | | | | | | | | | - Judith Currier
- University of California at Los Angeles, Los Angeles, CA
| | | | | | - Anchalee Avihingsanon
- HIV-NAT, Thai Red Cross AIDS Research Centre and TB RU; Faculty of Medicine, Chulalongkorn University, Thailand
| | | | | | - Kiat Ruxrungtham
- HIV-NAT, Thai Red Cross AIDS Research Centre and TB RU; Faculty of Medicine, Chulalongkorn University, Thailand
| | - Maria Saumoy
- Hospital de Bellvitge, l'Hospitalet de Llobregat, Spain
| | | | | | - Heather J Ribaudo
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, MA
| | | | | |
Collapse
|
26
|
Testosterone, HIV, and cardiovascular disease risk. Cardiovasc Endocrinol Metab 2021; 10:72-79. [PMID: 34124602 PMCID: PMC8189608 DOI: 10.1097/xce.0000000000000236] [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: 07/20/2020] [Accepted: 09/04/2020] [Indexed: 11/26/2022]
Abstract
There has been a recent increase in the use of testosterone supplementation among young adults in the United States, despite the controversy of testosterone replacement therapy (TRT) and cardiovascular safety. The lower testosterone levels and earlier age of TRT use in persons living with HIV (PLHIV) is of particular relevance for this population because cardiovascular disease (CVD) comorbidities are known to be increased among PLHIV. There is very limited data on TRT in PLHIV, as such, in this article, we sought to compile current evidence regarding the diagnosis and management of testosterone deficiency and its link to CVD risk including among PLHIV.
Collapse
|
27
|
Siedner MJ, Bibangambah P, Kim J, Lankowski A, Chang JL, Yang IT, Kwon DS, North CM, Triant VA, Longenecker C, Ghoshhajra B, Peck RN, Sentongo RN, Gilbert R, Kakuhikire B, Boum Y, Haberer JE, Martin JN, Tracy R, Hunt PW, Bangsberg DR, Tsai AC, Hemphill LC, Okello S. Treated HIV Infection and Progression of Carotid Atherosclerosis in Rural Uganda: A Prospective Observational Cohort Study. J Am Heart Assoc 2021; 10:e019994. [PMID: 34096320 PMCID: PMC8477876 DOI: 10.1161/jaha.120.019994] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background Although ≈70% of the world's population of people living with HIV reside in sub-Saharan Africa, there are minimal prospective data on the contributions of HIV infection to atherosclerosis in the region. Methods and Results We conducted a prospective observational cohort study of people living with HIV on antiretroviral therapy >40 years of age in rural Uganda, along with population-based comparators not infected with HIV. We collected data on cardiovascular disease risk factors and carotid ultrasound measurements annually. We fitted linear mixed effects models, adjusted for cardiovascular disease risk factors, to estimate the association between HIV serostatus and progression of carotid intima media thickness (cIMT). We enrolled 155 people living with HIV and 154 individuals not infected with HIV and collected cIMT images at 1045 visits during a median of 4 annual visits per participant (interquartile range 3-4, range 1-5). Age (median 50.9 years) and sex (49% female) were similar by HIV serostatus. At enrollment, there was no difference in mean cIMT by HIV serostatus (0.665 versus 0.680 mm, P=0.15). In multivariable models, increasing age, blood pressure, and non-high-density lipoprotein cholesterol were associated with greater cIMT (P<0.05), however change in cIMT per year was also no different by HIV serostatus (0.004 mm/year for HIV negative [95% CI, 0.001-0.007 mm], 0.006 mm/year for people living with HIV [95% CI, 0.003-0.008 mm], HIV×time interaction P=0.25). Conclusions In rural Uganda, treated HIV infection was not associated with faster cIMT progression. These results do not support classification of treated HIV infection as a risk factor for subclinical atherosclerosis progression in rural sub-Saharan Africa. Registration URL: https://www.ClinicalTrials.gov; Unique identifier: NCT02445079.
Collapse
Affiliation(s)
- Mark J. Siedner
- Department of MedicineHarvard Medical SchoolBostonMA,Departments of Medicine and PsychiatryMassachusetts General HospitalBostonMA,Faculty of MedicineMbarara University of Science and TechnologyMbararaUganda
| | - Prossy Bibangambah
- Faculty of MedicineMbarara University of Science and TechnologyMbararaUganda
| | - June‐Ho Kim
- Department of MedicineHarvard Medical SchoolBostonMA,Department of MedicineBrigham and Women's HospitalBostonMA
| | - Alexander Lankowski
- Department of MedicineUniversity of WashingtonSeattleWA,Vaccine and Infectious Disease DivisionFred Hutchinson Cancer Research CenterSeattleWA
| | - Jonathan L. Chang
- Department of MedicineHarvard Medical SchoolBostonMA,Department of MedicineBrigham and Women's HospitalBostonMA
| | - Isabelle T. Yang
- Department of MedicineGeisel School of Medicine at DartmouthHanoverNH
| | - Douglas S. Kwon
- Department of MedicineHarvard Medical SchoolBostonMA,Departments of Medicine and PsychiatryMassachusetts General HospitalBostonMA,Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and HarvardCambridgeMA
| | - Crystal M. North
- Department of MedicineHarvard Medical SchoolBostonMA,Departments of Medicine and PsychiatryMassachusetts General HospitalBostonMA
| | - Virginia A. Triant
- Department of MedicineHarvard Medical SchoolBostonMA,Departments of Medicine and PsychiatryMassachusetts General HospitalBostonMA
| | | | - Brian Ghoshhajra
- Department of MedicineHarvard Medical SchoolBostonMA,Departments of Medicine and PsychiatryMassachusetts General HospitalBostonMA
| | - Robert N. Peck
- Center for Global HealthWeill Cornell Medical CollegeNew YorkNY
| | - Ruth N. Sentongo
- Faculty of MedicineMbarara University of Science and TechnologyMbararaUganda
| | - Rebecca Gilbert
- Departments of Medicine and PsychiatryMassachusetts General HospitalBostonMA
| | - Bernard Kakuhikire
- Faculty of MedicineMbarara University of Science and TechnologyMbararaUganda
| | - Yap Boum
- Epicentre Research BaseMbararaUganda
| | - Jessica E. Haberer
- Department of MedicineHarvard Medical SchoolBostonMA,Departments of Medicine and PsychiatryMassachusetts General HospitalBostonMA
| | | | - Russell Tracy
- Department of Pathology and Laboratory MedicineUniversity of VermontBurlingtonVT
| | - Peter W. Hunt
- Department of MedicineUniversity of CaliforniaSan FranciscoCA
| | | | - Alexander C. Tsai
- Department of MedicineHarvard Medical SchoolBostonMA,Departments of Medicine and PsychiatryMassachusetts General HospitalBostonMA,Faculty of MedicineMbarara University of Science and TechnologyMbararaUganda
| | - Linda C. Hemphill
- Department of MedicineHarvard Medical SchoolBostonMA,Departments of Medicine and PsychiatryMassachusetts General HospitalBostonMA
| | - Samson Okello
- Faculty of MedicineMbarara University of Science and TechnologyMbararaUganda
| |
Collapse
|
28
|
Hoffmann U, Lu MT, Foldyna B, Zanni MV, Karady J, Taron J, Zhai BK, Burdo T, Fitch KV, Kileel EM, Williams K, Fichtenbaum CJ, Overton ET, Malvestutto C, Aberg J, Currier J, Sponseller CA, Melbourne K, Floris-Moore M, Van Dam C, Keefer MC, Koletar SL, Douglas PS, Ribaudo H, Mayrhofer T, Grinspoon SK. Assessment of Coronary Artery Disease With Computed Tomography Angiography and Inflammatory and Immune Activation Biomarkers Among Adults With HIV Eligible for Primary Cardiovascular Prevention. JAMA Netw Open 2021; 4:e2114923. [PMID: 34185068 PMCID: PMC8243232 DOI: 10.1001/jamanetworkopen.2021.14923] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
IMPORTANCE Cardiovascular disease (CVD) is increased among people with HIV (PWH), but little is known regarding the prevalence and extent of coronary artery disease (CAD) and associated biological factors in PWH with low to moderate traditional CVD risk. OBJECTIVES To determine unique factors associated with CVD in PWH and to assess CAD by coronary computed tomography angiography (CTA) and critical pathways of arterial inflammation and immune activation. DESIGN, SETTING, AND PARTICIPANTS This cohort study among male and female PWH, aged 40 to 75 years, without known CVD, receiving stable antiretroviral therapy, and with low to moderate atherosclerotic cardiovascular disease (ASCVD) risk according to the 2013 American College of Cardiology/American Heart Association pooled cohort equation, was part of the Randomized Trial to Prevent Vascular Events in HIV (REPRIEVE), a large, ongoing primary prevention trial of statin therapy among PWH conducted at 31 US sites. Participants were enrolled from May 2015 to February 2018. Data analysis was conducted from May to December 2020. EXPOSURE HIV disease. MAIN OUTCOMES AND MEASURES The primary outcome was the prevalence and composition of CAD assessed by coronary CTA and, secondarily, the association of CAD with traditional risk indices and circulating biomarkers, including insulin, monocyte chemoattractant protein 1 (MCP-1), interleukin (IL) 6, soluble CD14 (sCD14), sCD163, lipoprotein-associated phospholipase A2 (LpPLA2), oxidized low-density lipoprotein (oxLDL), and high-sensitivity C-reactive protein (hsCRP). RESULTS The sample included 755 participants, with a mean (SD) age of 51 (6) years, 124 (16%) female participants, 267 (35%) Black or African American participants, 182 (24%) Latinx participants, a low median (interquartile range) ASCVD risk (4.5% [2.6%-6.8%]), and well-controlled viremia. Overall, plaque was seen in 368 participants (49%), including among 52 of 175 participants (30%) with atherosclerotic CVD (ASCVD) risk of less than 2.5%. Luminal obstruction of at least 50% was rare (25 [3%]), but vulnerable plaque and high Leaman score (ie, >5) were more frequently observed (172 of 755 [23%] and 118 of 743 [16%], respectively). Overall, 251 of 718 participants (35%) demonstrated coronary artery calcium score scores greater than 0. IL-6, LpPLA2, oxLDL, and MCP-1 levels were higher in those with plaque compared with those without (eg, median [IQR] IL-6 level, 1.71 [1.05-3.04] pg/mL vs 1.45 [0.96-2.60] pg/mL; P = .008). LpPLA2 and IL-6 levels were associated with plaque in adjusted modeling, independent of traditional risk indices and HIV parameters (eg, IL-6: adjusted odds ratio, 1.07; 95% CI, 1.02-1.12; P = .01). CONCLUSIONS AND RELEVANCE In this study of a large primary prevention cohort of individuals with well-controlled HIV and low to moderate ASCVD risk, CAD, including noncalcified, nonobstructive, and vulnerable plaque, was highly prevalent. Participants with plaque demonstrated higher levels of immune activation and arterial inflammation, independent of traditional ASCVD risk and HIV parameters.
Collapse
Affiliation(s)
| | | | | | | | - Julia Karady
- Massachusetts General Hospital, Boston
- MTA-SE Cardiovascular Imaging Research Group, Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Jana Taron
- Massachusetts General Hospital, Boston
- University Hospital Freiburg, Freiburg, Germany
| | - Bingxue K. Zhai
- Massachusetts General Hospital, Boston
- Brigham and Women’s Hospital, Boston, Massachusetts
| | | | | | | | | | | | | | | | - Judith Aberg
- Icahn School of Medicine at Mount Sinai, New York, New York
| | | | | | | | | | - Cornelius Van Dam
- Greensboro Clinical Research Site, Cone Health, Greensboro, North Carolina
| | - Michael C. Keefer
- University of Rochester Adult HIV Therapeutic Strategies Network Clinical Research Site, Rochester, New York
| | | | - Pamela S. Douglas
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, North Carolina
| | - Heather Ribaudo
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts
| | - Thomas Mayrhofer
- Massachusetts General Hospital, Boston
- School of Business Studies, Stralsund University of Applied Sciences, Stralsund, Germany
| | | |
Collapse
|
29
|
Grinspoon SK, Douglas PS, Hoffmann U, Ribaudo HJ. Leveraging a Landmark Trial of Primary Cardiovascular Disease Prevention in Human Immunodeficiency Virus: Introduction From the REPRIEVE Coprincipal Investigators. J Infect Dis 2021; 222:S1-S7. [PMID: 32645161 DOI: 10.1093/infdis/jiaa098] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
The Randomized Trial to Prevent Vascular Events in HIV (REPRIEVE) is the largest study of cardiovascular disease in human immunodeficiency virus. Enrolling 7770 participants from 2015 to 2019 with sites across 5 continents, REPRIEVE will assess the effects of a statin as a cardiovascular disease prevention strategy in people with HIV (PWH) receiving antiretroviral therapy (ART). Although the primary purpose of REPRIEVE, and its substudy assessing coronary plaque, is to assess cardiovascular outcomes, the trial is a rich source of data on population characteristics and critical comorbidities in PWH, particularly across Global Burden of Disease (GBD) regions, reflective of the ethnic, racial, and gender diversity in this global epidemic. The purpose of this Supplement is to leverage the rich phenotyping in REPRIEVE, to provide data on detailed patterns of baseline ART and immune function by GBD region, reproductive aging among cisgender women, and data on the participation and clinical characteristics of transgender participants. We also leveraged REPRIEVE to assess critical comorbidities, including renal dysfunction, muscle function and frailty, and myocardial steatosis. REPRIEVE is a remarkable collaboration between funders, trial networks, clinical research sites, clinical and data coordinating centers, and willing participants who devoted their time to make the trial possible.
Collapse
Affiliation(s)
- Steven K Grinspoon
- Metabolism Unit, Massachusetts General Hospital, and Harvard Medical School, Boston, Massachusetts, USA
| | - Pamela S Douglas
- Division of Cardiology and Duke Clinical Research Center, Duke University School of Medicine, Durham, North Carolina, USA
| | - Udo Hoffmann
- Cardiovascular Imaging Research Center, Department of Radiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Heather J Ribaudo
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
30
|
deFilippi C, Toribio M, Wong LP, Sadreyev R, Grundberg I, Fitch KV, Zanni MV, Lo J, Sponseller CA, Sprecher E, Rashidi N, Thompson MA, Cagliero D, Aberg JA, Braun LR, Stanley TL, Lee H, Grinspoon SK. Differential Plasma Protein Regulation and Statin Effects in Human Immunodeficiency Virus (HIV)-Infected and Non-HIV-Infected Patients Utilizing a Proteomics Approach. J Infect Dis 2021; 222:929-939. [PMID: 32310273 DOI: 10.1093/infdis/jiaa196] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 04/16/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND People with human immunodeficiency virus (PWH) demonstrate increased atherosclerotic cardiovascular disease (ASCVD). Statins are being studied to prevent ASCVD in human immunodeficiency virus (HIV), but little is known regarding the effects of statins on a broad range of inflammatory and cardiovascular proteins in this population. METHODS We used a highly specific discovery proteomic approach (Protein Extension Assay), to determine statin effects on over 350 plasma proteins in relevant ASCVD pathways among HIV and non-HIV groups. Responses to pitavastatin calcium were assessed in 89 PWH in the INTREPID trial and 46 non-HIV participants with features of central adiposity and insulin resistance. History of cardiovascular disease was exclusionary for both studies. RESULTS Among participants with HIV, PCOLCE (enzymatic cleavage of type I procollagen) significantly increased after pitavastatin therapy and PLA2G7 (systemic marker of arterial inflammation) decreased. Among participants without HIV, integrin subunit alpha M (integrin adhesive function) and defensin alpha-1 (neutrophil function) increased after pitavastatin therapy and PLA2G7 decreased. At baseline, comparing participants with and without HIV, differentially expressed proteins included proteins involved in platelet and endothelial function and immune activation. CONCLUSIONS Pitavastatin affected proteins important to platelet and endothelial function and immune activation, and effects differed to a degree within PWH and participants without HIV.
Collapse
Affiliation(s)
- Chris deFilippi
- Inova Heart and Vascular Institute, Falls Church, Virginia, USA
| | - Mabel Toribio
- Massachusetts General Hospital, Metabolism Unit and Harvard Medical School, Boston, Massachusetts, USA
| | - Lai Ping Wong
- Massachusetts General Hospital, Department of Molecular Biology and Harvard Medical School, Boston, Massachusetts, USA
| | - Ruslan Sadreyev
- Massachusetts General Hospital, Department of Molecular Biology and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Kathleen V Fitch
- Massachusetts General Hospital, Metabolism Unit and Harvard Medical School, Boston, Massachusetts, USA
| | - Markella V Zanni
- Massachusetts General Hospital, Metabolism Unit and Harvard Medical School, Boston, Massachusetts, USA
| | - Janet Lo
- Massachusetts General Hospital, Metabolism Unit and Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | - Diana Cagliero
- Massachusetts General Hospital, Metabolism Unit and Harvard Medical School, Boston, Massachusetts, USA
| | - Judith A Aberg
- Mount Sinai Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Laurie R Braun
- Massachusetts General Hospital, Metabolism Unit and Harvard Medical School, Boston, Massachusetts, USA
| | - Takara L Stanley
- Massachusetts General Hospital, Metabolism Unit and Harvard Medical School, Boston, Massachusetts, USA
| | - Hang Lee
- Massachusetts General Hospital, Biostatistics Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Steven K Grinspoon
- Massachusetts General Hospital, Metabolism Unit and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
31
|
Liu Y, Zhang HG. Vigilance on New-Onset Atherosclerosis Following SARS-CoV-2 Infection. Front Med (Lausanne) 2021; 7:629413. [PMID: 33553222 PMCID: PMC7855580 DOI: 10.3389/fmed.2020.629413] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 12/23/2020] [Indexed: 01/08/2023] Open
Abstract
The pandemic of coronavirus disease 2019 (COVID-19), caused by SARS-CoV-2, has become a global challenge to public health. While its typical clinical manifestations are respiratory disorders, emerging evidence of cardiovascular complications indicates the adverse interaction between SARS-CoV-2 infection and cardiovascular outcomes. Given that viral infection has emerged as an additional risk factor for atherosclerosis, in this paper, we attempt to clarify the susceptibility to new-onset atherosclerosis in individuals infected with SARS-CoV-2. Mechanistically, serving as functional receptors for SARS-CoV-2, angiotensin-converting enzyme 2 (ACE2) mediates SARS-CoV-2 infection of endothelial cells (ECs) directly, leading to endothelial dysfunction and dysregulation of the renin-angiotensin system (RAS). In addition, high expression of CD147, an alternative receptor, and activation of the NLRP3 inflammasome may also contribute to atherosclerosis in the context of COVID-19. More importantly, SARS-CoV-2 attacks the immune system, which results in excessive inflammation and perpetuates a vicious cycle of deteriorated endothelial dysfunction that further promotes inflammation. The alterations in the blood lipid profile induced by COVID-19 should not be ignored in assessing the predisposition toward atherosclerosis in victims of COVID-19. A better understanding of the underlying mechanisms of SARS-CoV-2 infection and the long-term monitoring of inflammatory factors and endothelial function should be considered in the follow-up of patients who have recovered from COVID-19 for early detection and prevention of atherosclerosis.
Collapse
Affiliation(s)
| | - Hai-Gang Zhang
- Department of Pharmacology, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
32
|
Kettelhut A, Bowman E, Funderburg NT. Immunomodulatory and Anti-Inflammatory Strategies to Reduce Comorbidity Risk in People with HIV. Curr HIV/AIDS Rep 2020; 17:394-404. [PMID: 32535769 DOI: 10.1007/s11904-020-00509-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW In this review, we will discuss treatment interventions targeting drivers of immune activation and chronic inflammation in PWH. RECENT FINDINGS Potential treatment strategies to prevent the progression of comorbidities in PWH have been identified. These studies include, among others, the use of statins to modulate lipid alterations and subsequent innate immune receptor activation, probiotics to restore healthy gut microbiota and reduce microbial translocation, hydroxychloroquine to reduce immune activation by altering Toll-like receptors function and expression, and canakinumab to block the action of a major pro-inflammatory cytokine IL-1β. Although many of the treatment strategies discussed here show promise, due to the complex nature of chronic inflammation and comorbidities in PWH, larger clinical studies are needed to understand and target the prominent drivers and inflammatory cascades underlying these end-organ diseases.
Collapse
Affiliation(s)
- Aaren Kettelhut
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, Ohio State University College of Medicine, Columbus, OH, USA
| | - Emily Bowman
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, Ohio State University College of Medicine, Columbus, OH, USA
| | - Nicholas T Funderburg
- Division of Medical Laboratory Science, School of Health and Rehabilitation Sciences, Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
33
|
Rerkasem A, Pongtam S, Ounjaijean S, Kulprachakarn K, Wongthanee A, Chaiwarith R, Supparatpinyo K, Salee P, Arworn S, Rerkasem K. Ankle-Brachial Index and Carotid Intima-Media Thickness Progression by Using Ultrasound Among Patients With HIV Infection Versus End-Stage Renal Disease. INT J LOW EXTR WOUND 2020; 19:364-368. [DOI: 10.1177/1534734620971067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Human immunodeficiency virus (HIV) and end-stage renal disease (ESRD) patients contributed to accelerated cardiovascular disease. Comparing the effect on atherosclerosis of the 2 diseases has never been explored. A prospective cohort study enrolled participants who were more than 18 years of age without stroke, coronary, and peripheral arterial disease events. Each HIV-infected person had continuously used antiretroviral therapy and ESRD and required intermittent hemodialysis. We assessed patients using the ankle-brachial index (ABI) and carotid intimal media thickness (CIMT) at enrollment, and 1 year later. The main outcome was the progression of ABI and CIMT per year. Demographic, comorbidities, and serum profiles were collected on entry. A total of 789 HIV-positive and 41 ESRD with HIV-negative patients were recruited. After adjusting for potential confounders at baseline, the ESRD die not significantly decrease ABI by 0.015 in 1 year (P=0 .252). The HIV-infected group had a significantly decreased ABI by 0.020 in 1 year (P < .001), but the reduced rate in the HIV-infected group was not statistically different from those in the ESRD group (P = 0.901). When adjusted for potential confounders, the ESRD had a significant increase of CIMT by 0.111 mm in 1 year (P<0.001). The HIV patients had a significant increase of 0.250 mm CIMT in 1 year (P<0.001). This progression rate was statistically greater in the HIV-infected group versus the ESRD group. HIV infection and ESRD had comparable rates of ABI and CIMT progression in our study. Then, early prevention in asymptomatic atherosclerosis should include not only patients with ESRD but also HIV-infected patients.
Collapse
Affiliation(s)
- Amaraporn Rerkasem
- NCD Center of Excellence, Research Institute for Health Sciences, Chiang Mai University, Thailand
| | - Sasinat Pongtam
- NCD Center of Excellence, Research Institute for Health Sciences, Chiang Mai University, Thailand
| | - Sakaewan Ounjaijean
- NCD Center of Excellence, Research Institute for Health Sciences, Chiang Mai University, Thailand
| | - Kanokwan Kulprachakarn
- NCD Center of Excellence, Research Institute for Health Sciences, Chiang Mai University, Thailand
| | - Antika Wongthanee
- Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Maharaj Nakorn Chiang Mai Hospital, Chiang Mai, Thailand
| | - Romanee Chaiwarith
- Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Maharaj Nakorn Chiang Mai Hospital, Chiang Mai, Thailand
| | - Khuanchai Supparatpinyo
- NCD Center of Excellence, Research Institute for Health Sciences, Chiang Mai University, Thailand
- Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Maharaj Nakorn Chiang Mai Hospital, Chiang Mai, Thailand
| | - Parichat Salee
- Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Maharaj Nakorn Chiang Mai Hospital, Chiang Mai, Thailand
| | - Supapong Arworn
- Department of Surgery, Faculty of Medicine, Chiang Mai University, Maharaj Nakorn Chiang Mai Hospital, Chiang Mai, Thailand
| | - Kittipan Rerkasem
- NCD Center of Excellence, Research Institute for Health Sciences, Chiang Mai University, Thailand
- Department of Surgery, Faculty of Medicine, Chiang Mai University, Maharaj Nakorn Chiang Mai Hospital, Chiang Mai, Thailand
| |
Collapse
|
34
|
Bloch M, John M, Smith D, Rasmussen TA, Wright E. Managing HIV-associated inflammation and ageing in the era of modern ART. HIV Med 2020; 21 Suppl 3:2-16. [PMID: 33022087 DOI: 10.1111/hiv.12952] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVES This paper aims to address the concerns around ongoing immune activation, inflammation, and resistance in those ageing with HIV that represent current challenges for clinicians. METHODS Presentations at a symposium addressing issues of ageing with HIV infection were reviewed and synthesised. RESULTS The changing natural history and demographics of human immunodeficiency virus (HIV)-infected individuals means new challenges in contemporary management. In the early years of the epidemic,management was focussed on acute, potentially life-threatening AIDS-related complications. From initial monotherapy with first-generation antiretroviral therapy (ART), the development of combination highly active ART (HAART) allowed HIV control but ART toxicities, treatment adherence and drug resistance emerged as major issues. Today, the availability of potent and tolerable ART has made viral suppression achievable in most people living with HIV (PLHIV), and clinicians are confronted with managing a chronic condition among an ageing population. The combination of diseases of ageing and the co-morbidities associated with HIV-infection, even when well controlled, results in a complex set of challenges for many older PLHIV. There is a growing appreciation that many non-AIDS-related co-morbidities are caused, at least in part, by persistent, low-grade immune activation, inflammation, and hypercoagulability, despite suppressive ART. CONCLUSIONS In order to further improve HIV management, it is important to understand the enduring effects of chronically suppressed HIV infection, the potential contribution of these factors to the ageing process, the possibility of drug resistance, and the impact of different treatment strategies, including early ART initiation.
Collapse
Affiliation(s)
- M Bloch
- Holdsworth House Medical Practice, Sydney, NSW, Australia.,Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - M John
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia.,Royal Perth Hospital, Perth, WA, Australia.,Institute of Immunology and Infectious Disease, Perth, WA, Australia
| | - D Smith
- School of Public Health and Community Medicine, University of New South Wales, Sydney, NSW, Australia.,The Albion Centre, Sydney, NSW, Australia
| | - T A Rasmussen
- Doherty Institute for Infection and Immunity, Melbourne, Vic., Australia.,University of Melbourne, Melbourne, Vic., Australia
| | - E Wright
- The Alfred Hospital, Melbourne, Vic., Australia.,Centre for Inflammatory Diseases, Monash University, Melbourne, Vic., Australia.,The Burnett Institute, Melbourne, Vic., Australia
| |
Collapse
|
35
|
Implementation of Cholesterol-Lowering Therapy to Reduce Cardiovascular Risk in Persons Living with HIV. Cardiovasc Drugs Ther 2020; 36:173-186. [PMID: 32979175 DOI: 10.1007/s10557-020-07085-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/17/2020] [Indexed: 10/23/2022]
Abstract
The widespread availability of highly effective antiretroviral therapies has reduced mortality from opportunistic infections in persons living with HIV (PLHIV), resulting in an increase in atherosclerotic cardiovascular disease (ASCVD) and other chronic illnesses (Samji et al. 2013). Although there has been a decline in morbidity and mortality from ASCVD in the past several decades, contemporary studies continue to report higher rates of cardiovascular events (Rosenson et al. 2020). HIV has been identified as a risk enhancer for ASCVD by multiple professional guideline writing committees (Grundy Scott et al. 2019, Mach et al. 2020); however, the utilization of cholesterol-lowering therapies in PLHIV remains low (Rosenson et al. 2018). Moreover, the use of statin therapy in PLHIV is complicated by drug-drug interactions that may either elevate or lower the blood statin concentrations resulting in increased toxicity or reduced efficacy respectively. Other comorbidities commonly associated with HIV present other challenges for the use of cholesterol-lowering therapies. This review will summarize the data on lipoprotein-associated ASCVD risk in PLHIV and discuss the challenges with effective treatment. Finally, we present a clinical algorithm to optimize cardiovascular risk reduction in this high-risk population.
Collapse
|
36
|
Abdelrahman KM, Chen MY, Dey AK, Virmani R, Finn AV, Khamis RY, Choi AD, Min JK, Williams MC, Buckler AJ, Taylor CA, Rogers C, Samady H, Antoniades C, Shaw LJ, Budoff MJ, Hoffmann U, Blankstein R, Narula J, Mehta NN. Coronary Computed Tomography Angiography From Clinical Uses to Emerging Technologies: JACC State-of-the-Art Review. J Am Coll Cardiol 2020; 76:1226-1243. [PMID: 32883417 PMCID: PMC7480405 DOI: 10.1016/j.jacc.2020.06.076] [Citation(s) in RCA: 152] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 05/08/2020] [Accepted: 06/10/2020] [Indexed: 12/14/2022]
Abstract
Evaluation of coronary artery disease (CAD) using coronary computed tomography angiography (CCTA) has seen a paradigm shift in the last decade. Evidence increasingly supports the clinical utility of CCTA across various stages of CAD, from the detection of early subclinical disease to the assessment of acute chest pain. Additionally, CCTA can be used to noninvasively quantify plaque burden and identify high-risk plaque, aiding in diagnosis, prognosis, and treatment. This is especially important in the evaluation of CAD in immune-driven conditions with increased cardiovascular disease prevalence. Emerging applications of CCTA based on hemodynamic indices and plaque characterization may provide personalized risk assessment, affect disease detection, and further guide therapy. This review provides an update on the evidence, clinical applications, and emerging technologies surrounding CCTA as highlighted at the 2019 National Heart, Lung and Blood Institute CCTA Summit.
Collapse
Affiliation(s)
- Khaled M Abdelrahman
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Marcus Y Chen
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Amit K Dey
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Renu Virmani
- Department of Pathology, CVPath Institute, Gaithersburg, Maryland
| | - Aloke V Finn
- Department of Pathology, CVPath Institute, Gaithersburg, Maryland
| | - Ramzi Y Khamis
- Vascular Sciences Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Andrew D Choi
- Division of Cardiology and Department of Radiology, The George Washington University School of Medicine, Washington, DC
| | - James K Min
- Department of Radiology, New York-Presbyterian Hospital and Weill Cornell Medicine, New York, New York
| | - Michelle C Williams
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom; Edinburgh Imaging, Queen's Medical Research Institute University of Edinburgh, Edinburgh, United Kingdom
| | | | | | | | - Habib Samady
- Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia
| | - Charalambos Antoniades
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Leslee J Shaw
- Department of Radiology, New York-Presbyterian Hospital and Weill Cornell Medicine, New York, New York
| | - Matthew J Budoff
- Lundquist Institute at Harbor-UCLA Medical Center, Torrance, California
| | - Udo Hoffmann
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ron Blankstein
- Departments of Medicine (Cardiovascular Division) and Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jagat Narula
- Zena and Michael A. Wiener Cardiovascular Institute, Marie-Josée and Henry R. Kravis Center for Cardiovascular Health Icahn School of Medicine at Mount Sinai, Mount Sinai Heart, New York, New York
| | - Nehal N Mehta
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|
37
|
Guaraldi G, Milic J, Prandini N, Ligabue G, Esposito F, Ciusa G, Malagoli A, Scaglioni R, Besutti G, Beghetto B, Nardini G, Roncaglia E, Mussini C, Raggi P. 18Fluoride-based molecular imaging of coronary atherosclerosis in HIV infected patients. Atherosclerosis 2020; 297:127-135. [PMID: 32113050 DOI: 10.1016/j.atherosclerosis.2020.02.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 01/18/2020] [Accepted: 02/14/2020] [Indexed: 11/25/2022]
Abstract
BACKGROUND AND AIMS Molecular imaging with 18Fluorodeoxyglucose (FDG) and 18F-sodium-fluoride (NaF) captures arterial inflammation and micro-calcification and can reveal potentially unstable atherosclerotic plaques. METHODS We performed FDG and NaF PET/CT imaging in two clinically similar cohorts of patients living with HIV (PLWH) with no symptomatic cardiovascular disease. The prevalence and intensity of coronary artery uptake of each tracer, measured as target-to-background ratio (TBR), were assessed in patients at low and high cardiovascular risk. RESULTS Ninety-three PLWH were submitted to PET/CT imaging with FDG (N = 43) and NaF (N = 50); 42% were at low and 58% at high cardiovascular risk. The intensity of uptake and multivessel coronary artery uptake were significantly higher with NaF than FDG both in low and high-risk patients. When each 18F-tracer was tested in low and high-risk patients, an equal proportion of subjects showed no vessel, single and multivessel NaF uptake; the same was true for no and single vessel uptake of FDG (no multivessel FDG uptake was noted). Waist circumference, CRP, D-dimer, HIV duration and treatment with nucleoside reverse transcriptase inhibitors were associated with high NaF uptake in univariable analyses; D-dimer remained significant in multivariable analyses (OR = 1.05; p=0.02). There were no significant associations with FDG uptake. CONCLUSIONS The prevalence of coronary artery uptake was higher with NaF compared to FDG both in high and low risk patients, hence microcalcification imaging may be a more sensitive tool to detect coronary atherosclerosis than inflammation imaging. However, the uptake of each 18Fluoride tracer was similar between low and high-risk subjects, and this underscores the discordance between clinical and imaging based risk assessment. Future investigation should address the prognostic significance of NaF coronary artery uptake.
Collapse
Affiliation(s)
- Giovanni Guaraldi
- Modena HIV Metabolic Clinic, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41124, Modena, Italy
| | - Jovana Milic
- Modena HIV Metabolic Clinic, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41124, Modena, Italy; Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, 41124, Modena, Italy
| | - Napoleone Prandini
- Department of Nuclear Medicine, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41124, Modena, Italy
| | - Guido Ligabue
- Department of Radiology, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41124, Modena, Italy
| | - Francesco Esposito
- Modena HIV Metabolic Clinic, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41124, Modena, Italy
| | - Giacomo Ciusa
- Modena HIV Metabolic Clinic, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41124, Modena, Italy
| | - Andrea Malagoli
- Modena HIV Metabolic Clinic, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41124, Modena, Italy
| | - Riccardo Scaglioni
- Department of Radiology, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41124, Modena, Italy
| | - Giulia Besutti
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, 41124, Modena, Italy; Department of Radiology, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41124, Modena, Italy
| | - Barbara Beghetto
- Modena HIV Metabolic Clinic, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41124, Modena, Italy
| | - Giulia Nardini
- Modena HIV Metabolic Clinic, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41124, Modena, Italy
| | - Enrica Roncaglia
- Modena HIV Metabolic Clinic, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41124, Modena, Italy
| | - Cristina Mussini
- Modena HIV Metabolic Clinic, Azienda Ospedaliero-Universitaria di Modena, University of Modena and Reggio Emilia, 41124, Modena, Italy
| | - Paolo Raggi
- Division of Cardiology and Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, 11220, 83rd Avenue, Suite 5A9-014, Edmonton, AB T6G 2B7, Canada.
| |
Collapse
|
38
|
HIV-associated Extracranial Arterial Aneurysms: A Systematic Review. AMERICAN JOURNAL OF MEDICAL CASE REPORTS 2020; 8:128-133. [PMID: 32432158 PMCID: PMC7236995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Human immunodeficiency virus/acquired immunodeficiency syndrome (HIV/AIDS) has been found to be associated with an increased risk of cardiovascular disease, and the development of arterial aneurysms in particular, intracranial aneurysms. In this review, we will review the reported HIV-associated extracranial aneurysms (HECAA) and their possible association with HIV/AIDS. We will discuss the proposed pathogenetic pathways leading to arterial aneurysms. HECAA, a subset of HIV/AIDS-associated arterial aneurysm (HAA), is more commonly seen in the adult population and in those with lower CD4+ T-cell counts and higher HIV viral loads. There also appears to be an advantage to early diagnosis of HECAA. There are viable treatment options available, as 61.4% of patients with HECAA underwent a corrective procedure. Furthermore, the mortality rate of 1.75% in HECAA was much lower when compared to HICAA.
Collapse
|
39
|
Abstract
Antiretroviral therapy has largely transformed HIV infection into a chronic disease condition. As such, physicians and other providers caring for individuals living with HIV infection need to be aware of the potential cardiovascular complications of HIV infection and the nuances of how HIV infection increases the risk of cardiovascular diseases, including acute myocardial infarction, stroke, peripheral artery disease, heart failure and sudden cardiac death, as well as how to select available therapies to reduce this risk. In this Review, we discuss the epidemiology and clinical features of cardiovascular disease, with a focus on coronary heart disease, in the setting of HIV infection, which includes a substantially increased risk of myocardial infarction even when the HIV infection is well controlled. We also discuss the mechanisms underlying HIV-associated atherosclerotic cardiovascular disease, such as the high rates of traditional cardiovascular risk factors in patients with HIV infection and HIV-related factors, including the use of antiretroviral therapy and chronic inflammation in the setting of effectively treated HIV infection. Finally, we highlight available therapeutic strategies, as well as approaches under investigation, to reduce the risk of cardiovascular disease and lower inflammation in patients with HIV infection.
Collapse
Affiliation(s)
- Priscilla Y Hsue
- University of California-San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, CA, USA.
| | - David D Waters
- University of California-San Francisco, Zuckerberg San Francisco General Hospital, San Francisco, CA, USA
| |
Collapse
|
40
|
Thompson-Paul AM, Palella FJ, Rayeed N, Ritchey MD, Lichtenstein KA, Patel D, Yang Q, Gillespie C, Loustalot F, Patel P, Buchacz K. Excess heart age in adult outpatients in routine HIV care. AIDS 2019; 33:1935-1942. [PMID: 31274539 PMCID: PMC8428771 DOI: 10.1097/qad.0000000000002304] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Cardiovascular disease (CVD) is a common cause of morbidity and mortality among persons living with HIV (PLWH). We used individual cardiovascular risk factor profiles to estimate heart age for PLWH in medical care in the United States. DESIGN Cross-sectional analyses of HIV Outpatient Study (HOPS) data METHODS:: Included in this analysis were participants aged 30-74 years, without prior CVD, with at least two HOPS clinic visits during 2010-2017, at least 1-year of follow-up, and available covariate data. We calculated age and race/ethnicity-adjusted heart age and excess heart age (chronological age - heart age), using a Framingham risk score-based model. RESULTS We analyzed data from 2467 men and 619 women (mean chronologic age 49.3 and 49.1 years, and 23.6% and 54.6% Non-Hispanic/Latino black, respectively). Adjusted excess heart age was 11.5 years (95% confidence interval, 11.1-12.0) among men and 13.1 years (12.0-14.1) among women. Excess heart age was seen among all age groups beginning with persons aged 30-39 years [men, 7.8 (6.9-8.8); women, 7.7 (4.9-10.4)], with the highest excess heart age among participants aged 50-59 years [men, 13.7 years (13.0-14.4); women, 16.4 years (14.8-18.0)]. More than 50% of participants had an excess heart age of at least 10 years. CONCLUSIONS Excess heart age is common among PLWH, begins in early adulthood, and impacts both women and men. Among PLWH, CVD risk factors should be addressed early and proactively. Routine use of the heart age calculator may help optimize CVD risk stratification and facilitate interventions for aging PLWH.
Collapse
Affiliation(s)
- Angela M. Thompson-Paul
- Division for Heart Disease and Stroke Prevention, National Center for Chronic Disease Prevention and Health Promotion, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Frank J. Palella
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | | | - Matthew D. Ritchey
- Division for Heart Disease and Stroke Prevention, National Center for Chronic Disease Prevention and Health Promotion, Centers for Disease Control and Prevention, Atlanta, Georgia
| | | | - Deesha Patel
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, & TB Prevention, Centers for Disease Control and Prevention, Atlanta
| | - Quanhe Yang
- Division for Heart Disease and Stroke Prevention, National Center for Chronic Disease Prevention and Health Promotion, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Cathleen Gillespie
- Division for Heart Disease and Stroke Prevention, National Center for Chronic Disease Prevention and Health Promotion, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Fleetwood Loustalot
- Division for Heart Disease and Stroke Prevention, National Center for Chronic Disease Prevention and Health Promotion, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Pragna Patel
- Division of Global Health Protection, Center for Global Health, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Kate Buchacz
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, & TB Prevention, Centers for Disease Control and Prevention, Atlanta
| | | |
Collapse
|
41
|
Conceptualizing the Risks of Coronary Heart Disease and Heart Failure Among People Aging with HIV: Sex-Specific Considerations. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2019; 21:41. [DOI: 10.1007/s11936-019-0744-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
42
|
Feinstein MJ, Hsue PY, Benjamin L, Bloomfield GS, Currier JS, Freiberg MS, Grinspoon SK, Levin J, Longenecker CT, Post. WS. Characteristics, Prevention, and Management of Cardiovascular Disease in People Living With HIV: A Scientific Statement From the American Heart Association. Circulation 2019; 140:e98-e124. [PMID: 31154814 PMCID: PMC7993364 DOI: 10.1161/cir.0000000000000695] [Citation(s) in RCA: 393] [Impact Index Per Article: 78.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
As early and effective antiretroviral therapy has become more widespread, HIV has transitioned from a progressive, fatal disease to a chronic, manageable disease marked by elevated risk of chronic comorbid diseases, including cardiovascular diseases (CVDs). Rates of myocardial infarction, heart failure, stroke, and other CVD manifestations, including pulmonary hypertension and sudden cardiac death, are significantly higher for people living with HIV than for uninfected control subjects, even in the setting of HIV viral suppression with effective antiretroviral therapy. These elevated risks generally persist after demographic and clinical risk factors are accounted for and may be partly attributed to chronic inflammation and immune dysregulation. Data on long-term CVD outcomes in HIV are limited by the relatively recent epidemiological transition of HIV to a chronic disease. Therefore, our understanding of CVD pathogenesis, prevention, and treatment in HIV relies on large observational studies, randomized controlled trials of HIV therapies that are underpowered to detect CVD end points, and small interventional studies examining surrogate CVD end points. The purpose of this document is to provide a thorough review of the existing evidence on HIV-associated CVD, in particular atherosclerotic CVD (including myocardial infarction and stroke) and heart failure, as well as pragmatic recommendations on how to approach CVD prevention and treatment in HIV in the absence of large-scale randomized controlled trial data. This statement is intended for clinicians caring for people with HIV, individuals living with HIV, and clinical and translational researchers interested in HIV-associated CVD.
Collapse
Affiliation(s)
| | - Priscilla Y. Hsue
- University of California-San Francisco School of Medicine, San Francisco, CA
| | | | | | - Judith S. Currier
- University of California-Los Angeles School of Medicine, Los Angeles, CA
| | | | | | - Jules Levin
- National AIDS Treatment Advocacy Program, New York, NY
| | | | - Wendy S. Post.
- Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
43
|
Grinspoon SK, Fitch KV, Overton ET, Fichtenbaum CJ, Zanni MV, Aberg JA, Malvestutto C, Lu MT, Currier JS, Sponseller CA, Waclawiw M, Alston-Smith B, Cooper-Arnold K, Klingman KL, Desvigne-Nickens P, Hoffmann U, Ribaudo HJ, Douglas PS. Rationale and design of the Randomized Trial to Prevent Vascular Events in HIV (REPRIEVE). Am Heart J 2019; 212:23-35. [PMID: 30928825 PMCID: PMC6535121 DOI: 10.1016/j.ahj.2018.12.016] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 12/15/2018] [Indexed: 01/14/2023]
Abstract
BACKGROUND Cardiovascular disease (CVD) is more frequent among people with HIV (PWH) and may relate to traditional and nontraditional factors, including inflammation and immune activation. A critical need exists to develop effective strategies to prevent CVD in this population. METHODS The Randomized Trial to Prevent Vascular Events in HIV (REPRIEVE) (A5332) is a prospective, randomized, placebo-controlled trial of a statin strategy for the primary prevention of major adverse cardiovascular events (MACE) in PWH with low to moderate traditional risk. At least 7,500 PWH, 40-75 years of age, on stable antiretroviral therapy, will be randomized to pitavastatin calcium (4 mg/d) or identical placebo and followed for up to 8 years. Participants are enrolled based on the 2013 American College of Cardiology (ACC)/American Heart Association (AHA) atherosclerotic cardiovascular disease (ASCVD) risk score and low-density lipoprotein cholesterol (LDL-C) level with a goal to identify a low- to moderate-risk population who might benefit from a pharmacologic CVD prevention strategy. Potential participants with a risk score ≤ 15% were eligible based on decreasing LDL-C thresholds for increasing risk score >7.5% (LDL-C <190 mg/dL for risk score <7.5%, LDL-C <160 mg/dL for risk score 7.6%-10%, and LDL-C<130 mg/dL for risk score 10.1%-15%). The primary objective is to determine effects on a composite end point of MACE. Formal and independent adjudication of clinical events will occur using standardized criteria. Key secondary end points include effects on MACE components, all-cause mortality, specified non-CVD events, AIDS and non-AIDS events, and safety. RESULTS To date, REPRIEVE has enrolled >7,500 participants at approximately 120 sites across 11 countries, generating a diverse and representative population of PWH to investigate the primary objective of the trial. CONCLUSIONS REPRIEVE is the first trial investigating a primary CVD prevention strategy in PWH. REPRIEVE will inform the field of the efficacy and safety of a statin strategy among HIV-infected participants on antiretroviral therapy and provide critical information on CVD mechanisms and non-CVD events in PWH.
Collapse
Affiliation(s)
- Steven K Grinspoon
- MGH Program in Nutritional Metabolism and Harvard Medical School, Boston, MA.
| | - Kathleen V Fitch
- MGH Program in Nutritional Metabolism and Harvard Medical School, Boston, MA
| | - Edgar Turner Overton
- Division of Infectious Diseases, University of Alabama at Birmingham School of Medicine, Birmingham, AL
| | - Carl J Fichtenbaum
- Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Markella V Zanni
- MGH Program in Nutritional Metabolism and Harvard Medical School, Boston, MA
| | - Judith A Aberg
- Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Carlos Malvestutto
- Division of Infectious Diseases, Ohio State University Wexner Medical Center, Columbus, OH
| | - Michael T Lu
- Cardiac MR PET CT Program and Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Judith S Currier
- David Geffen School of Medicine at University of California Los Angeles, Division of Infectious Diseases, Los Angeles, CA
| | | | - Myron Waclawiw
- National Institutes of Health/National Heart, Lung, and Blood Institute, Bethesda, MD
| | | | | | | | | | - Udo Hoffmann
- Cardiac MR PET CT Program and Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Heather J Ribaudo
- Center for Biostatistics in AIDS Research, Department of Biostatistics, Harvard TH Chan School of Public Health, Boston, MA
| | - Pamela S Douglas
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| |
Collapse
|
44
|
Hoffmann U, Lu MT, Olalere D, Adami EC, Osborne MT, Ivanov A, Aluru JS, Lee S, Arifovic N, Overton ET, Fichtenbaum CJ, Aberg JA, Alston-Smith B, Klingman KL, Waclawiw M, Burdo TH, Williams KC, Zanni MV, Desvigne-Nickens P, Cooper-Arnold K, Fitch KV, Ribaudo H, Douglas PS, Grinspoon SK. Rationale and design of the Mechanistic Substudy of the Randomized Trial to Prevent Vascular Events in HIV (REPRIEVE): Effects of pitavastatin on coronary artery disease and inflammatory biomarkers. Am Heart J 2019; 212:1-12. [PMID: 30928823 PMCID: PMC6596304 DOI: 10.1016/j.ahj.2019.02.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 02/23/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND People with HIV (PWH) have increased cardiovascular events, inflammation, and high-risk coronary atherosclerosis. Statin therapy has been shown to lower the risk of cardiovascular disease (CVD) in the general population, but whether this results from reductions in coronary atherosclerosis and is mediated by decreased inflammation remains unknown. METHODS REPRIEVE is a randomized, placebo-controlled trial of pitavastatin calcium (4 mg/day) vs. placebo enrolling at least 7500 PWH between 40-75 years, on antiretroviral therapy (ART), with low to moderate traditional CVD risk. The Mechanistic Substudy of REPRIEVE (A5333s) is co-enrolling 800 participants from 31 US sites. These participants undergo serial contrast enhanced coronary computed tomography angiography (CCTA) and measurements of biomarkers of inflammation and immune activation at baseline and after 2 years of follow-up. The primary objectives are to determine the effects of pitavastatin on noncalcified coronary atherosclerotic plaque (NCP) volume, low attenuation plaque, and positive remodeling and on changes in immune activation and inflammation and to assess relationships between the two. Changes in CAD will be assessed in a standardized fashion by a core lab with expert readers blinded to time points and participant information; immune activation and inflammation assessment is also performed centrally. RESULTS To date the Mechanistic Substudy has completed planned enrollment, with 805 participants. CONCLUSION This study represents the first large, randomized, CCTA-based assessment of the effects of a primary prevention strategy for CVD on high-risk CAD, immune activation and inflammation among PWH. The study will assess pitavastatin's effects on coronary plaque, and the interrelationship of these changes with biomarkers of immune activation and inflammation in PWH to determine mechanisms of CVD prevention and improved outcomes in this population.
Collapse
Affiliation(s)
- Udo Hoffmann
- Cardiac MR PET CT Program and Department of Radiology, Massachusetts General Hospital, Boston, MA.
| | - Michael T Lu
- Cardiac MR PET CT Program and Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Devvora Olalere
- Cardiac MR PET CT Program and Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Elizabeth C Adami
- Cardiac MR PET CT Program and Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Michael T Osborne
- Cardiac MR PET CT Program and Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Alex Ivanov
- Cardiac MR PET CT Program and Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - John Sukumar Aluru
- Cardiac MR PET CT Program and Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Saeyun Lee
- Cardiac MR PET CT Program and Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Nadja Arifovic
- Cardiac MR PET CT Program and Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Edgar Turner Overton
- Division of Infectious Diseases, University of Alabama at Birmingham School of Medicine, Birmingham, AL
| | - Carl J Fichtenbaum
- Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Judith A Aberg
- Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY
| | | | | | - Myron Waclawiw
- National Institutes of Health/National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Tricia H Burdo
- Department of Neuroscience, Temple University School of Medicine, Philadelphia, PA
| | | | - Markella V Zanni
- MGH Program in Nutritional Metabolism and Harvard Medical School, Boston, MA
| | | | | | - Kathleen V Fitch
- MGH Program in Nutritional Metabolism and Harvard Medical School, Boston, MA
| | - Heather Ribaudo
- Center for Biostatistics in AIDS Research, Department of Biostatistics, Harvard TH Chan School of Public Health, Boston, MA
| | - Pamela S Douglas
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC
| | - Steven K Grinspoon
- MGH Program in Nutritional Metabolism and Harvard Medical School, Boston, MA
| |
Collapse
|
45
|
Alencherry B, Erem G, Mirembe G, Ssinabulya I, Yun CH, Hung CL, Siedner MJ, Bittencourt M, Kityo C, McComsey GA, Longenecker CT. Coronary artery calcium, HIV and inflammation in Uganda compared with the USA. Open Heart 2019; 6:e001046. [PMID: 31218009 PMCID: PMC6546194 DOI: 10.1136/openhrt-2019-001046] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/11/2019] [Accepted: 04/26/2019] [Indexed: 12/22/2022] Open
Abstract
Objectives To compare the prevalence of detectable coronary artery calcium (CAC) among higher risk, older people living with HIV (PLWH) and uninfected persons in Uganda versus the USA, and second to explore associations of CAC with HIV-specific variables and biomarkers of inflammation. Methods This cross-sectional study of 430 total subjects compared 100 PLWH on antiretroviral therapy and 100 age-matched and sex-matched HIV-uninfected controls in Uganda with 167 PLWH on antiretroviral therapy and 63 uninfected controls in the USA. Multivariable logistic regression was used to examine associations with detectable CAC (CAC >0). Results Compared with US subjects, Ugandans were older (mean age 56 vs 52 years) and were more likely to have diabetes (36% vs 3%) and hypertension (85% vs 36%), but were less likely to be male (38% vs 74%) or smokers (4% vs 56%). After adjustment for HIV serostatus, age, sex and traditional risk factors, Ugandans had substantially lower odds of CAC >0 (adjusted OR 0.07 (95% CI 0.03 to 0.17), p<0.001). HIV was not associated with CAC >0 in either country (p>0.1). Among all PLWH, nadir CD4 count was associated with the presence of CAC, and among Ugandans soluble intercellular adhesion molecule (p=0.044), soluble CD163 (p=0.004) and oxidised low-density lipoprotein (p=0.043) were all associated with the presence of CAC. Conclusions Ugandans had a dramatically lower prevalence of any coronary calcification compared with US subjects. The role of HIV infection and inflammation as risk factors for subclinical coronary disease in sub-Saharan Africa merits further investigation.
Collapse
Affiliation(s)
- Ben Alencherry
- Medicine and Pediatrics, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Geoffrey Erem
- Radiology, St Francis Hospital Nsambya, Kampala, Uganda.,Radiology and Medicine, Makerere University College of Health Sciences, Kampala, Uganda
| | - Grace Mirembe
- HIV Medicine, Joint Clinical Research Centre, Kampala, Uganda
| | - Isaac Ssinabulya
- Radiology and Medicine, Makerere University College of Health Sciences, Kampala, Uganda.,Cardiology, Uganda Heart Institute, Kampala, Uganda
| | - Chun-Ho Yun
- Radiology, Mackay Memorial Hospital, Taipei, Taiwan
| | | | - Mark J Siedner
- Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA.,Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | | | - Cissy Kityo
- HIV Medicine, Joint Clinical Research Centre, Kampala, Uganda
| | - Grace A McComsey
- Medicine and Pediatrics, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA.,Pediatric Infectious Diseases, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Chris T Longenecker
- Cardiology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
46
|
Mild renal impairment is associated with calcified plaque parameters assessed by computed tomography angiography in people living with HIV. AIDS 2019; 33:219-227. [PMID: 30325774 DOI: 10.1097/qad.0000000000002055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVE To investigate the association of mild renal impairment and coronary plaque in people living with HIV (PLHIV). METHODS PLHIV and non-HIV controls with serum creatinine less than 1.5 mg/dl were investigated. Estimated glomerular filtration rate (eGFR) (calculated by CKD-EPI formula) was related to coronary plaque indices obtained by CT angiography. RESULTS One hundred and eighty-four PLHIV [HIV viral load, 49 (47,49) copies/ml, CD4+ cell count, median 536 (370, 770) cells/μl, duration HIV, 15 ± 7 years] and 72 HIV-negative controls without known cardiovascular disease (CVD) were studied. The two groups were well matched for traditional CVD risk factors. Serum creatinine (0.9 ± 0.2 vs. 0.9 ± 0.2 mg/dl, P = 0.96) and eGFR (96 ± 22 vs. 96 ± 24 ml/min per 1.73 m(2), P = 0.99) were similar between PLHIV and non-HIV, respectively. In PLHIV, eGFR inversely related to total severity of coronary plaque score (r = -0.27, P = 0.002), total coronary segments with plaque (r = -0.21, P = 0.005), calcified plaque segments (r = -0.15, P = 0.045), and Agatston score (r = -0.21, P = 0.006). Adjusting for total Framingham point score, BMI, and HIV parameters, eGFR remained significantly associated with calcified plaque and Agatston score in PLHIV. In HIV negative controls, eGFR did not correlate with calcified plaque (r = -0.20, P = 0.10) or Agatston score (r = -0.13, P = 0.29). Among PLHIV, those with eGFR less than 90 ml/min per 1.73 m(2) demonstrated increased total severity of coronary plaque score compared with those with eGFR greater than or equal to 90, P = 0.02). This relationship was stronger in PLHIV than the non-HIV group. CONCLUSION Our data highlight a robust relationship between subclinical renal impairment and coronary artery disease among PLHIV. Further research is needed to understand the relationship between mild renal impairment and CVD in HIV.
Collapse
|
47
|
Ryan T, Affandi JS, Gahungu N, Dwivedi G. Noninvasive Cardiovascular Imaging: Emergence of a Powerful Tool for Early Identification of Cardiovascular Risk in People Living With HIV. Can J Cardiol 2018; 35:260-269. [PMID: 30825948 DOI: 10.1016/j.cjca.2018.11.036] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 11/06/2018] [Accepted: 11/20/2018] [Indexed: 12/31/2022] Open
Abstract
Antiretroviral therapy (ART) has been pivotal in prolonging the lifespan of people living with HIV (PLWH). However, this also simultaneously increases their risk of cardiovascular disease (CVD) either related to ART, aging, hypertension, immunosenescence, inflammation, immune activation, or other comorbidities. Although the use of risk markers has greatly enhanced the field of cardiovascular (CV) medicine and improved the prognosis and early diagnosis in the general population, this strategy has not been clearly elucidated in PLWH. Developing accurate risk algorithms for PLWH requires an innate understanding of mechanistic factors influencing their risks. Early identification of CV risk will significantly enhance the prospects of PLWH living longer and relatively healthily. Herein, we discuss the use of multimodality noninvasive CV imaging as robust markers for ameliorating CV risk. The ability to prognosticate CV risk and hence prevent CV events in PLWH would represent an important advance in CV medicine, allowing precise detection and early institution of preventative strategies. Using novel CV imaging modalities and strategies would have a positive impact on precision medicine in this patient cohort.
Collapse
Affiliation(s)
- Timothy Ryan
- Fiona Stanley Hospital, Murdoch, Western Australia, Australia
| | - Jacquita S Affandi
- School of Public Health, Curtin University, Bentley, Western Australia, Australia; Harry Perkins Institute of Medical Research, Murdoch, Western Australia, Australia
| | - Nestor Gahungu
- Royal Perth Hospital, Perth, Western Australia, Australia
| | - Girish Dwivedi
- Fiona Stanley Hospital, Murdoch, Western Australia, Australia; Harry Perkins Institute of Medical Research, Murdoch, Western Australia, Australia; The University of Western Australia, Crawley, Western Australia, Australia.
| |
Collapse
|
48
|
Sinha A, Feinstein MJ. Coronary Artery Disease Manifestations in HIV: What, How, and Why. Can J Cardiol 2018; 35:270-279. [PMID: 30825949 DOI: 10.1016/j.cjca.2018.11.029] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 11/13/2018] [Accepted: 11/22/2018] [Indexed: 12/24/2022] Open
Abstract
Understanding why persons with human immunodeficiency virus (HIV) have accelerated atherosclerosis and its sequelae, including coronary artery disease (CAD) and myocardial infarction, is necessary to provide appropriate care to a large and aging population with HIV. In this review, we delineate the diverse pathophysiologies underlying HIV-associated CAD and discuss how these are implicated in the clinical manifestations of CAD among persons with HIV. Several factors contribute to HIV-associated CAD, with chronic inflammation and immune activation likely representing the primary drivers. Increased monocyte activation, inflammation, and hyperlipidemia present in chronic HIV infection also mirror the pathophysiology of plaque rupture. Furthermore, mechanisms central to plaque erosion, such as activation of toll-like receptor 2 and formation of neutrophil extracellular traps, are also abundant in HIV. In addition to inflammation and immune activation in general, persons with HIV have a higher prevalence than uninfected persons of traditional cardiovascular risk factors, including dyslipidemia, hypertension, insulin resistance, and tobacco use. Antiretroviral therapies, although clearly necessary for HIV treatment and survival, have had varied effects on CAD, but newer generation regimens have reduced cardiovascular toxicities. From a clinical standpoint, this mix of risk factors is implicated in earlier CAD among persons with HIV than uninfected persons; whether the distribution and underlying plaque content of CAD for persons with HIV differs considerably from uninfected persons has not been definitively studied. Furthermore, the role of cardiovascular risk estimators in HIV remains unclear, as does the role of traditional and emerging therapies; no trials of CAD therapies powered to detect clinical events have been completed among persons with HIV.
Collapse
Affiliation(s)
- Arjun Sinha
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Matthew J Feinstein
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA; Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA.
| |
Collapse
|
49
|
Hemmat N, Ebadi A, Badalzadeh R, Memar MY, Baghi HB. Viral infection and atherosclerosis. Eur J Clin Microbiol Infect Dis 2018; 37:2225-2233. [PMID: 30187247 DOI: 10.1007/s10096-018-3370-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 08/28/2018] [Indexed: 12/22/2022]
Abstract
Several risk factors have been described for the pathogenesis of atherosclerosis. Infectious diseases are suggested to be a causative factor, and some viruses have been studied for their relation with atherosclerotic diseases. Studies report two hypotheses, direct and indirect effects, for the role of viral infections in atherogenesis. Viruses are able to initiate atherosclerosis by two different pathways. They can exert their direct effects on atherogenesis by infecting vascular cells and then inducing inflammation in the endothelium and smooth muscle cells. Alternatively, they can also apply indirect effects by infecting non-vascular cells and inducing systemic inflammation. In this review, we consider the available data about the effects and correlations of DNA and RNA viruses on atherosclerosis.
Collapse
Affiliation(s)
- Nima Hemmat
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, PO Box 5165665931, Tabriz, Iran
| | - Amin Ebadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, PO Box 5165665931, Tabriz, Iran
| | - Reza Badalzadeh
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Yousef Memar
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, PO Box 5165665931, Tabriz, Iran.,Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Bannazadeh Baghi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, PO Box 5165665931, Tabriz, Iran. .,Department of Microbiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
50
|
D'Antoni ML, Mitchell BI, McCurdy S, Byron MM, Ogata-Arakaki D, Chow D, Mehta NN, Boisvert WA, Lefebvre E, Shikuma CM, Ndhlovu LC, Baumer Y. Cenicriviroc inhibits trans-endothelial passage of monocytes and is associated with impaired E-selectin expression. J Leukoc Biol 2018; 104:1241-1252. [PMID: 30088682 DOI: 10.1002/jlb.5a0817-328rrr] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 07/12/2018] [Accepted: 07/13/2018] [Indexed: 01/23/2023] Open
Abstract
Incidences of cardiovascular diseases (CVD) are high among virologically suppressed HIV-infected individuals. Monocyte activation and trafficking are key mechanisms in the evolution of CVD. We studied the ability of cenicriviroc (CVC), a dual C-C chemokine receptor type 2 (CCR2) and CCR5 antagonist, to influence the migration of monocytes from HIV-infected individuals on antiretroviral therapy (ART). Monocytes were derived from 23 ART-suppressed HIV-infected and 16 HIV-uninfected donors. In a trans-endothelial migration model, monocytes, and human aortic endothelial cells (HAoECs) were exposed to cenicriviroc and migrated monocytes, quantified. Expression of CCR2 and CCR5 on monocytes and adhesion molecules (E-selectin, ICAM-1, VCAM-1, PECAM-1, and CD99) on HAoECs were measured. The single antagonists, BMS-22 (CCR2), and maraviroc (CCR5), served as controls. When both HAoECs and monocytes together were exposed to the antagonists, cenicriviroc led to a greater decrease in monocyte migration compared to BMS-22 or vehicle in both HIV-infected and HIV-uninfected groups (P < 0.05), with maraviroc having no inhibitory effect. Cenicriviroc treatment of HAoECs alone decreased monocyte migration in the HIV-infected group when compared to vehicle (P < 0.01). Inhibition of migration was not evident when monocytes alone were exposed to cenicriviroc, BMS-22 or maraviroc. Incubation of HAoECs with cenicriviroc decreased E-selectin expression (P = 0.045) but had limited effects on the other adhesion molecules. Cenicriviroc inhibits monocyte trans-endothelial migration more effectively than single chemokine receptor blockade, which may be mediated via disruption of monocyte-endothelial tethering through reduced E-selectin expression. Cenicriviroc should be considered as a therapeutic intervention to reduce detrimental monocyte trafficking.
Collapse
Affiliation(s)
- Michelle L D'Antoni
- Hawaii Center for HIV/AIDS, University of Hawaii, Hawaii, USA.,Department of Tropical Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Brooks I Mitchell
- Hawaii Center for HIV/AIDS, University of Hawaii, Hawaii, USA.,Department of Tropical Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Sara McCurdy
- Department of Medicine, Center for Cardiovascular Research, University of Hawaii, Hawaii, USA
| | - Mary Margaret Byron
- Hawaii Center for HIV/AIDS, University of Hawaii, Hawaii, USA.,Department of Tropical Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | | | - Dominic Chow
- Hawaii Center for HIV/AIDS, University of Hawaii, Hawaii, USA
| | - Nehal N Mehta
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - William A Boisvert
- Department of Medicine, Center for Cardiovascular Research, University of Hawaii, Hawaii, USA
| | | | | | - Lishomwa C Ndhlovu
- Hawaii Center for HIV/AIDS, University of Hawaii, Hawaii, USA.,Department of Tropical Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, Hawaii, USA
| | - Yvonne Baumer
- Hawaii Center for HIV/AIDS, University of Hawaii, Hawaii, USA.,Department of Medicine, Center for Cardiovascular Research, University of Hawaii, Hawaii, USA.,Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|