1
|
Shi M, Feinstein MJ. Immune Dysregulation in Ischemic Heart Disease Among Individuals with Human Immunodeficiency Virus. Heart Fail Clin 2025; 21:227-239. [PMID: 40107801 PMCID: PMC11960833 DOI: 10.1016/j.hfc.2024.12.005] [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: 03/22/2025]
Abstract
Individuals with human immunodeficiency virus (HIV) experience an elevated risk of ischemic heart disease and related cardiovascular sequelae. This is due to a combination of factors including traditional comorbidities, adverse effects of antiretroviral therapy, low-level viremia, viral coinfection, mucosal injury, and chronic immune activation and dysregulation. Understanding the underlying mechanisms, especially as they relate to inflammation, has implications for prevention, diagnostics, and therapeutics. This review highlights some of the foundational and recent literature on clinical phenotypes, diagnostic tools, and promising pharmacotherapies, along with future directions for translational research and clinical implementation.
Collapse
Affiliation(s)
- Meilynn Shi
- Division of Cardiology, Department of Medicine, Bluhm Cardiovascular Institute, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Matthew J Feinstein
- Division of Cardiology, Department of Medicine, Bluhm Cardiovascular Institute, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
2
|
Durstenfeld MS, Hsue PY. Advances in the Management of Cardiovascular Disease in the Setting of Human Immunodeficiency Virus. Infect Dis Clin North Am 2024; 38:517-530. [PMID: 38871571 PMCID: PMC11305916 DOI: 10.1016/j.idc.2024.04.006] [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: 06/15/2024]
Abstract
This state-of-the art review discusses the underlying mechanisms that contribute to atherosclerotic cardiovascular disease, heart failure and arrhythmias among people with human immunodeficiency virus (HIV), risk prediction and prevention, management, and outstanding research questions, including a discussion of how the Randomized Trial to Prevent Vascular Events in HIV may inform clinical practice.
Collapse
Affiliation(s)
- Matthew S Durstenfeld
- Department of Medicine, University of California, San Francisco, CA, USA; Division of Cardiology, Zuckerberg San Francisco General, San Francisco, CA, USA.
| | - Priscilla Y Hsue
- Department of Medicine, University of California, San Francisco, CA, USA; Division of Cardiology, Zuckerberg San Francisco General, San Francisco, CA, USA
| |
Collapse
|
3
|
Cyktor JC, Yeh E, Ribaudo H, Hoeth D, Naqvi A, Bell T, Ridker PM, Fichtenbaum C, Daar ES, Havlir D, Tawakol A, Lederman MM, Stein JH, Deeks SG, Currier JS, Hsue PY, Mellors JW. Brief Report: Low-Dose Methotrexate Does Not Affect Measures of HIV-1 Persistence in Individuals With Chronically Treated HIV-1 Infection. J Acquir Immune Defic Syndr 2024; 96:481-485. [PMID: 39287554 PMCID: PMC11408752 DOI: 10.1097/qai.0000000000003453] [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: 10/20/2023] [Accepted: 03/27/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND People with HIV-1 often have chronic inflammation leading to severe non-AIDS morbidity and mortality. The AIDS Clinical Trials Group Study A5314 sought to lower inflammation with low-dose methotrexate (LDMTX). The primary study outcomes were reported previously but here we present the impact of LDMTX on multiple measures of HIV-1 persistence. METHODS A5314 was a phase 2 randomized, double-blind, multicenter trial in 176 adult people with HIV-1 on virally suppressive antiretroviral therapy. LDMTX (5-15 mg/wk) was administered for 24 weeks with an additional 12 weeks of participant follow-up. The current analyses of HIV-1 persistence were restricted to 60 participants (30 LDMTX and 30 placebo) randomly selected from the total population. Plasma HIV-1 RNA, total HIV-1 DNA, and cell-associated HIV-1 RNA (CA HIV-1 RNA) were measured by sensitive quantitative PCR assays. RESULTS LDMTX treatment had no significant effect on sensitive measures of plasma HIV-1 RNA, HIV-1 DNA, CA HIV-1 RNA, or CA HIV-1 RNA/DNA ratio at any time point or from baseline to week 24. As observed in the main study, absolute peripheral CD4+ and CD8+ T-cell numbers decreased from baseline to week 24 among the 30 participants receiving LDMTX compared with placebo (median decrease of -31.5 CD4+ T cells/µL, -83.5 CD8+ T cells/µL). CONCLUSIONS LDMTX had no significant effect on any measure of HIV-1 persistence in plasma or peripheral blood mononuclear cells. Further studies are needed to determine whether other immunosuppressive and/or immunoreductive interventions are safe and capable of affecting HIV-1 persistence.
Collapse
Affiliation(s)
- Joshua C Cyktor
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Eunice Yeh
- Department of Biostatistics, Harvard TH Chan Sch Public Health, Boston, MA
| | - Heather Ribaudo
- Department of Biostatistics, Harvard TH Chan Sch Public Health, Boston, MA
| | - Dianna Hoeth
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Asma Naqvi
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| | - Tanvir Bell
- The University of Texas Health Science Center at Houston, Houston, TX
| | - Paul M Ridker
- Center for Cardiovascular Disease Prevention, Cardiology Division, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Carl Fichtenbaum
- Division of Infectious Diseases, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Eric S Daar
- Lundquist Institute at Harbor-UCLA Medical Center, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Diane Havlir
- University of California, Biostatistics, Epidemiology, and Computational Precision Health, Berkeley, CA
| | - Ahmed Tawakol
- Cardiovascular Imaging Research Center and Division of Cardiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Michael M Lederman
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH
| | - James H Stein
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Steven G Deeks
- Division of HIV, Infectious Diseases and Global Medicine, Department of Medicine, University of California, San Francisco, CA
| | - Judith S Currier
- Division of Infectious Diseases, Department of Medicine, University of California, Los Angeles, CA; and
| | - Priscilla Y Hsue
- Division of Cardiology, Department of Medicine, University of California, San Francisco, CA
| | - John W Mellors
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
4
|
Hmiel L, Zhang S, Obare LM, Santana MADO, Wanjalla CN, Titanji BK, Hileman CO, Bagchi S. Inflammatory and Immune Mechanisms for Atherosclerotic Cardiovascular Disease in HIV. Int J Mol Sci 2024; 25:7266. [PMID: 39000373 PMCID: PMC11242562 DOI: 10.3390/ijms25137266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 06/26/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
Atherosclerotic vascular disease disproportionately affects persons living with HIV (PLWH) compared to those without. The reasons for the excess risk include dysregulated immune response and inflammation related to HIV infection itself, comorbid conditions, and co-infections. Here, we review an updated understanding of immune and inflammatory pathways underlying atherosclerosis in PLWH, including effects of viral products, soluble mediators and chemokines, innate and adaptive immune cells, and important co-infections. We also present potential therapeutic targets which may reduce cardiovascular risk in PLWH.
Collapse
Affiliation(s)
- Laura Hmiel
- Department of Medicine, Division of Infectious Disease, MetroHealth Medical Center and Case Western Reserve University, Cleveland, OH 44109, USA
| | - Suyu Zhang
- Department of Medicine, Emory University, Atlanta, GA 30322, USA
| | - Laventa M. Obare
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | - Celestine N. Wanjalla
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Boghuma K. Titanji
- Division of Infectious Diseases, Emory University, Atlanta, GA 30322, USA
| | - Corrilynn O. Hileman
- Department of Medicine, Division of Infectious Disease, MetroHealth Medical Center and Case Western Reserve University, Cleveland, OH 44109, USA
| | - Shashwatee Bagchi
- Division of Infectious Diseases, Washington University in St. Louis, St. Louis, MO 63110, USA
| |
Collapse
|
5
|
Obare LM, Temu T, Mallal SA, Wanjalla CN. Inflammation in HIV and Its Impact on Atherosclerotic Cardiovascular Disease. Circ Res 2024; 134:1515-1545. [PMID: 38781301 PMCID: PMC11122788 DOI: 10.1161/circresaha.124.323891] [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
People living with HIV have a 1.5- to 2-fold increased risk of developing cardiovascular disease. Despite treatment with highly effective antiretroviral therapy, people living with HIV have chronic inflammation that makes them susceptible to multiple comorbidities. Several factors, including the HIV reservoir, coinfections, clonal hematopoiesis of indeterminate potential (CHIP), microbial translocation, and antiretroviral therapy, may contribute to the chronic state of inflammation. Within the innate immune system, macrophages harbor latent HIV and are among the prominent immune cells present in atheroma during the progression of atherosclerosis. They secrete inflammatory cytokines such as IL (interleukin)-6 and tumor necrosis-α that stimulate the expression of adhesion molecules on the endothelium. This leads to the recruitment of other immune cells, including cluster of differentiation (CD)8+ and CD4+ T cells, also present in early and late atheroma. As such, cells of the innate and adaptive immune systems contribute to both systemic inflammation and vascular inflammation. On a molecular level, HIV-1 primes the NLRP3 (NLR family pyrin domain containing 3) inflammasome, leading to an increased expression of IL-1β, which is important for cardiovascular outcomes. Moreover, activation of TLRs (toll-like receptors) by HIV, gut microbes, and substance abuse further activates the NLRP3 inflammasome pathway. Finally, HIV proteins such as Nef (negative regulatory factor) can inhibit cholesterol efflux in monocytes and macrophages through direct action on the cholesterol transporter ABCA1 (ATP-binding cassette transporter A1), which promotes the formation of foam cells and the progression of atherosclerotic plaque. Here, we summarize the stages of atherosclerosis in the context of HIV, highlighting the effects of HIV, coinfections, and antiretroviral therapy on cells of the innate and adaptive immune system and describe current and future interventions to reduce residual inflammation and improve cardiovascular outcomes among people living with HIV.
Collapse
Affiliation(s)
- Laventa M. Obare
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN (L.M.O., S.A.M., C.N.W.)
| | - Tecla Temu
- Department of Pathology, Harvard Medical School, Boston, MA (T.T.)
| | - Simon A. Mallal
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN (L.M.O., S.A.M., C.N.W.)
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN (S.A.M.)
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN (S.A.M.)
- Institute for Immunology and Infectious Diseases, Murdoch University, WA, Western Australia (S.A.M.)
| | - Celestine N. Wanjalla
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN (L.M.O., S.A.M., C.N.W.)
| |
Collapse
|
6
|
McCutcheon K, Nqebelele U, Murray L, Thomas TS, Mpanya D, Tsabedze N. Cardiac and Renal Comorbidities in Aging People Living With HIV. Circ Res 2024; 134:1636-1660. [PMID: 38781295 PMCID: PMC11122746 DOI: 10.1161/circresaha.124.323948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Contemporary World Health Organization data indicates that ≈39 million people are living with the human immunodeficiency virus. Of these, 24 million have been reported to have successfully accessed combination antiretroviral therapy. In 1996, the World Health Organization endorsed the widespread use of combination antiretroviral therapy, transforming human immunodeficiency virus infection from being a life-threatening disease to a chronic illness characterized by multiple comorbidities. The increased access to combination antiretroviral therapy has translated to people living with human immunodeficiency virus (PLWH) no longer having a reduced life expectancy. Although aging as a biological process increases exposure to oxidative stress and subsequent systemic inflammation, this effect is likely enhanced in PLWH as they age. This narrative review engages the intricate interplay between human immunodeficiency virus associated chronic inflammation, combination antiretroviral therapy, and cardiac and renal comorbidities development in aging PLWH. We examine the evolving demographic profile of PLWH, emphasizing the increasing prevalence of aging individuals within this population. A central focus of the review discusses the pathophysiological mechanisms that underpin the heightened susceptibility of PLWH to renal and cardiac diseases as they age.
Collapse
Affiliation(s)
| | - Unati Nqebelele
- Non-Communicable Diseases Research Unit, South African Medical Research Council, Cape Town, Western Cape, South Africa (U.N.)
- Department of Medicine, Faculty of Health Sciences, University of Cape Town, Western Cape, South Africa (U.N.)
| | - Lyle Murray
- Division of Infectious Diseases, Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand and the Charlotte Maxeke Johannesburg Academic Hospital, South Africa (L.M.)
| | - Teressa Sumy Thomas
- Division of Endocrinology and Metabolism, Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand and the Chris Hani Baragwanath Academic Hospital, Johannesburg, Gauteng, South Africa (T.S.T.)
| | - Dineo Mpanya
- Division of Cardiology, Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Gauteng, South Africa (D.M., N.T.)
| | - Nqoba Tsabedze
- Division of Cardiology, Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Gauteng, South Africa (D.M., N.T.)
| |
Collapse
|
7
|
Hanley S, Moodley D, Naidoo M, Brummel SS. The Impact of Regular Screening and Lifestyle Modification on Cardiovascular Disease Risk Factors in South African Women Living With HIV. J Acquir Immune Defic Syndr 2024; 96:23-33. [PMID: 38427932 PMCID: PMC11008438 DOI: 10.1097/qai.0000000000003387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 01/03/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND The Integration of cardiovascular disease SCreening and prevention in the HIV MAnagement plan for women of reproductive age study set out to determine the effectiveness of screening and lifestyle modification in modifying cardiovascular disease (CVD) risk factors in women living with HIV (WLHIV). METHODS In this prospective, quasiexperimental, intervention study, WLHIV aged 18-<50 years were enrolled from 2 clinics (intervention [I-arm]) and (control arms [C-arm]) in Umlazi, South Africa, between November 2018 and May 2019. Women in the I-arm received lifestyle modification advice on diet, physical activity, alcohol use, and smoking cessation and underwent annual screening for CVD risk. The CVD risk factors were assessed through standardized questionnaires and clinical and laboratory procedures at baseline and at end of 3 years of follow-up. Prevalence of metabolic syndrome and other CVD indices were compared between arms at end-of-study (EOS). RESULTS Total of 269 WLHIV (149 I-arm and 120 C-arm) with a mean ± SD age of 36 ± 1 years were included in the EOS analyses after 32 ± 2 months of follow-up. The metabolic syndrome prevalence at EOS was 16.8% (25/149) in the I-arm and 24% (24/120) in the C-arm (risk ratio 0.9; 95% CI: 0.5 to 1.1; P 0.86). Proportion of women with fasting blood glucose >5.6 mmol/L in the I-arm and C-arm were 2.7% (4/149) and 13.3% (16/120) respectively (risk ratio 0.2; 95% CI: 0.069 to 0.646; P < 0.01). High-density lipoprotein improved with the intervention arm from baseline to EOS (95% CI: -0.157 to -0.034; P < 0.05). CONCLUSIONS Although there was no significant difference in the prevalence of metabolic syndrome between study arms, we observed decreased blood glucose levels in the I-arm compared with the C-arm and improved high-density lipoprotein within the I-arm, following lifestyle modification and regular screening for CVD risk factors in WLHIV.
Collapse
Affiliation(s)
- Sherika Hanley
- Department of Family Medicine, University of KwaZulu-Natal, Durban, South Africa
- Umlazi CRS, Centre for AIDS Programme of Research in South Africa, Durban, South Africa
| | - Dhayendre Moodley
- Umlazi CRS, Centre for AIDS Programme of Research in South Africa, Durban, South Africa
- Department of Obstetrics and Gynaecology, University of KwaZulu-Natal, Durban, South Africa; and
| | - Mergan Naidoo
- Department of Family Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Sean S. Brummel
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, MA
| |
Collapse
|
8
|
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
|
9
|
Rohani R, Malakismail J, Njoku E. Pharmacological and Behavioral Interventions to Mitigate Premature Aging in Patients with HIV. Curr HIV/AIDS Rep 2023; 20:394-404. [PMID: 37917387 DOI: 10.1007/s11904-023-00677-7] [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] [Accepted: 10/18/2023] [Indexed: 11/04/2023]
Abstract
PURPOSE OF REVIEW We sought to review pharmacological and behavioral interventions that have been publicly presented, published, or are currently ongoing to prevent or mitigate the effect of premature HIV-associated comorbidities. RECENT FINDINGS Multiple studies have been conducted in hopes of finding an effective intervention. While the choice of antiretroviral regimen influences recovery of immune function, several drugs used as adjunct treatments have proven effective to mitigate premature aging. Additionally, few behavioral interventions have exhibited some efficacy. Statins, angiotensin-receptor blockers, and anti-hyperglycemic agents as well as optimal adherence, exercise, and intermittent fasting among others have had beneficial impact on markers of immune activation and levels of inflammatory biomarkers. However, several investigations had inconclusive outcomes so further studies with larger sample sizes are warranted.
Collapse
Affiliation(s)
- Roxane Rohani
- Discipline of Cellular and Molecular Pharmacology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Road BSB 3.266, North Chicago, IL, USA.
- Department of Pharmacy, Captain James A. Lovell Federal Health Care Center, North Chicago, IL, USA.
| | - Jacob Malakismail
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Emmanuel Njoku
- Section of Infectious Disease, Captain James A. Lovell Federal Health Care Center, North Chicago, IL, USA
- Discipline of Internal Medicine, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| |
Collapse
|
10
|
Boczar KE, Faller E, Zeng W, Wang J, Small GR, Corrales-Medina VF, deKemp RA, Ward NC, Beanlands RSB, MacPherson P, Dwivedi G. Anti-inflammatory effect of rosuvastatin in patients with HIV infection: An FDG-PET pilot study. J Nucl Cardiol 2022; 29:3057-3068. [PMID: 34820771 DOI: 10.1007/s12350-021-02830-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/22/2021] [Indexed: 01/29/2023]
Abstract
AIMS This study aimed to evaluate markers of systemic as well as imaging markers of inflammation in the ascending aorta, bone marrow, and spleen measured by 18F-FDG PET/CT, in HIV+ patients at baseline and following therapy with rosuvastatin. METHODS AND RESULTS Of the 35 HIV+ patients enrolled, 17 were randomized to treatment with 10 mg/day rosuvastatin and 18 to usual care for 6 months. An HIV- control cohort was selected for baseline comparison of serum inflammatory markers and monocyte markers of inflammation. 18F-FDG-PET/CT imaging of bone marrow, spleen, and thoracic aorta was performed in the HIV+ cohort at baseline and 6 months. While CD14++CD16- and CCR2 expressions were reduced, serum levels of IL-7, IL-8, and MCP-1 were elevated in the HIV+ population compared to the controls. There was a significant drop in FDG uptake in the bone marrow (TBRmax), spleen (SUVmax) and thoracic aortic (TBRmax) in the statin-treated group compared to the control group (bone marrow: - 10.3 ± 16.9% versus 5.0 ± 18.9%, p = .0262; spleen: - 9.8 ± 20.3% versus 11.3 ± 28.8%, p = .0497; thoracic aorta: - 19.1 ± 24.2% versus 4.3 ± 15.4%, p = .003). CONCLUSIONS HIV+ patients had significantly markers of systemic inflammation including monocyte activation. Treatment with low-dose rosuvastatin in the HIV+ cohort significantly reduced bone marrow, spleen and thoracic aortic FDG uptake.
Collapse
Affiliation(s)
- Kevin E Boczar
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, ON, Canada
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
| | - Elliot Faller
- The Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Wanzhen Zeng
- The Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Jerry Wang
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, ON, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Gary R Small
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, ON, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Vicente F Corrales-Medina
- The Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Robert A deKemp
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, ON, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Natalie C Ward
- School of Public Health, Curtin University, Perth, Australia
- School of Medicine, University of Western Australia, Perth, Australia
| | - Rob S B Beanlands
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, ON, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Paul MacPherson
- The Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Girish Dwivedi
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ottawa, ON, Canada.
- School of Medicine, University of Western Australia, Perth, Australia.
- Department of Advanced Clinical and Translational Cardiovascular Imaging, Harry Perkins Institute of Medical Research, The University of Western Australia, Murdoch, Australia.
- Department of Cardiology, Fiona Stanley Hospital, Murdoch, WA, 6009, Australia.
- School of Biomedical Sciences at Curtin University, Perth, WA, Australia.
| |
Collapse
|
11
|
Liu A, Feinstein M. Addressing gaps in cardiovascular care for people with HIV: bridging scientific evidence and practice. Curr Opin HIV AIDS 2022; 17:279-285. [PMID: 35938461 PMCID: PMC9373406 DOI: 10.1097/coh.0000000000000754] [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: 12/14/2022]
Abstract
PURPOSE OF REVIEW People with HIV continue to have an excess burden of cardiovascular disease compared to the general population. The reasons for these disparities in cardiovascular disease include HIV-specific risk enhancers, traditional atherosclerotic cardiovascular disease risk factors, and sociodemographic disparities, all of which are ripe targets for intervention. RECENT FINDINGS Accurate risk prediction of atherosclerotic cardiovascular disease remains difficult, and cardiovascular risk for people with HIV may be underestimated in the absence of HIV-specific risk enhancers. Despite this increased cardiovascular risk, people with HIV are undertreated and often placed on inadequate lipid lowering therapy. Structural racism and HIV-related stigma play a role, and provider-level and structural-level interventions to encourage early identification and treatment of persons at high risk are necessary. SUMMARY Persons with HIV should be screened with existing cardiovascular risk prediction tools, and those at high risk cardiovascular disease should be promptly referred for lifestyle and pharmacologic interventions as appropriate. System-level implementation research is ongoing in attempts to narrow the gap in cardiovascular care, particularly for vulnerable communities in low resource settings.
Collapse
Affiliation(s)
- Albert Liu
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine
| | - Matthew Feinstein
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine
| |
Collapse
|
12
|
Freeman ML, Clagett BM, Moisi D, Yeh E, Morris CD, Ryu A, Rodriguez B, Stein JH, Deeks SG, Currier JS, Hsue PY, Anthony DD, Calabrese LH, Ribaudo HJ, Lederman MM. Methotrexate Inhibits T Cell Proliferation but Not Inflammatory Cytokine Expression to Modulate Immunity in People Living With HIV. Front Immunol 2022; 13:924718. [PMID: 35967371 PMCID: PMC9374564 DOI: 10.3389/fimmu.2022.924718] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/10/2022] [Indexed: 11/13/2022] Open
Abstract
Inflammation associated with increased risk of comorbidities persists in people living with HIV (PWH) on combination antiretroviral therapy (ART). A recent placebo-controlled trial of low-dose methotrexate (MTX) in PWH found that numbers of total CD4 and CD8 T cells decreased in the low-dose MTX arm. In this report we analyzed T cell phenotypes and additional plasma inflammatory indices in samples from the trial. We found that cycling (Ki67+) T cells lacking Bcl-2 were reduced by MTX but plasma inflammatory cytokines were largely unaffected. In a series of in vitro experiments to further investigate the mechanisms of MTX activity, we found that MTX did not inhibit effector cytokine production but inhibited T cell proliferation downstream of mTOR activation, mitochondrial function, and cell cycle entry. This inhibitory effect was reversible with folinic acid, suggesting low-dose MTX exerts anti-inflammatory effects in vivo in PWH largely by blocking T cell proliferation via dihydrofolate reductase inhibition, yet daily administration of folic acid did not rescue this effect in trial participants. Our findings identify the main mechanism of action of this widely used anti-inflammatory medicine in PWH and may provide insight into how MTX works in the setting of other inflammatory conditions.
Collapse
Affiliation(s)
- Michael L. Freeman
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals, Cleveland Medical Center, Cleveland, OH, United States
| | - Brian M. Clagett
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals, Cleveland Medical Center, Cleveland, OH, United States
| | - Daniela Moisi
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals, Cleveland Medical Center, Cleveland, OH, United States
| | - Eunice Yeh
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Charles D. Morris
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals, Cleveland Medical Center, Cleveland, OH, United States
| | - Angela Ryu
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals, Cleveland Medical Center, Cleveland, OH, United States
| | - Benigno Rodriguez
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals, Cleveland Medical Center, Cleveland, OH, United States
| | - James H. Stein
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Steven G. Deeks
- Division of HIV, Infectious Diseases and Global Medicine, Department of Medicine, University of San Francisco School of Medicine, San Francisco, CA, United States
| | - Judith S. Currier
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Priscilla Y. Hsue
- Division of Cardiology, Department of Medicine, University of California, San Francisco School of Medicine, San Francisco, CA, United States
| | - Donald D. Anthony
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals, Cleveland Medical Center, Cleveland, OH, United States
- Louis Stokes Cleveland Veterans Affairs Medical Center, US Department of Veterans Affairs, Cleveland, OH, United States
- Division of Rheumatic Diseases, MetroHealth Medical Center, Cleveland, OH, United States
| | - Leonard H. Calabrese
- Fasenmyer Center for Immunology, Division of Rheumatic Diseases, Cleveland Clinic, Cleveland, OH, United States
| | - Heather J. Ribaudo
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Michael M. Lederman
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Case Western Reserve University/University Hospitals, Cleveland Medical Center, Cleveland, OH, United States
| |
Collapse
|
13
|
Wagle A, Goerlich E, Post WS, Woldu B, Wu KC, Hays AG. HIV and Global Cardiovascular Health. Curr Cardiol Rep 2022; 24:1149-1157. [PMID: 35802233 DOI: 10.1007/s11886-022-01741-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/19/2022] [Indexed: 11/30/2022]
Abstract
PURPOSE OF REVIEW Because of effective combination antiretroviral therapy, people living with HIV (PLWH) are living longer but developing chronic age-related conditions including cardiovascular disease (CVD), the leading cause of death globally. This review aims to discuss the epidemiology, mechanisms, and clinical considerations of CVD in PLWH from a global perspective. RECENT FINDINGS PLWH are at greater risk for CVD at chronologically younger ages than those without HIV. Potential underlying mechanisms for CVD in PLWH include systemic inflammation, comorbidities, immune-mediated, or treatment-related mechanisms. There is also risk factor variation based on geographical location, including non-traditional CVD risk factors. CVD is prevalent in PLWH and increasing on a global scale. Further understanding the unique epidemiology, risk factors, and treatment of CVD in this population will improve the care of PLWH.
Collapse
Affiliation(s)
- Anjali Wagle
- Department of Medicine, Johns Hopkins Division of Cardiology, Baltimore, MD, USA
| | - Erin Goerlich
- Department of Medicine, Johns Hopkins Division of Cardiology, Baltimore, MD, USA
| | - Wendy S Post
- Department of Medicine, Johns Hopkins Division of Cardiology, Baltimore, MD, USA.,Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bethel Woldu
- Department of Medicine, Johns Hopkins Division of Cardiology, Baltimore, MD, USA.,MedStar Heart and Vascular Institute, Baltimore, MD, USA.,Department of Medicine, Division of Cardiology, MedStar Georgetown University, Washington, DC, USA
| | - Katherine C Wu
- Department of Medicine, Johns Hopkins Division of Cardiology, Baltimore, MD, USA.,Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Allison G Hays
- Department of Medicine, Johns Hopkins Division of Cardiology, Baltimore, MD, USA. .,Ciccarone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
14
|
McCutcheon K, Manga P. Human Immunodeficiency Virus and Cardiovascular Disease: Revisiting the Inflammation-Thrombosis Axis. Thromb Haemost 2022; 122:476-479. [PMID: 34689321 DOI: 10.1055/s-0041-1736445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Keir McCutcheon
- Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| | - Pravin Manga
- Department of Internal Medicine, School of Clinical Medicine, Faculty of Health Sciences, University of Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
15
|
Masters MC, Landay AL, Robbins PD, Tchkonia T, Kirkland JL, Kuchel GA, Niedernhofer LJ, Palella FJ. Chronic HIV Infection and Aging: Application of a Geroscience-Guided Approach. J Acquir Immune Defic Syndr 2022; 89:S34-S46. [PMID: 35015744 PMCID: PMC8751288 DOI: 10.1097/qai.0000000000002858] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 09/27/2021] [Indexed: 12/16/2022]
Abstract
ABSTRACT The ability of virally suppressive antiretroviral therapy use to extend the life span of people with HIV (PWH) implies that the age of PWH will also increase. Among PWH, extended survival comes at a cost of earlier onset and increased rates of aging-associated comorbidities and geriatric syndromes, with persistent inflammation and immune dysregulation consequent to chronic HIV infection and to antiretroviral therapy use contributing to an overall decrease in health span. The geroscience hypothesis proposes that the root causes of most aging-related chronic diseases and conditions is the aging process itself. Hence, therapeutically targeting fundamental aging processes could have a greater impact on alleviating or delaying aging-associated comorbidities than addressing each disease individually. Extending the geroscience hypothesis to PWH, we speculate that targeting basic mechanisms of aging will improve overall health with age. Clinical features and pathophysiologic mechanisms of chronic diseases in PWH qualitatively resemble those seen in older adults without HIV. Therefore, drugs that target any of the pillars of aging, including metformin, rapamycin, and nicotinamide adenine dinucleotide precursors, may also slow the rate of onset of age-associated comorbidities and geriatric syndromes in PWH. Drugs that selectively induce apoptosis of senescent cells, termed senolytics, may also improve health span among PWH. Preliminary evidence suggests that senescent cell burden is increased in PWH, implying that senescent cells are an excellent therapeutic target for extending health span. Recently initiated clinical trials evaluating senolytics in age-related diseases offer insights into the design and potential implementation of similar trials for PWH.
Collapse
Affiliation(s)
- Mary C. Masters
- Department of Medicine, Division of Infectious Diseases, Northwestern University, Chicago, IL
| | - Alan L. Landay
- Department of Internal Medicine, Section of Geriatric Medicine Rush University Medical Center, Chicago, IL
| | - Paul D. Robbins
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; and
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN; and
| | - James L. Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN; and
| | | | - Laura J. Niedernhofer
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN
| | - Frank J. Palella
- Department of Medicine, Division of Infectious Diseases, Northwestern University, Chicago, IL
| |
Collapse
|
16
|
Bernal E, Martinez M, Campillo JA, Puche G, Baguena C, Tomás C, Jimeno A, Alcaraz MJ, Alcaraz A, Muñoz A, Oliver E, de la Torre A, Marín I, Cano A, Minguela A. Moderate to intense physical activity is associated with improved clinical, CD4/CD8 ratio and immune activation status in HIV infected patients on ART. Open Forum Infect Dis 2021; 9:ofab654. [PMID: 35146043 PMCID: PMC8826224 DOI: 10.1093/ofid/ofab654] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 12/27/2021] [Indexed: 11/27/2022] Open
Abstract
Background Physical activity has anti-inflammatory effects and reduces morbidity and mortality in the general population, but its role in the clinical, CD4/CD8 ratio, and immune activation status of HIV-infected patients has been poorly studied. Methods A cross-sectional study was carried out in a cohort of 155 HIV-infected patients on stable antiretroviral therapy (ART) to compare clinical, biochemical, CD4/CD8 ratio, and immune activation status according to their physical activity in the last 2 years (sedentary/low vs moderate/intense) assessed by the iPAQ. A binary logistic regression and mixed analysis of variance were performed to evaluate the impact of levels of physical activity on CD4/CD8 ratio. Results In our series, 77 (49.7%) out of 155 patients were sedentary, and 78 (50.3%) practiced moderate/intense physical activity. Moderate/intense physical activity was associated with better metabolic control (lower body mass index, P = .024; glucose, P = .024; and triglyceride, P = .002) and CDC HIV stage (P = .046), lower CD8+ (P = .018), CD4+CD8+ (P = .026), CD4+CD86+ (P = .045), CD4+HLA-DR+ (P = .011), CD8+HLA-DR+ (P = .048) T lymphocytes and CD16+HLA-DR+ natural killer cells (P = .026), and higher CD3+CD4+ T lymphocytes (P = .016) and CD4/CD8 ratio (P = .001). Sedentary lifestyle (odds ratio [OR], 2.12; P = .042), CD4 nadir (OR, 1.005; P < .001), and CD8+CD38+ T cells (OR, 1.27; P = .006) were independently associated with low CD4/CD8 ratio (<0.8). Earlier and more intense CD4/CD8 ratio recovery was observed in patients with higher physical activity in the 2-year follow-up with a significant interaction between these variables: F(2, 124) = 3.31; P = .049; partial η2 = 0.042. Conclusions Moderate to high physical activity is associated with beneficial health effects, improvement in metabolic profile, and reduction of chronic inflammation in patients with HIV. Although more studies and clinical trials are needed to confirm these findings, a healthy lifestyle including at least moderate physical activity should be recommended to HIV patients on stable ART.
Collapse
Affiliation(s)
- Enrique Bernal
- Infectious Disease Unit, Reina Sofia University Hospital and Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Monica Martinez
- Infectious Disease Unit, Reina Sofia University Hospital and Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - José Antonio Campillo
- Immunology Service, Virgen de la Arrrixaca University Clinical Hospital (HCUVA), Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Gabriel Puche
- Infectious Disease Unit, Reina Sofia University Hospital and Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Carlos Baguena
- Infectious Disease Unit, Reina Sofia University Hospital and Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Cristina Tomás
- Infectious Disease Unit, Reina Sofia University Hospital and Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Amaya Jimeno
- Internal Medicine Service, Infectious Disease Unit. Santa Lucía University Hospital, Cartagena, Spain
| | - Maria Jose Alcaraz
- Infectious Disease Unit, Reina Sofia University Hospital and Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Antonia Alcaraz
- Infectious Disease Unit, Reina Sofia University Hospital and Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Angeles Muñoz
- Infectious Disease Unit, Reina Sofia University Hospital and Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Eva Oliver
- Infectious Disease Unit, Reina Sofia University Hospital and Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Alejandro de la Torre
- Infectious Disease Unit, Reina Sofia University Hospital and Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Irene Marín
- Cardiology Unit, Reina Sofia University Hospital and Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Alfredo Cano
- Infectious Disease Unit, Reina Sofia University Hospital and Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Alfredo Minguela
- Immunology Service, Virgen de la Arrrixaca University Clinical Hospital (HCUVA), Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| |
Collapse
|
17
|
Thakkar AB, Ma Y, Dela Cruz M, Wu Y, Arechiga V, Swaminathan S, Ganz P, Wu AHB, Scherzer R, Deeks S, Hsue PY. Effect of HIV-1 Infection on Angiopoietin 1 and 2 Levels and Measures of Microvascular and Macrovascular Endothelial Dysfunction. J Am Heart Assoc 2021; 10:e021397. [PMID: 34726064 PMCID: PMC8751943 DOI: 10.1161/jaha.121.021397] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Individuals infected with HIV have an increased risk of developing cardiovascular disease; yet, the underlying mechanisms remain unknown. Recent evidence has implicated the Tie-2 tyrosine kinase receptor system and its associated ligands ANG1 (angiopoietin 1) and ANG2 (angiopoietin 2) in maintaining vascular homeostasis. In the general population, lower ANG1 levels and higher ANG2 levels are strongly correlated with the development of cardiovascular disease. In this study, we aim to investigate the associations of HIV infection with angiopoietin levels and endothelial dysfunction. Methods and Results In this cross-sectional study, we compared measures of ANG1, ANG2, and endothelial dysfunction using flow-mediated vasodilation of the brachial artery in 39 untreated subjects infected with HIV, 47 treated subjects infected with HIV, and 46 uninfected subjects from the SCOPE (Observational Study of the Consequences of the Protease Inhibitor Era) cohort. Compared with uninfected controls, treated individuals infected with HIV had 53.1% lower mean ANG1 levels (P<0.01) and similar ANG2 levels. On the other hand, untreated individuals infected with HIV had similar ANG1 levels, and 29.2% had higher ANG2 levels (P<0.01) compared with uninfected controls. When compared with individuals with untreated HIV infection, those with treated HIV infection had 56% lower ANG1 levels (P<0.01) and 22% lower ANG2 levels (P<0.01).Both treated and untreated HIV infection were associated with significant impairment in hyperemic velocity, a key measure of microvascular dysfunction (median 61 versus 72 cm/s, P<0.01), compared with uninfected controls (median 73 cm/s). This difference persisted after adjustment for ANG1 and ANG2 levels. Interestingly, when compared with untreated individuals infected with HIV, treated individuals infected with HIV had worse hyperemic velocity (-12.35 cm/s, P=0.05). In contrast, HIV status, ANG1 levels, and ANG2 levels were not associated with macrovascular dysfunction as measured by flow-mediated dilatation and brachial artery diameter, 2 other measures of vascular homeostasis. Conclusions HIV infection affects the balance between levels of ANG1 and ANG2 and may disturb endothelial homeostasis through disruption of vascular homeostasis. Individuals with treated HIV had decreased ANG1 levels and similar ANG2 levels, whereas individuals with untreated HIV had similar ANG1 levels and increased ANG2 levels, suggesting that treatment status may alter the balance between ANG1 and ANG2. HIV also promotes endothelial dysfunction via impairment of microvascular dysfunction, independent of the Tie-2 receptor system; the finding of worse microvascular dysfunction in the setting of treated HIV infection may reflect the impact of viral persistence on the microvasculature or toxicities of specific antiretroviral regimens. Further research to clarify the mechanism of HIV-mediated endothelial dysfunction is necessary to advance treatment of cardiovascular complications of HIV infection.
Collapse
Affiliation(s)
- Anjali B Thakkar
- Division of Cardiology Department of Medicine Zuckerberg San Francisco General HospitalUniversity of California San Francisco San Francisco CA
| | - Yifei Ma
- Division of Cardiology Department of Medicine Zuckerberg San Francisco General HospitalUniversity of California San Francisco San Francisco CA
| | - Mark Dela Cruz
- Section of Cardiology Department of Medicine University of Chicago Medical Center Chicago IL
| | - Yuaner Wu
- Division of Cardiology Department of Medicine Zuckerberg San Francisco General HospitalUniversity of California San Francisco San Francisco CA
| | - Victor Arechiga
- Division of Cardiology Department of Medicine Zuckerberg San Francisco General HospitalUniversity of California San Francisco San Francisco CA
| | - Shreya Swaminathan
- Division of Cardiology Department of Medicine Zuckerberg San Francisco General HospitalUniversity of California San Francisco San Francisco CA
| | - Peter Ganz
- Division of Cardiology Department of Medicine Zuckerberg San Francisco General HospitalUniversity of California San Francisco San Francisco CA
| | - Alan H B Wu
- Division of Clinical Chemistry Department of Laboratory Medicine Zuckerberg San Francisco General HospitalUniversity of California San Francisco San Francisco CA
| | - Rebecca Scherzer
- San Francisco Veteran's Affairs Medical Center San Francisco CA.,Department of Medicine University of California San Francisco CA
| | - Steven Deeks
- Positive Health Program Zuckerberg San Francisco General HospitalUniversity of California San Francisco CA
| | - Priscilla Y Hsue
- Division of Cardiology Department of Medicine Zuckerberg San Francisco General HospitalUniversity of California San Francisco San Francisco CA
| |
Collapse
|
18
|
HIV-Related Immune Activation and Inflammation: Current Understanding and Strategies. J Immunol Res 2021; 2021:7316456. [PMID: 34631899 PMCID: PMC8494587 DOI: 10.1155/2021/7316456] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/20/2021] [Indexed: 02/07/2023] Open
Abstract
Although antiretroviral therapy effectively controls human immunodeficiency virus (HIV) replication, a residual chronic immune activation/inflammation persists throughout the disease. This aberrant immune activation and inflammation are considered an accelerator of non-AIDS-related events and one of the driving forces of CD4+ T cell depletion. Unfortunately, HIV-associated immune activation is driven by various factors, while the mechanism of excessive inflammation has not been formally clarified. To date, several clinical interventions or treatment candidates undergoing clinical trials have been proposed to combat this systemic immune activation/inflammation. However, these strategies revealed limited results, or their nonspecific anti-inflammatory properties are similar to previous interventions. Here, we reviewed recent learnings of immune activation and persisting inflammation associated with HIV infection, as well as the current directions to overcome it. Of note, a more profound understanding of the specific mechanisms for aberrant inflammation is still imperative for identifying an effective clinical intervention strategy.
Collapse
|
19
|
Hixson EA, Borker PV, Jackson EK, Macatangay BJ. The Adenosine Pathway and Human Immunodeficiency Virus-Associated Inflammation. Open Forum Infect Dis 2021; 8:ofab396. [PMID: 34557556 PMCID: PMC8454523 DOI: 10.1093/ofid/ofab396] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 07/23/2021] [Indexed: 12/15/2022] Open
Abstract
Human immunodeficiency virus (HIV) is associated with an increased risk of age-associated comorbidities and mortality compared to people without HIV. This has been attributed to HIV-associated chronic inflammation and immune activation despite viral suppression. The adenosine pathway is an established mechanism by which the body regulates persistent inflammation to limit tissue damage associated with inflammatory conditions. However, HIV infection is associated with derangements in the adenosine pathway that limits its ability to control HIV-associated inflammation. This article reviews the function of purinergic signaling and the role of the adenosine signaling pathway in HIV-associated chronic inflammation. This review also discusses the beneficial and potential detrimental effects of pharmacotherapeutic strategies targeting this pathway among people with HIV.
Collapse
Affiliation(s)
- Emily A Hixson
- Department of Infectious Disease and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA
| | - Priya V Borker
- Division of Pulmonary Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Edwin K Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Bernard J Macatangay
- Department of Infectious Disease and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA.,Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pennsylvania, USA
| |
Collapse
|
20
|
Bonou M, Kapelios CJ, Protogerou AD, Mavrogeni S, Aggeli C, Markousis-Mavrogenis G, Psichogiou M, Barbetseas J. Cardiac adiposity as a modulator of cardiovascular disease in HIV. HIV Med 2021; 22:879-891. [PMID: 34514685 DOI: 10.1111/hiv.13166] [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] [Received: 05/13/2021] [Revised: 08/02/2021] [Accepted: 08/13/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND With the number of people living with human immunodeficiency virus (HIV) steadily increasing, cardiovascular disease has emerged as a leading cause of non-HIV related mortality. People living with HIV (PLWH) appear to be at increased risk of coronary artery disease and heart failure (HF), while the underlying mechanism appears to be multifactorial. In the general population, ectopic cardiac adiposity has been highlighted as an important modulator of accelerated coronary artery atherosclerosis, arrhythmogenesis and HF with preserved ejection fraction (HFpEF). Cardiac adiposity is also strongly linked with obesity, especially with visceral adipose tissue accumulation. AIMS This review aims to summarize the possible role of cardiac fat depositions, assessed by imaging modalities,as potential contributors to the increased cardiac morbidity and mortality seen in PLWH, as well as therapeutic targets in the current ART era. MATERIALS & METHODS Review of contemporary literature on this topic. DISCUSSION Despite antiretroviral therapy (ART), PLWH have evidence of persistent, HIV-related systemic inflammation and body fat alterations. Cardiac adiposity can play an additional role in the pathogenesis of cardiovascular disease in the HIV setting. Imaging modalities such as echocardiography, cardiac multidetector computed tomography and cardiac magnetic resonance have demonstrated increased adipose tissue. Studies show that high cardiac fat depots play an additive role in promoting coronary artery atherosclerosis and HFpEF in PLWH. Systemic inflammation due to HIV infection, metabolic adverse effects of ART, adipose alterations in the ageing HIV population, inflammation and immune activation are likely important mechanisms for adipose dysfunction and disproportionately occurrence of ectopic fat depots in the heart among PLWH. CONCLUSIONS High cardiac adiposity seems to plays an additive role in promoting coronary artery atherosclerosis and HFpEF in PLWH. The underlying mechanisms are multiple and warrant further investigation. Improved understanding of the regulating mechanisms that increase cardiovascular risk in HIV infection may give rise to more tailored therapeutic strategies targeting cardiac fat depots.
Collapse
Affiliation(s)
- Maria Bonou
- Department of Cardiology, Laiko General Hospital, Athens, Greece
| | - Chris J Kapelios
- Department of Cardiology, Laiko General Hospital, Athens, Greece
| | - Athanase D Protogerou
- Cardiovascular Prevention & Research Unit, Clinic and Laboratory of Pathophysiology, National and Kapodistrian University Athens School of Medicine, Athens, Greece
| | - Sophie Mavrogeni
- Department of Cardiology, Onassis Cardiac Surgery Center, Athens, Greece
| | - Constantina Aggeli
- First Department of Cardiology, Hippokration General Hospital, National and Kapodistrian University Athens School of Medicine, Athens, Greece
| | | | - Mina Psichogiou
- First Department of Internal Medicine, Laiko General Hospital, National and Kapodistrian University Athens School of Medicine, Athens, Greece
| | - John Barbetseas
- Department of Cardiology, Laiko General Hospital, Athens, Greece
| |
Collapse
|
21
|
Kelly C, Tinago W, Alber D, Hunter P, Luckhurst N, Connolly J, Arrigoni F, Garcia Abner A, Kamn’gona R, Sheha I, Chammudzi M, Jambo K, Mallewa J, Rapala A, Mallon PWG, Mwandumba H, Klein N, Khoo S. Inflammatory pathways amongst people living with HIV in Malawi differ according to socioeconomic status. PLoS One 2021; 16:e0256576. [PMID: 34432828 PMCID: PMC8386842 DOI: 10.1371/journal.pone.0256576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 08/11/2021] [Indexed: 11/18/2022] Open
Abstract
Background Non-communicable diseases (NCDs) are increased amongst people living with HIV (PLWH) and are driven by persistent immune activation. The role of socioeconomic status (SES) in immune activation amongst PLWH is unknown, especially in low-income sub-Saharan Africa (SSA), where such impacts may be particularly severe. Methods We recruited Malawian adults with CD4<100 cells/ul two weeks after starting ART in the REALITY trial (NCT01825031), as well as volunteers without HIV infection. Clinical assessment, socioeconomic evaluation, blood draw for immune activation markers and carotid femoral pulse wave velocity (cfPWV) were carried out at 2- and 42-weeks post-ART initiation. Socioeconomic risk factors for immune activation and arterial stiffness were assessed using linear regression models. Results Of 279 PLWH, the median (IQR) age was 36 (31–43) years and 122 (44%) were female. Activated CD8 T-cells increased from 70% amongst those with no education to 88% amongst those with a tertiary education (p = 0.002); and from 71% amongst those earning less than 10 USD/month to 87% amongst those earning between 100–150 USD/month (p = 0.0001). Arterial stiffness was also associated with higher SES (car ownership p = 0.003, television ownership p = 0.012 and electricity access p = 0.029). Conversely, intermediate monocytes were higher amongst those with no education compared to a tertiary education (12.6% versus 7.3%; p = 0.01) and trended towards being higher amongst those earning less than 10 USD/month compared to 100–150 USD/month (10.5% versus 8.0%; p = 0.08). Water kiosk use showed a protective association against T cell activation (p = 0.007), as well as endothelial damage (MIP1β, sICAM1 and sVCAM1 p = 0.047, 0.026 and 0.031 respectively). Conclusions Socioeconomic risk factors for persistent inflammation amongst PLWH in SSA differ depending on the type of inflammatory pathway. Understanding these pathways and their socioeconomic drivers will help identify those at risk and target interventions for NCDs. Future studies assessing drivers of inflammation in HIV should include an SES assessment.
Collapse
Affiliation(s)
- Christine Kelly
- Cente for Experimental Pathogen Host Research (CEPHR), University College Dublin, Dublin, Ireland
- Malawi Liverpool Wellcome Trust Clinical Research Program, Blantyre, Malawi
- Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
- * E-mail:
| | - Willard Tinago
- Cente for Experimental Pathogen Host Research (CEPHR), University College Dublin, Dublin, Ireland
| | - Dagmar Alber
- Institute of Child Health, University College London, London, United Kingdom
| | - Patricia Hunter
- Institute of Child Health, University College London, London, United Kingdom
| | | | | | | | - Alejandro Garcia Abner
- Cente for Experimental Pathogen Host Research (CEPHR), University College Dublin, Dublin, Ireland
| | - Raphael Kamn’gona
- Malawi Liverpool Wellcome Trust Clinical Research Program, Blantyre, Malawi
| | - Irene Sheha
- Malawi Liverpool Wellcome Trust Clinical Research Program, Blantyre, Malawi
| | - Mishek Chammudzi
- Malawi Liverpool Wellcome Trust Clinical Research Program, Blantyre, Malawi
| | - Kondwani Jambo
- Malawi Liverpool Wellcome Trust Clinical Research Program, Blantyre, Malawi
| | - Jane Mallewa
- College of Medicine, University of Malawi, Blantyre, Malawi
| | - Alicja Rapala
- Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Patrick W. G. Mallon
- Cente for Experimental Pathogen Host Research (CEPHR), University College Dublin, Dublin, Ireland
| | - Henry Mwandumba
- Malawi Liverpool Wellcome Trust Clinical Research Program, Blantyre, Malawi
| | - Nigel Klein
- Institute of Child Health, University College London, London, United Kingdom
| | - Saye Khoo
- Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
22
|
Bourgeois C, Gorwood J, Olivo A, Le Pelletier L, Capeau J, Lambotte O, Béréziat V, Lagathu C. Contribution of Adipose Tissue to the Chronic Immune Activation and Inflammation Associated With HIV Infection and Its Treatment. Front Immunol 2021; 12:670566. [PMID: 34220817 PMCID: PMC8250865 DOI: 10.3389/fimmu.2021.670566] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 05/24/2021] [Indexed: 12/12/2022] Open
Abstract
White adipose tissue (AT) contributes significantly to inflammation – especially in the context of obesity. Several of AT’s intrinsic features favor its key role in local and systemic inflammation: (i) large distribution throughout the body, (ii) major endocrine activity, and (iii) presence of metabolic and immune cells in close proximity. In obesity, the concomitant pro-inflammatory signals produced by immune cells, adipocytes and adipose stem cells help to drive local inflammation in a vicious circle. Although the secretion of adipokines by AT is a prime contributor to systemic inflammation, the lipotoxicity associated with AT dysfunction might also be involved and could affect distant organs. In HIV-infected patients, the AT is targeted by both HIV infection and antiretroviral therapy (ART). During the primary phase of infection, the virus targets AT directly (by infecting AT CD4 T cells) and indirectly (via viral protein release, inflammatory signals, and gut disruption). The initiation of ART drastically changes the picture: ART reduces viral load, restores (at least partially) the CD4 T cell count, and dampens inflammatory processes on the whole-body level but also within the AT. However, ART induces AT dysfunction and metabolic side effects, which are highly dependent on the individual molecules and the combination used. First generation thymidine reverse transcriptase inhibitors predominantly target mitochondrial DNA and induce oxidative stress and adipocyte death. Protease inhibitors predominantly affect metabolic pathways (affecting adipogenesis and adipocyte homeostasis) resulting in insulin resistance. Recently marketed integrase strand transfer inhibitors induce both adipocyte adipogenesis, hypertrophy and fibrosis. It is challenging to distinguish between the respective effects of viral persistence, persistent immune defects and ART toxicity on the inflammatory profile present in ART-controlled HIV-infected patients. The host metabolic status, the size of the pre-established viral reservoir, the quality of the immune restoration, and the natural ageing with associated comorbidities may mitigate and/or reinforce the contribution of antiretrovirals (ARVs) toxicity to the development of low-grade inflammation in HIV-infected patients. Protecting AT functions appears highly relevant in ART-controlled HIV-infected patients. It requires lifestyle habits improvement in the absence of effective anti-inflammatory treatment. Besides, reducing ART toxicities remains a crucial therapeutic goal.
Collapse
Affiliation(s)
- Christine Bourgeois
- CEA - Université Paris Saclay - INSERM U1184, Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Jennifer Gorwood
- Sorbonne Université, INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-métabolisme et Nutrition (ICAN), FRM EQU201903007868, Paris, France
| | - Anaelle Olivo
- CEA - Université Paris Saclay - INSERM U1184, Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Fontenay-aux-Roses, France
| | - Laura Le Pelletier
- Sorbonne Université, INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-métabolisme et Nutrition (ICAN), FRM EQU201903007868, Paris, France
| | - Jacqueline Capeau
- Sorbonne Université, INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-métabolisme et Nutrition (ICAN), FRM EQU201903007868, Paris, France
| | - Olivier Lambotte
- CEA - Université Paris Saclay - INSERM U1184, Center for Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Fontenay-aux-Roses, France.,AP-HP, Groupe Hospitalier Universitaire Paris Saclay, Hôpital Bicêtre, Service de Médecine Interne et Immunologie Clinique, Le Kremlin-Bicêtre, France
| | - Véronique Béréziat
- Sorbonne Université, INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-métabolisme et Nutrition (ICAN), FRM EQU201903007868, Paris, France
| | - Claire Lagathu
- Sorbonne Université, INSERM UMR_S 938, Centre de Recherche Saint-Antoine, Institut Hospitalo-Universitaire de Cardio-métabolisme et Nutrition (ICAN), FRM EQU201903007868, Paris, France
| |
Collapse
|
23
|
Verhoeven F, Prati C, Chouk M, Demougeot C, Wendling D. Methotrexate and cardiovascular risk in rheumatic diseases:A comprehensive review. Expert Rev Clin Pharmacol 2021; 14:1105-1112. [PMID: 34006152 DOI: 10.1080/17512433.2021.1932461] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Introduction: Management of inflammatory rheumatic diseases has evolved based on improved treatment strategies and better management of comorbidities, specifically cardiovascular risk. Methotrexate is one of the first-line treatments in the management of inflammatory rheumatic diseases, but its cardiovascular effects are poorly understood. The purpose of this review is to assess the cardiovascular impact of methotrexate in inflammatory rheumatic disease.Areas covered: Current knowledge about the mechanism of action of methotrexate on cardiovascular tissue is presented. A review of the literature in the Medline, Cochrane and Embase databases was performed. Current data about the cardiovascular effects of methotrexate in rheumatoid arthritis, psoriatic arthritis, and psoriasis are presented.Expert opinion: Mechanism of action of methotrexate is based on the antagonism of purines. It reduces systemic inflammation and oxidative stress and improves the major cardiovascular risk factors. Methotrexate improves cardiovascular risk in rheumatoid arthritis, psoriasis and psoriatic arthritis, but the mechanisms involved are partially identified. Data are controversial regarding its effects on endothelial function and atherosclerosis. Conversely, in the general population and in patients with HIV infection, methotrexate does not modify cardiovascular outcomes. Thus, methotrexate only improves cardiovascular risk by reducing systemic inflammation, and should not be used to prevent cardiovascular events.
Collapse
Affiliation(s)
- Frank Verhoeven
- Department of Rheumatology, Service De Rhumatologie, CHRU De Besançon, Besancon, France.,EA 4267 : "PEPITE", FHU Increase, Université Bourgogne - Franche Comte, UFR Santé, Besancon, France
| | - Clément Prati
- Department of Rheumatology, Service De Rhumatologie, CHRU De Besançon, Besancon, France.,EA 4267 : "PEPITE", FHU Increase, Université Bourgogne - Franche Comte, UFR Santé, Besancon, France
| | - Mickaël Chouk
- Department of Rheumatology, Service De Rhumatologie, CHRU De Besançon, Besancon, France
| | - Céline Demougeot
- EA 4267 : "PEPITE", FHU Increase, Université Bourgogne - Franche Comte, UFR Santé, Besancon, France
| | - Daniel Wendling
- Department of Rheumatology, Service De Rhumatologie, CHRU De Besançon, Besancon, France.,EA 4266 : « EPILAB », Université Bourgogne - Franche Comte, UFR Santé, Besancon, France
| |
Collapse
|
24
|
A randomized, placebo-controlled, double-blinded clinical trial of colchicine to improve vascular health in people living with HIV. AIDS 2021; 35:1041-1050. [PMID: 33587443 PMCID: PMC8916096 DOI: 10.1097/qad.0000000000002845] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVES People living with HIV (PWH) experience an increased burden of coronary artery disease (CAD) believed to be related, in part, to an interplay of chronically increased inflammation and traditional risk factors. Recent trials suggest cardiovascular benefits of the anti-inflammatory, colchicine, in HIV-seronegative CAD patients. However, the impact of colchicine on impaired vascular health, as measured by coronary endothelial function (CEF), an independent contributor to CAD, has not been studied in PWH. We tested the hypothesis that colchicine improves vascular health in PWH. DESIGN This was a randomized, placebo-controlled, double-blinded trial in 81 PWH to test whether low-dose colchicine (0.6 mg daily) improves CEF over 8-24 weeks. METHODS Coronary and systemic endothelial function and serum inflammatory markers were measured at baseline, and at 8 and 24 weeks. The primary endpoint was CEF, measured as the change in coronary blood flow from rest to that during an isometric handgrip exercise, an endothelial-dependent stressor, measured with non-invasive MRI at 8 weeks. RESULTS Colchicine was well tolerated and not associated with increased adverse events. However, there were no significant improvements in coronary or systemic endothelial function or reductions in serum inflammatory markers at 8 or 24 weeks with colchicine as compared to placebo. CONCLUSIONS In PWH with no history of CAD, low-dose colchicine was well tolerated but did not improve impaired coronary endothelial function, a predictor of cardiovascular events. These findings suggest that this anti-inflammatory approach using colchicine in PWH does not improve vascular health, the central, early driver of coronary atherosclerosis.
Collapse
|
25
|
Durstenfeld MS, Hsue PY. Mechanisms and primary prevention of atherosclerotic cardiovascular disease among people living with HIV. Curr Opin HIV AIDS 2021; 16:177-185. [PMID: 33843806 PMCID: PMC8064238 DOI: 10.1097/coh.0000000000000681] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
PURPOSE OF REVIEW To highlight mechanisms of elevated risk of atherosclerotic cardiovascular disease (ASCVD) among people living with HIV (PLWH), discuss therapeutic strategies, and opportunities for primary prevention. RECENT FINDINGS HIV-associated ASCVD risk is likely multifactorial and due to HIV-specific factors and traditional risk factors even in the setting of treated and suppressed HIV disease. Although a growing body of evidence suggests that inflammation and immune activation are key drivers of atherogenesis, therapies designed to lower inflammation including colchicine and low-dose methotrexate have not improved secondary cardiovascular endpoints among PLWH. Statins continue to be the mainstay of management of hyperlipidemia in HIV, but the impact of newer lipid therapies including proprotein convertase subtilisin/kexin type 9 inhibitors on ASCVD risk among PLWH is under investigation. Aside from the factors mentioned above, healthcare disparities are particularly prominent among PLWH and thus likely contribute to increased ASCVD risk. SUMMARY Our understanding of mechanisms of elevated ASCVD risk in HIV continues to evolve, and the optimal treatment for CVD in HIV aside from targeting traditional risk factors remains unknown. Future studies including novel therapies to lower inflammation, control of risk factors, and implementation science are needed to ascertain optimal ways to treat and prevent ASCVD among PLWH.
Collapse
Affiliation(s)
- Matthew S Durstenfeld
- Division of Cardiology, UCSF at Zuckerberg San Francisco General Hospital
- Department of Medicine, University of California, San Francisco, California, USA
| | - Priscilla Y Hsue
- Division of Cardiology, UCSF at Zuckerberg San Francisco General Hospital
- Department of Medicine, University of California, San Francisco, California, USA
| |
Collapse
|
26
|
Kelly C, Tinago W, Alber D, Hunter P, Luckhurst N, Connolly J, Arrigoni F, Abner AG, Kamngona R, Sheha I, Chammudzi M, Jambo K, Mallewa J, Rapala A, Heyderman RS, Mallon PWG, Mwandumba H, Walker AS, Klein N, Khoo S. Inflammatory Phenotypes Predict Changes in Arterial Stiffness Following Antiretroviral Therapy Initiation. Clin Infect Dis 2021; 71:2389-2397. [PMID: 32103268 PMCID: PMC7713681 DOI: 10.1093/cid/ciaa186] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 02/24/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Inflammation drives vascular dysfunction in HIV, but in low-income settings causes of inflammation are multiple, and include infectious and environmental factors. We hypothesized that patients with advanced immunosuppression could be stratified into inflammatory phenotypes that predicted changes in vascular dysfunction on ART. METHODS We recruited Malawian adults with CD4 <100 cells/μL 2 weeks after starting ART in the REALITY trial (NCT01825031). Carotid femoral pulse-wave velocity (cfPWV) measured arterial stiffness 2, 12, 24, and 42 weeks post-ART initiation. Plasma inflammation markers were measured by electrochemiluminescence at weeks 2 and 42. Hierarchical clustering on principal components identified inflammatory clusters. RESULTS 211 participants with HIV grouped into 3 inflammatory clusters representing 51 (24%; cluster-1), 153 (73%; cluster-2), and 7 (3%; cluster-3) individuals. Cluster-1 showed markedly higher CD4 and CD8 T-cell expression of HLADR and PD-1 versus cluster-2 and cluster-3 (all P < .0001). Although small, cluster-3 had significantly higher levels of cytokines reflecting inflammation (IL-6, IFN-γ, IP-10, IL-1RA, IL-10), chemotaxis (IL-8), systemic and vascular inflammation (CRP, ICAM-1, VCAM-1), and SAA (all P < .001). In mixed-effects models, cfPWV changes over time were similar for cluster-2 versus cluster-1 (relative fold-change, 0.99; 95% CI, .86-1.14; P = .91), but greater in cluster-3 versus cluster-1 (relative fold-change, 1.45; 95% CI, 1.01-2.09; P = .045). CONCLUSIONS Two inflammatory clusters were identified: one defined by high T-cell PD-1 expression and another by a hyperinflamed profile and increases in cfPWV on ART. Further clinical characterization of inflammatory phenotypes could help target vascular dysfunction interventions to those at highest risk. CLINICAL TRIALS NETWORK NCT01825031.
Collapse
Affiliation(s)
- Christine Kelly
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Dublin, Ireland.,Malawi Liverpool Wellcome Trust Clinical Research Program, Blantyre, Malawi.,Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Willard Tinago
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Dublin, Ireland
| | - Dagmar Alber
- Institute of Child Health, University College London, London, United Kingdom
| | - Patricia Hunter
- Institute of Child Health, University College London, London, United Kingdom
| | | | | | | | - Alejandro Garcia Abner
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Dublin, Ireland
| | - Ralph Kamngona
- Malawi Liverpool Wellcome Trust Clinical Research Program, Blantyre, Malawi
| | - Irene Sheha
- Malawi Liverpool Wellcome Trust Clinical Research Program, Blantyre, Malawi
| | - Mishek Chammudzi
- Malawi Liverpool Wellcome Trust Clinical Research Program, Blantyre, Malawi
| | - Kondwani Jambo
- Malawi Liverpool Wellcome Trust Clinical Research Program, Blantyre, Malawi.,College of Medicine, University of Malawi, Blantyre, Malawi.,Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Jane Mallewa
- College of Medicine, University of Malawi, Blantyre, Malawi
| | - Alicja Rapala
- Institute of Cardiovascular Science, University College London, London, United Kingdom
| | - Robert S Heyderman
- Institute of Child Health, University College London, London, United Kingdom
| | - Patrick W G Mallon
- Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Dublin, Ireland
| | - Henry Mwandumba
- Malawi Liverpool Wellcome Trust Clinical Research Program, Blantyre, Malawi.,College of Medicine, University of Malawi, Blantyre, Malawi.,Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - A Sarah Walker
- 9MRC Clinical Trials Unit, University College London, London, United Kingdom
| | - Nigel Klein
- Institute of Child Health, University College London, London, United Kingdom
| | - Saye Khoo
- Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
27
|
Zhang Y, Jiang T, Li A, Li Z, Hou J, Gao M, Huang X, Su B, Wu H, Zhang T, Jiang W. Adjunct Therapy for CD4 + T-Cell Recovery, Inflammation and Immune Activation in People Living With HIV: A Systematic Review and Meta-Analysis. Front Immunol 2021; 12:632119. [PMID: 33679779 PMCID: PMC7925844 DOI: 10.3389/fimmu.2021.632119] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 01/22/2021] [Indexed: 01/21/2023] Open
Abstract
Background: HIV infection results in immune homeostasis perturbations, which is characterized by CD4+ T-cell depletion, immune activation, and inflammation. Effective antiretroviral therapy (ART) does not fully restore immunologic and clinical health in people living with HIV (PLWH). Various drugs have been used to improve their immune status and CD4+ T-cell counts, but no measures have been tested effective. Here we conduct a systematic review and meta-analysis of existing clinical studies on improving CD4+ T-cell count while decreasing inflammation and immune activation. Methods: We retrieved possible relevant publications from a total of five electronic databases and selected eligible studies, which dealt with outcomes of medical therapy for CD4+ T-cell count recovery, inflammation, and immune activation with or without ART. We paid particular attention to immunologic non-responders with a favorable treatment regimen. Results: Thirty-three articles were included in the systematic review and meta-analysis. However, there were no safe and effective medications specific for improving CD4+ T-cell reconstitution. The immunological benefits or adverse events mainly depend on the safety, dosage, and duration of the candidate medication use, as well as whether it is combined with ART. Conclusion: Under the “safe, combined, adequate and long (SCAL)” principles, alternative approaches are needed to accelerate the recovery of CD4+ T-cells, and to prevent adverse long-term outcomes in PLWH with standard ART treatment.
Collapse
Affiliation(s)
- Yang Zhang
- Center for Infectious Diseases, Beijing YouAn Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of AIDS Research, Beijing, China.,Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Taiyi Jiang
- Center for Infectious Diseases, Beijing YouAn Hospital, Capital Medical University, Beijing, China
| | - Aixin Li
- Center for Infectious Diseases, Beijing YouAn Hospital, Capital Medical University, Beijing, China
| | - Zhen Li
- Center for Infectious Diseases, Beijing YouAn Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of AIDS Research, Beijing, China
| | - Jianhua Hou
- Center for Infectious Diseases, Beijing YouAn Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of AIDS Research, Beijing, China
| | - Meixia Gao
- Center for Infectious Diseases, Beijing YouAn Hospital, Capital Medical University, Beijing, China
| | - Xiaojie Huang
- Center for Infectious Diseases, Beijing YouAn Hospital, Capital Medical University, Beijing, China
| | - Bin Su
- Center for Infectious Diseases, Beijing YouAn Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of AIDS Research, Beijing, China
| | - Hao Wu
- Center for Infectious Diseases, Beijing YouAn Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of AIDS Research, Beijing, China
| | - Tong Zhang
- Center for Infectious Diseases, Beijing YouAn Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of AIDS Research, Beijing, China
| | - Wei Jiang
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.,Division of Infectious Diseases, Department of Medicine, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
28
|
Bonou M, Kapelios CJ, Athanasiadi E, Mavrogeni SI, Psichogiou M, Barbetseas J. Imaging modalities for cardiovascular phenotyping in asymptomatic people living with HIV. Vasc Med 2021; 26:326-337. [PMID: 33475050 DOI: 10.1177/1358863x20978702] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cardiovascular disease (CVD) has emerged as a leading cause of non-HIV-related mortality among people living with HIV (PLWH). Despite the growing CVD burden in PLWH, there is concern that general population risk score models may underestimate CVD risk in these patients. Imaging modalities have received mounting attention lately to better understand the pathophysiology of subclinical CVD and provide improved risk assessment in this population. To date, traditional and well-established techniques such as echocardiography, pulse wave velocity, and carotid intima thickness continue to be the basis for the diagnosis and subsequent monitoring of vascular atherosclerosis and heart failure. Furthermore, novel imaging tools such as cardiac computed tomography (CT) and cardiac CT angiography (CCTA), positron emission tomography/CT (PET/CT), and cardiac magnetic resonance (CMR) have provided new insights into accelerated cardiovascular abnormalities in PLWH and are currently evaluated with regards to their potential to improve risk stratification.
Collapse
Affiliation(s)
- Maria Bonou
- Department of Cardiology Department, Laiko General Hospital, Athens, Greece
| | - Chris J Kapelios
- Department of Cardiology Department, Laiko General Hospital, Athens, Greece
| | - Eleni Athanasiadi
- Department of Cardiology Department, Laiko General Hospital, Athens, Greece
| | | | - Mina Psichogiou
- First Department of Internal Medicine, Laiko General Hospital, National and Kapodistrian University of Athens School of Medicine, Athens, Greece
| | - John Barbetseas
- Department of Cardiology Department, Laiko General Hospital, Athens, Greece
| |
Collapse
|
29
|
Stein JH, Kime N, Korcarz CE, Ribaudo H, Currier JS, Delaney JC. Effects of HIV Infection on Arterial Endothelial Function: Results From a Large Pooled Cohort Analysis. Arterioscler Thromb Vasc Biol 2021; 41:512-522. [PMID: 33327750 PMCID: PMC7770018 DOI: 10.1161/atvbaha.120.315435] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/26/2020] [Indexed: 12/26/2022]
Abstract
OBJECTIVE To determine the effects of HIV serostatus and disease severity on endothelial function in a large pooled cohort study of people living with HIV infection and HIV- controls. Approach and Results: We used participant-level data from 9 studies: 7 included people living with HIV (2 treatment-naïve) and 4 had HIV- controls. Brachial artery flow-mediated dilation (FMD) was measured using a standardized ultrasound imaging protocol with central reading. After data harmonization, multiple linear regression was used to examine the effects of HIV- serostatus, HIV disease severity measures, and cardiovascular disease risk factors on FMD. Of 2533 participants, 986 were people living with HIV (mean 44.4 [SD 11.8] years old) and 1547 were HIV- controls (42.9 [12.2] years old). The strongest and most consistent associates of FMD were brachial artery diameter, age, sex, and body mass index. The effect of HIV+ serostatus on FMD was strongly influenced by kidney function. In the highest tertile of creatinine (1.0 mg/dL), the effect of HIV+ serostatus was strong (β=-1.59% [95% CI, -2.58% to -0.60%], P=0.002), even after covariate adjustment (β=-1.36% [95% CI, -2.46% to -0.47%], P=0.003). In the lowest tertile (0.8 mg/dL), the effect of HIV+ serostatus was strong (β=-1.90% [95% CI, -2.58% to -1.21%], P<0.001), but disappeared after covariate adjustment. HIV RNA viremia, CD4+ T-cell count, and use of antiretroviral therapy were not meaningfully associated with FMD. CONCLUSIONS The significant effect of HIV+ serostatus on FMD suggests that people living with HIV are at increased cardiovascular disease risk, especially if they have kidney disease.
Collapse
Affiliation(s)
- James H. Stein
- University of Wisconsin School of Medicine and Public Health; Madison, WI
| | - Noah Kime
- University of Washington Collaborative Health Studies Coordinating Center, Seattle, WA
| | - Claudia E. Korcarz
- University of Wisconsin School of Medicine and Public Health; Madison, WI
| | | | - Judith S. Currier
- David Geffen School of Medicine at University of California -Los Angeles; Los Angeles, CA
| | - Joseph C. Delaney
- University of Washington Collaborative Health Studies Coordinating Center, Seattle, WA
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba; Winnipeg, MB
| |
Collapse
|
30
|
Gingrich D, Deitchman AN, Kantor A, Huang L, Stein JH, Currier JS, Hsue PY, Ribaudo HJ, Aweeka FT. Methotrexate Decreases Tenofovir Exposure in Antiretroviral-Suppressed Individuals Living With HIV. J Acquir Immune Defic Syndr 2020; 85:651-658. [PMID: 33177476 PMCID: PMC8132144 DOI: 10.1097/qai.0000000000002502] [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: 11/26/2022]
Abstract
BACKGROUND To mitigate increased risk of premature cardiovascular disease in antiretroviral therapy (ART) suppressed adults living with HIV (PWH), low-dose methotrexate (LDMTX) was evaluated in a multicenter randomized placebo controlled clinical trial of 176 PWH taking various ART regimens (ACTG A5314). Given shared methotrexate (MTX) and tenofovir (TFV) pharmacokinetic (PK) pathways, a substudy was conducted to investigate whether LDMTX alters TFV exposure. METHODS Adults virally suppressed on ART for >24 weeks were randomized to LDMTX or placebo. The first 66 participants taking a tenofovir disoproxil fumarate-containing regimen underwent intensive PK sampling over 24 hours after the second dose of LDMTX 10 mg or placebo. TFV and MTX levels were quantified using validated mass spectrometry methods. TFV PK between LDMTX and placebo groups were compared and MTX PK was characterized. RESULTS Forty-eight participants completed this substudy (n = 20 on LDMTX and 28 on placebo). Baseline characteristics were balanced except for protease inhibitor (PI)-use (25% in LDMTX and 43% in placebo groups). For TFV, AUC6 (primary endpoint), and AUC24,imputed, Cmax, and Cmin (secondary endpoints) were on average 22%, and 24%, 27%, and 31% less in the LDMTX versus placebo groups, with reductions in secondary endpoints reaching statistical significance. Additional analyses suggested a greater reduction in the absence of PI although not significant. CONCLUSION Lower TFV AUC24,imputed and Cmax indicates that LDMTX reduces TFV exposure in PWH. However, this change was modest, not warranting a change in TFV dosing at this time. Further studies of TFV PK with LDMTX, especially without PI co-administration, are warranted.
Collapse
Affiliation(s)
- David Gingrich
- Drug Research Unit, Department of Clinical Pharmacy University of California, San Francisco, CA 94110
| | - Amelia N Deitchman
- Drug Research Unit, Department of Clinical Pharmacy University of California, San Francisco, CA 94110
| | - Amy Kantor
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Liusheng Huang
- Drug Research Unit, Department of Clinical Pharmacy University of California, San Francisco, CA 94110
| | - James H Stein
- University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Judith S Currier
- David Geffen School of Medicine at University of California – Los Angeles; Los Angeles, CA
| | | | - Heather J Ribaudo
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Francesca T Aweeka
- Drug Research Unit, Department of Clinical Pharmacy University of California, San Francisco, CA 94110
| | | |
Collapse
|
31
|
Blanco JR, Negredo E, Bernal E, Blanco J. Impact of HIV infection on aging and immune status. Expert Rev Anti Infect Ther 2020; 19:719-731. [PMID: 33167724 DOI: 10.1080/14787210.2021.1848546] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Introduction: Thanks to antiretroviral therapy (ART), persons living with HIV (PLWH), have a longer life expectancy. However, immune activation and inflammation remain elevated, even after viral suppression, and contribute to morbidity and mortality in these individuals.Areas covered: We review aspects related to immune activation and inflammation in PLWH, their consequences, and the potential strategies to reduce immune activation in HIV-infected individuals on ART.Expert opinion: When addressing a problem, it is necessary to thoroughly understand the topic. This is the main limitation faced when dealing with immune activation and inflammation in PLWH since there is no consensus on the ideal markers to evaluate immune activation or inflammation. To date, the different interventions that have addressed this problem by targeting specific mediators have not been able to significantly reduce immune activation or its consequences. Given that there is currently no curative intervention for HIV infection, more studies are necessary to understand the mechanism underlying immune activation and help to identify potential therapeutic targets that contribute to improving the life expectancy of HIV-infected individuals.
Collapse
Affiliation(s)
- Jose-Ramon Blanco
- Servicio de Enfermedades Infecciosas, Hospital Universitario San Pedro- Centro De Investigación Biomédica De La Rioja (CIBIR), La Rioja, Spain
| | - Eugenia Negredo
- Lluita Contra La Sida Foundation, Germans Trias I Pujol University Hospital, Badalona, Spain. Centre for Health and Social Care Research (CESS), Faculty of Medicine, University of Vic - Central University of Catalonia (Uvic - UCC), Catalonia, Spain
| | - Enrique Bernal
- Unidad De Enfermedades Infecciosas, Hospital General Universitario Reina Sofía, Universidad De Murcia, Murcia, Spain
| | - Juliá Blanco
- AIDS Research Institute-IrsiCaixa, Badalona, Barcelona, Spain.,Universitat De Vic-Central De Catalunya (UVIC-UCC), Vic, Spain
| |
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: 1.8] [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
|
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: 5.4] [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
|
34
|
Erlandson KM, Wilson MP, MaWhinney S, Rapaport E, Liu J, Wilson CC, Rahkola JT, Janoff EN, Brown TT, Campbell TB, Jankowski CM. The Impact of Moderate or High-Intensity Combined Exercise on Systemic Inflammation Among Older Persons With and Without HIV. J Infect Dis 2020; 223:1161-1170. [PMID: 32779711 DOI: 10.1093/infdis/jiaa494] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 07/31/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND We investigated whether higher-intensity exercise provided greater decrease in markers of inflammation, and whether responses differed by HIV serostatus. METHODS People with HIV (PWH; n = 32) and controls (n = 37) aged 50-75 years completed 12 weeks moderate-intensity exercise, then were randomized to moderate- or high-intensity exercise for 12 additional weeks (n = 27 and 29, respectively). Inflammation biomarkers were measured at 0, 12, 24 weeks. Mixed and multiple regression models were adjusted for baseline inflammation, age, and body mass index. RESULTS Baseline tumor necrosis factor-α (TNF-α), soluble TNF receptor 2 (sTNFR2), and soluble CD14 (sCD14) were significantly higher among PWH than controls (P < .04). From week 0-12, changes in interleukin-6 (IL-6), TNF-α, and sTNFR1 were not significantly different by HIV serostatus. We found no significant interaction between HIV serostatus/exercise intensity on week 12-24 changes in IL-6, TNF-α, and sTNFR1. Among high-intensity exercisers, PWH and controls had significant increases in sCD14 (P ≤ .003), controls significant increases in IL-10 (P = .01), and PWH nonsignificant decrease in highly sensitive C-reactive protein (P = .07). Other markers were not significantly different by serostatus or intensity. CONCLUSIONS Moderate and high-intensity exercise elicited similar effects on inflammation among PWH and controls, with additional beneficial effects seen among high-intensity exercisers. Increase in sCD14 and attenuated IL-10 increase (PWH only) merit further study. CLINICAL TRIALS REGISTRATION NCT02404792.
Collapse
Affiliation(s)
- Kristine M Erlandson
- Department of Medicine, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Melissa P Wilson
- School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Samantha MaWhinney
- School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Eric Rapaport
- Department of Medicine, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jay Liu
- Department of Medicine, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Cara C Wilson
- Department of Medicine, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jeremy T Rahkola
- Department of Medicine, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Edward N Janoff
- Department of Medicine, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado, USA
| | - Todd T Brown
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism, Johns Hopkins University, Baltimore, Maryland, USA
| | - Thomas B Campbell
- Department of Medicine, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Catherine M Jankowski
- College of Nursing, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
35
|
Premeaux TA, Javandel S, Hosaka KRJ, Greene M, Therrien N, Allen IE, Corley MJ, Valcour VG, Ndhlovu LC. Associations Between Plasma Immunomodulatory and Inflammatory Mediators With VACS Index Scores Among Older HIV-Infected Adults on Antiretroviral Therapy. Front Immunol 2020; 11:1321. [PMID: 32695109 PMCID: PMC7338430 DOI: 10.3389/fimmu.2020.01321] [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] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 05/26/2020] [Indexed: 12/13/2022] Open
Abstract
The prevalence of age-related comorbidities is increased in people living with HIV, even in those well-controlled on combination antiretroviral therapy (ART). Persistent immune activation and inflammation may play pivotal roles in the pathogenesis; however, the burden of morbidities in the older HIV infected population may be exacerbated and driven by distinct mechanisms. In a cross sectional study of 45 HIV-infected participants 60 years or older, we examined the relationships between 14 immunomodulatory and inflammatory factors and the Veterans Aging Cohort Study (VACS) Index, a metric of multimorbidity and mortality comprised of age, CD4 count, hemoglobin, Fibrosis-4 [FIB-4], and estimated glomerular filtration rate [eGFR], by linear regression analysis. All participants were virally suppressed (<50 HIV RNA copies/mL), on ART, and primarily Caucasian (86.7%), and male (91.1%). Plasma levels of monocyte/macrophage-associated (neopterin, IP-10, sCD163, sCD14, and MCP-1) and glycan-binding immunomodulatory factors (galectin (Gal)-1, Gal-3, and Gal-9) were assessed, as well as inflammatory biomarkers previously linked to the VACS Index (i.e., CRP, cystatin C, TNF-α, TNFRI, IL-6, and D-dimer) for comparison. In regression analysis, higher VACS index scores were associated with higher levels of neopterin, cystatin C, TNFRI, and Gal-9 (all p < 0.05), potentially driven by correlations found with individual VACS components, including age, CD4 count, FIB-4, and eGFR. Gal-9, cystatin C, and TNFRI directly correlated with the extent of multimorbidity. Multiple correlations among markers were observed, suggesting an interplay of overlapping, but distinct, pathways. Collectively, in addition to cystatin C and TNFRI, both galectin-9 and neopterin, independently emerged as novel fluid markers of the VACS Index and burden of comorbidity and may further guide in understanding pathogenic mechanisms of age-related disorders in older HIV-infected individuals on suppressive ART.
Collapse
Affiliation(s)
- Thomas A Premeaux
- Department of Tropical Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
| | - Shireen Javandel
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, United States
| | - Kalei R J Hosaka
- Department of Tropical Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
| | - Meredith Greene
- Division of Geriatric Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Nicholas Therrien
- Department of Tropical Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States
| | - Isabel E Allen
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, United States
| | - Michael J Corley
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Victor G Valcour
- Department of Neurology, Memory and Aging Center, University of California, San Francisco, San Francisco, CA, United States.,Division of Geriatric Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Lishomwa C Ndhlovu
- Department of Tropical Medicine, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI, United States.,Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
36
|
Mullis C, Swartz TH. NLRP3 Inflammasome Signaling as a Link Between HIV-1 Infection and Atherosclerotic Cardiovascular Disease. Front Cardiovasc Med 2020; 7:95. [PMID: 32596261 PMCID: PMC7301651 DOI: 10.3389/fcvm.2020.00095] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 05/06/2020] [Indexed: 01/06/2023] Open
Abstract
36.9 million people worldwide are living with HIV-1. The disease remains incurable and HIV-infected patients have increased risk of atherosclerosis. Inflammation is a key driver of atherosclerosis, but no targeted molecular therapies have been developed to reduce cardiovascular risk in people with HIV-1 (PWH). While the mechanism is unknown, there are several important inflammatory signaling events that are implicated in the development of chronic inflammation in PWH and in the inflammatory changes that lead to atherosclerosis. Here we describe the pro-inflammatory state of HIV-1 infection that leads to increased risk of cardiovascular disease, the role of the NLR Family Pyrin Domain Containing 3 (NLRP3) inflammasome in HIV-1 infection, the role of the NLRP3 inflammasome in cardiovascular disease (CVD), and outline a model whereby HIV-1 infection can lead to atherosclerotic disease through NLRP3 inflammasome activation. Our discussion highlights the literature supporting HIV-1 infection as a stimulator of the NLRP3 inflammasome as a driver of atherosclerosis.
Collapse
Affiliation(s)
- Caroline Mullis
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Talia H Swartz
- Division of Infectious Diseases, Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
37
|
McGettrick P, Mallon PWG, Sabin CA. Cardiovascular disease in HIV patients: recent advances in predicting and managing risk. Expert Rev Anti Infect Ther 2020; 18:677-688. [PMID: 32306781 DOI: 10.1080/14787210.2020.1757430] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Cardiovascular disease (CVD) is one of the leading causes of mortality in virally suppressed people living with HIV (PLWH) and with an aging population, is likely to become one of the leading challenges in maintaining good health outcomes in HIV infection. However, factors driving the risk of CVD in PLWH are multiple and may be different from those of the general population, raising challenges to predicting and managing CVD risk in this population. AREAS COVERED In this review, we examine the relevant data regarding CVD in HIV infection including CVD prevalence, pathogenesis, and other contributing factors. We review the data regarding CVD risk prediction in PLWH and summarize factors, both general and HIV specific, that may influence CVD risk in this population. And finally, we discuss appropriate management of CVD risk in PLWH and explore potential therapeutic pathways which may mitigate CVD risk in the future in this population. EXPERT OPINION Following a comprehensive review of CVD risk in PLWH, we give our opinion on the primary issues in risk prediction and management of CVD in HIV infected individuals and discuss the future direction of CVD management in this population.
Collapse
Affiliation(s)
- Padraig McGettrick
- Centre for Pathogen Host Research, UCD School of Medicine, University College Dublin , Dublin, Ireland
| | - Patrick W G Mallon
- Centre for Pathogen Host Research, UCD School of Medicine, University College Dublin , Dublin, Ireland.,Department of Infectious Diseases, St. Vincent's University Hospital , Dublin, Ireland
| | - Caroline A Sabin
- Centre for Clinical Research, Epidemiology, Modelling and Evaluation, Institute for Global Health, University College London , London, UK
| |
Collapse
|
38
|
Nguyen I, Kim AS, Chow FC. Prevention of stroke in people living with HIV. Prog Cardiovasc Dis 2020; 63:160-169. [PMID: 32014514 PMCID: PMC7237326 DOI: 10.1016/j.pcad.2020.01.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 01/29/2020] [Indexed: 12/13/2022]
Abstract
In the era of effective antiretroviral therapy (ART), HIV has become a manageable disease marked by an elevated risk of non-AIDS-related comorbidities, including stroke. Rates of stroke are higher in people living with HIV (PLWH) compared with the general population. Elevated stroke risk may be attributable to traditional risk factors, HIV-associated chronic inflammation and immune dysregulation, and possible adverse effects of long-standing ART use. Tailoring stroke prevention strategies for PLWH requires knowledge of how stroke pathogenesis may differ from non-HIV-associated stroke, knowledge of long-term stroke outcomes in HIV, and accurate stroke risk assessment tools. As a result, the approach to primary and secondary stroke prevention in PLWH relies heavily on guidelines developed for the general population, with an emphasis on optimization of traditional vascular risk factors and early initiation of ART. This review summarizes existing evidence on HIV-associated stroke mechanisms and considerations for stroke prevention for PLWH.
Collapse
Affiliation(s)
- Ivy Nguyen
- Department of Neurology, University of California, San Francisco, CA, United States of America
| | - Anthony S Kim
- Department of Neurology, University of California, San Francisco, CA, United States of America
| | - Felicia C Chow
- Department of Neurology, University of California, San Francisco, CA, United States of America; Department of Medicine, Division of Infectious Diseases, University of California San Francisco, CA, United States of America.
| |
Collapse
|
39
|
Titanji B, Gavegnano C, Hsue P, Schinazi R, Marconi VC. Targeting Inflammation to Reduce Atherosclerotic Cardiovascular Risk in People With HIV Infection. J Am Heart Assoc 2020; 9:e014873. [PMID: 31973607 PMCID: PMC7033865 DOI: 10.1161/jaha.119.014873] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Boghuma Titanji
- Division of Infectious Diseases Emory University School of Medicine Atlanta GA
| | - Christina Gavegnano
- Center for AIDS Research Laboratory of Biochemical Pharmacology Department of Pediatrics Emory University Atlanta GA
| | - Priscilla Hsue
- Department of Cardiology Zuckerberg San Francisco General Hospital University of California-San Francisco CA
| | - Raymond Schinazi
- Center for AIDS Research Laboratory of Biochemical Pharmacology Department of Pediatrics Emory University Atlanta GA
| | - Vincent C Marconi
- Division of Infectious Diseases Emory University School of Medicine Atlanta GA.,Emory Vaccine Center Atlanta GA.,Rollins School of Public Health Emory University Atlanta GA.,Atlanta VA Medical Center Decatur GA
| |
Collapse
|
40
|
Lipinski J, Margevicius S, Schluchter MD, Wilson DL, McComsey GA, Longenecker CT. Statin effect on coronary calcium distribution, mass and volume scores and associations with immune activation among HIV+ persons on antiretroviral therapy. Antivir Ther 2020; 25:419-427. [PMID: 33901017 DOI: 10.3851/imp3389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2020] [Indexed: 10/21/2022]
Abstract
BACKGROUND Inflammation has been associated with whole heart coronary artery calcification (CAC) among people with HIV (PWH) on antiretroviral therapy (ART); however, prior studies have not evaluated the distribution of calcium or separated mass versus volume scores, which are differentially associated with clinical events in the general population. Statins may also have a greater effect on CAC mass compared with volume. METHODS 147 PWH were randomized 1:1 to rosuvastatin 10 mg or placebo and followed for 96 weeks. We re-analysed coronary calcium scans from 0, 48 and 96 weeks to determine mass and volume scores and measures of CAC diffusivity. Mixed effects models and generalized estimating equations were used to examine longitudinal associations of CAC with treatment and biomarkers. RESULTS Median age at study entry was 46 years; 78% were male and 68% African American. Median CD4+ was 613 and half were on protease inhibitors. Randomization to statin therapy was not associated with a change in mass score, volume score, number of involved vessels or diffusivity index (all P>0.1). Soluble CD14 was associated with the presence of CAC (P=0.05) and borderline associated with number of involved vessels (P=0.07) across all three time points. CONCLUSIONS In PWH on ART, moderate intensity rosuvastatin does not appear to have a significant effect on volume, mass or regional distribution of CAC over 96 weeks. We extend previous cross-sectional observations to show that soluble CD14 is associated with whole heart CAC over time and independently of age and systolic blood pressure.
Collapse
Affiliation(s)
- Jerry Lipinski
- Department of Internal Medicine, University of California, San Diego, San Diego, CA, USA
| | | | - Mark D Schluchter
- School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - David L Wilson
- Case Western Reserve University School of Engineering, Cleveland, OH, USA
| | - Grace A McComsey
- School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Chris T Longenecker
- School of Medicine, Case Western Reserve University, Cleveland, OH, USA
- University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| |
Collapse
|
41
|
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
|
42
|
Stein JH, Yeh E, Weber JM, Korcarz C, Ridker PM, Tawakol A, Hsue PY, Currier JS, Ribaudo H, Mitchell CKC. Brachial Artery Echogenicity and Grayscale Texture Changes in HIV-Infected Individuals Receiving Low-Dose Methotrexate. Arterioscler Thromb Vasc Biol 2019; 38:2870-2878. [PMID: 30571173 DOI: 10.1161/atvbaha.118.311807] [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] [Indexed: 02/06/2023]
Abstract
Objective- We evaluated the biological effects of low-dose methotrexate on 3 novel brachial artery grayscale ultrasound measures that may indicate subclinical arterial injury. Approach and Results- Exploratory analysis from a clinical trial of people with HIV infection at increased cardiovascular disease risk who were randomly assigned to low-dose methotrexate (target dose 15 mg/wk) or placebo. Brachial artery ultrasound grayscale median, gray level difference statistic texture-contrast (GLDS-CON), and gray level texture entropy were measured at baseline and after 24 weeks of intervention. Findings from the intention-to-treat (N=148) and adequately-dosed (N=118) populations were consistent, so the adequately-dosed population results are presented. Participants were a median (Q1, Q3) age of 54 (50, 60) years. After 24 weeks, the low-dose methotrexate intervention was associated with a 25.4% (-18.1, 58.6; P=0.007) increase in GLDS-CON compared with 1.3% (-29.1, 44.7; P=0.97) with placebo ( P=0.05) and a 0.10 u (-0.06, 0.23; P=0.026) increase in entropy compared with 0.02 u (-0.11, 0.14; P=0.54) with placebo ( P=0.14). At week 24, changes in CD4+ T cells correlated inversely with changes in GLDS-CON (ρ=-0.20; P=0.031), and entropy (ρ=-0.21; P=0.023). Changes in D-dimer levels, but no other inflammatory biomarkers, also correlated inversely with changes in GLDS-CON (ρ=-0.23; P=0.014) and entropy (ρ=-0.26; P=0.005). Conclusions- Brachial artery GLDS-CON and entropy increased after 24 weeks of low-dose methotrexate, though the latter was not significantly different from placebo. Grayscale changes were associated with decreases in CD4+ T-cell and D-dimer concentrations and may indicate favorable arterial structure changes.
Collapse
Affiliation(s)
- James H Stein
- From the Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison (J.H.S., J.M.W., C.K., C.K.C.M.)
| | - Eunice Yeh
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, MA (E.Y., H.R.)
| | - Joanne M Weber
- From the Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison (J.H.S., J.M.W., C.K., C.K.C.M.)
| | - Claudia Korcarz
- From the Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison (J.H.S., J.M.W., C.K., C.K.C.M.)
| | - Paul M Ridker
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston (P.M.R., A.T.)
| | - Ahmed Tawakol
- Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Boston (P.M.R., A.T.)
| | - Priscilla Y Hsue
- Department of Medicine, University of California-San Francisco School of Medicine (P.Y.H.)
| | - Judith S Currier
- Division of Infectious Diseases, David Geffen School of Medicine at University of California-Los Angeles (J.S.C.)
| | - Heather Ribaudo
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, MA (E.Y., H.R.)
| | - Carol K C Mitchell
- From the Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison (J.H.S., J.M.W., C.K., C.K.C.M.)
| |
Collapse
|
43
|
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: 421] [Impact Index Per Article: 70.2] [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
|
44
|
Lawal IO, Ankrah AO, Stoltz AC, Sathekge MM. Radionuclide imaging of inflammation in atherosclerotic vascular disease among people living with HIV infection: current practice and future perspective. Eur J Hybrid Imaging 2019; 3:5. [PMID: 34191183 PMCID: PMC8218042 DOI: 10.1186/s41824-019-0053-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/11/2019] [Indexed: 01/03/2023] Open
Abstract
People living with human immunodeficiency virus (HIV) infection have twice the risk of atherosclerotic vascular disease compared with non-infected individuals. Inflammation plays a critical role in the development and progression of atherosclerotic vascular disease. Therapies targeting inflammation irrespective of serum lipid levels have been shown to be effective in preventing the occurrence of CVD. Radionuclide imaging is a viable method for evaluating arterial inflammation. This evaluation is useful in quantifying CVD risk and for assessing the effectiveness of anti-inflammatory treatment. The most tested radionuclide method for quantifying arterial inflammation among people living with HIV infection has been with F-18 FDG PET/CT. The level of arterial uptake of F-18 FDG correlates with vascular inflammation and with the risk of development and progression of atherosclerotic disease. Several limitations exist to the use of F-18 FDG for PET quantification of arterial inflammation. Many targets expressed on macrophage, a significant player in arterial inflammation, have the potential for use in evaluating arterial inflammation among people living with HIV infection. The review describes the clinical utility of F-18 FDG PET/CT in assessing arterial inflammation as a risk for atherosclerotic disease among people living with HIV infection. It also outlines potential newer probes that may quantify arterial inflammation in the HIV-infected population by targeting different proteins expressed on macrophages.
Collapse
Affiliation(s)
- Ismaheel O. Lawal
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Private Bag X169, Pretoria, 0001 South Africa
| | - Alfred O. Ankrah
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Private Bag X169, Pretoria, 0001 South Africa
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen & University of Groningen, Groningen, The Netherlands
| | - Anton C. Stoltz
- Infectious Disease Unit, Department of Internal Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria, South Africa
| | - Mike M. Sathekge
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Private Bag X169, Pretoria, 0001 South Africa
| |
Collapse
|
45
|
Mechanisms of Cardiovascular Disease in the Setting of HIV Infection. Can J Cardiol 2018; 35:238-248. [PMID: 30825947 DOI: 10.1016/j.cjca.2018.12.024] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 12/11/2018] [Accepted: 12/11/2018] [Indexed: 12/12/2022] Open
Abstract
Although the initial reports of increased cardiovascular (CV) disease in the setting of advanced AIDS were reported approximately 30 years ago, advances in antiretroviral therapy and immediate initiation of therapy on diagnosis have transformed what was once a deadly infectious disease into a chronic health condition. Accordingly, the types of CV diseases occurring in HIV have shifted from pericardial effusions and dilated cardiomyopathy to atherosclerosis and heart failure. The underlying pathophysiology of HIV-associated CV disease remains poorly understood, partly because of the rapidly evolving nature of HIV treatment and because clinical endpoints take many years to develop. The gut plays an important role in the early pathogenesis of HIV infection as HIV preferentially infects CD4+ T cells, 80% of which are located in gut mucosa. The loss of these T cells damages gut mucosa resulting in increased gut permeability and microbial translocation, which incites chronic inflammation and immune activation. Antiretroviral therapy does not cure HIV infection and immune abnormalities persist. These abnormalities correlate with mortality and CV events. The effects of antiretroviral therapy on CV risk are complex; treatment reduces inflammation and other markers of CV risk but induces lipid abnormalities, most commonly hypertriglyceridemia. On a molecular level, monocytes/macrophages, platelet reactivity, and immune cell activation, which play a role in the general population, may be heightened in the setting of HIV and contribute to HIV-associated atherosclerosis. Chronic inflammation represents an inviting therapeutic target in HIV, as it does in uninfected persons with atherosclerosis.
Collapse
|