Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Nov 28.
Published in final edited form as: AIDS. 2012 Nov 28;26(18):2409–2412. doi: 10.1097/QAD.0b013e32835a9950

Increased Coronary Atherosclerosis and Immune Activation in HIV-1 Elite Controllers

Florencia Pereyra 1,*, Janet Lo 1,*, Virginia A Triant 1, Jeffrey Wei 1, Maria J Buzon 1, Kathleen V Fitch 1, Janice Hwang 1, Jennifer H Campbell 1, Tricia H Burdo 1, Kenneth C Williams 1, Suhny Abbara 1, Steven K Grinspoon 1
PMCID: PMC3660105  NIHMSID: NIHMS471827  PMID: 23032411

Summary

HIV-1 elite controllers (EC) spontaneously maintain suppressed levels of viremia, but exhibit significant immune activation. We investigated coronary atherosclerosis by coronary CT angiography (CTA) in: 1) EC, 2) non EC, chronically HIV-1 infected, ART-treated patients with undetectable viral load (“chronic HIV”), and 3) HIV-negative controls. Prevalence of atherosclerosis (78% vs. 42%, P<0.05) and markers of immune activation were increased in EC compared to HIV-negative controls. sCD163, a monocyte activation marker, was increased in EC compared to chronic HIV-1 (P<0.05) and compared to HIV-negative controls (P< 0.05). These data suggest a significant degree of coronary atherosclerosis and monocyte activation among EC.

Introduction

Recent data suggest that immune activation – even among virologically suppressed patients – may contribute to increased atherosclerotic disease in HIV-infected patients[1, 2]. HIV-1 elite controllers (EC) represent an ideal population to explore the potential role played by longstanding immune activation in atherosclerotic disease, without the confounding effects of high level viral replication or ART. Despite tight control of viral replication, residual low-level viremia in EC is associated with persistent T-cell activation and inflammation[3-5]. In contrast, little is known about monocyte/ macrophage activation in EC and direct investigation of coronary atherosclerosis has not been performed in this population.

Methods

Ten HIV-1 EC were identified from the International HIV Controllers Study[6, 7], with approximately 400 controllers, of whom less than half had no history of viral blips and/or ART use. ART-naïve subjects, ages 40-60, with HIV-1 viral load below the detection limit of an ultrasensitive assay (<48 copies/mL), and no history of “viral blips”, CVD or renal disease were chosen. Subjects underwent assessment of CTA and coronary artery calcium (CAC)[8]. Viral reservoir was measured by cell associated HIV-1 DNA[9, 10]. Comparison was made to data previously obtained in non EC, chronic HIV-1 patients, receiving ART, with undetectable HIV viral load (“chronic HIV”) and HIV-negative controls[1, 8]. Subjects in the two comparison groups were similar to the EC with respect to age and lack of known CVD. All participants gave informed consent to participate. This study was approved by the Massachusetts General Hospital IRB.

Data were compared between the groups using ANOVA/Kruskal-Wallis test depending on normality. Pairwise comparisons were performed using Student's T-test or the Wilcoxon test for variables significant in overall ANOVA. Logistic regression was used to control for traditional CVD risk factors (SAS JMP).

Results

EC were similar in age, gender and traditional CVD risk factors, including smoking and Framingham score, to the two comparison groups (Table). Duration of HIV diagnosis was 15 or more years for both EC and chronic HIV-1 patients. Longitudinal plasma viral loads spanning a median of 8 years were obtained in EC. HIV reservoir, measured by copies per CD4 cell, of total HIV-1 DNA (median 1.4 × 10-4), 2-LTR circles (median undetectable), and integrated HIV-1 DNA (median 4.0 × 10-6) were low, consistent with previous data in EC[10]. Among chronic HIV-1, ART duration was 8.5 years.

Table 1.

HIV-1 Elite Controllers (n=10) Chronic HIV-1 (n=103) HIV-negative (n=49) P-value
Demographics
Age (yrs) 52.2 ± 5.1a,b 49.1 ± 5.0 48.2 ± 4.5 0.06
Gender (%male) 80% 69% 67% 0.71
Duration since HIV Diagnosis (yrs) 17.8 ± 9.1 14.8 ± 6.2 N/A 0.16
Currently on ART (%) 0% 100% N/A N/A
Duration of ART (yrs) 0 ± 0 8.5 ± 4.4 N/A N/A
Percent on PI 0% 55% N/A N/A
Duration of PI (yrs) 0 ± 0 4.4 ± 4.5 N/A N/A
Percent on NRTI 0% 95% N/A N/A
Duration of NRTI (yrs) 0 ± 0 8.2 ± 4.5 N/A N/A
Percent on NNRTI 0% 39% N/A N/A
Duration of NNRTI (yrs) 0 ± 0 3.0 ± 4.0 N/A N/A
CD4+ count (cells/μL) 934 ± 513 571 ± 281 N/A 0.0005
Nadir CD4+ count (cells/μL) 582 ± 372 186 ±160 N/A < 0.0001
HIV Viral load (copies/mL) <48 [<48, <48] <50 [<50, <50] N/A 0.006*
Undetectable VL (%) 100% 100% N/A N/A
CMV IgG Ab Test (% positive) 89% 92%a 71% 0.004
Cardiovascular Risk Factors
BMI (kg/m2) 28.6 ± 5.1 27.1 ± 4.7 27.8 ± 4.8 0.44
Framingham risk score (total points) 10.7 ± 3.5 9.6 ± 3.0 9.0 ± 4.0 0.34
Total cholesterol (mg/dL) 182 ± 50 185 ± 41 182 ± 37 0.85
Triglycerides (mg/dL) 110 ± 66 143 ± 122 111 ± 68 0.18
HDL (mg/dL) 54 ± 15 54 ± 18 51 ± 13 0.55
LDL (mg/dL) 106 ± 44 103 ± 31 109 ± 31 0.55
Fasting Glucose (mg/dL) 92 ± 19 94 ± 29 88 ± 12 0.45
SBP (mm Hg) 119 ± 13 121 ± 14 117 ± 15 0.36
WHR 0.95 ± 0.05 0.95 ± 0.07 0.93 ± 0.07 0.66
HTN (% prevalence) 30% 26% 16% 0.37
Diabetes (% prevalence) 10% 12% 4% 0.26
Statin Use (% prevalence) 10% 21%a 6% 0.11
Current Smoker (% prevalence) 40% 42% 41% 0.98
CT Angiography and Coronary Artery Calcium
Presence of coronary plaque (% prevalence) 78%a 60%a 42% 0.049
Total Plaque Segments 2.5 [0.3, 5.8] 1 [0, 3] 0 [0, 3] 0.14*
Calcium Score 16 [0, 96] 0 [0, 16] 0 [0, 30] 0.15*
Calcium Score >0 (% prevalence) 70% 41% 34% 0.11
Non-Calcified Segments 0.5 [0, 2] 0 [0, 2]a 0 [0, 1] 0.07*
Mixed Calcified and Non-Calcified Segments 1 [0, 2.8] 0 [0, 1] 0 [0, 2] 0.27*
Calcified Segments 0 [0, 1] 0 [0, 0] 0 [0, 0] 0.27*
Stenosis >50% (% prevalence) 25% 11% 6% 0.35
Markers of Inflammation and Immune Activation
sCD163 (ng/mL) 2841 [1722, 3427]a,b 1247 [829, 1883]a 847 [624, 1230] 0.0002*
sCD14 (ng/mL) 1530 [499, 1919]a 416 [218, 1614]a 241 [134, 395] 0.001*
hsIL6 (pg/mL) 1.74 [0.99, 6.33]a 1.05 [0.73, 1.65] 0.93 [0.55, 1.57] 0.13*
CXCL10 (pg/mL) 197 [173, 498]a 132 [100, 267] 109 [80, 175] 0.04*
hsCRP (mg/L) 0.4 [0.3, 2.4] 1.4 [0.6, 3.9] 1.2 [0.5, 3.1] 0.25*
MCP-1 (pg/mL) 319 [179, 452] 267 [200, 366] 232 [192, 286] 0.25*
%CD14+CD16+ monocytes 31.7 [12.8, 36.1] 18.3 [12.6, 29.9] 16.0 [9.3, 24.4] 0.24*
%CD38+HLA-DR+CD4 cells 0.9 [0.8, 1.7]a 1.2 [0.9, 1.9]a 0.6 [0.5, 0.7] < 0.0001*
%CD38+HLA-DR+CD8 cells 4.3 [1.8, 13.1] 1.9 [1.2, 4.2] 3.4 [2.1, 8.5] 0.10*
*

Nonparametric Wilcoxon/Kruskal-Wallis Test

a

P < 0.05 vs HIV-negative

b

P < 0.05 vs Chronic HIV-1 treated

For one HIV-1 elite controller, only calcium score was measured. Presence of coronary plaque could not be excluded. Measurements of coronary segments with plaque in table were obtained from 9 total HIV-1 elite controllers.

Mean ± SD for normally distributed parameters

Median [IQR] for non-normally distributed parameters

CXCL10 data were available in 56 participants (9 HIV-1 Elite controllers, 33 Chronic HIV-1, 14 HIV-negative)

Presence of plaque was significantly increased in EC compared to HIV- negative controls (78% vs. 42%, P< 0.05), and was relatively, but not statistically, increased compared to the prevalence of plaque seen in chronic HIV-1 (78% vs. 60%, P=0.28) (Table). The overall presence of plaque remained significantly increased in EC compared to HIV-negative controls after adjusting for known cardiovascular risk factors, including Framingham point score alone (P=0.03) or Framingham score and use of lipid lowering therapy (P=0.04). The proportion of EC with >50% stenosis of any coronary vessel (25%) tended to be greater than among chronic HIV-1 (11%) or HIV-negative controls (6%), but did not meet statistical significance due to the small number of EC. The percentage of patients with CAC tended to be higher in EC (Table).

sCD163, sCD14 and CXCL10 were significantly different between groups (P=0.0002, P=0.001 and P=0.04 respectively) and were higher in the EC than the HIV-negative controls (Table). Furthermore, sCD163 was significantly increased in EC compared to the chronicHIV-1. In contrast, hsCRP was not increased in EC. The %CD38+HLA-DR+ CD4+ was increased in both EC and chronic HIV-1 compared to HIV-negative controls (P<0.0001). The %CD38+HLA-DR+ CD8+ tended to be higher in EC, although not statistically significant.

Discussion

This study is the first to investigate coronary atherosclerosis using CTA in a carefully chosen group of art-naive EC without prior “viral blips”[11]. EC were compared to chronically HIV-1-infected, ART-treated, virologically suppressed patients, and HIV-negative controls, both of similar age.

We demonstrate an unexpectedly high degree of coronary atherosclerosis and elevated markers of immune activation in EC. Interestingly, the degree of atherosclerosis was similar, if not greater, compared to chronic HIV-1 receiving long-term ART with suppressed viremia, and was associated with a high degree of luminal stenosis, giving relevance to the data and emphasizing the added value of CTA beyond other measurements of CVD. This increase in plaque could not be explained by differences in traditional CVD risk factors or ART exposure, as the EC were ART naïve and CVD risk factors were similar between the groups.

Previous studies have shown that cIMT is higher in EC than in HIV-negative controls and comparable to that observed in chronic ART-treated HIV-1 patients[12]. Furthermore, increased T-cell activation has been associated to cIMT[2] and monocyte activation to noncalcified coronary plaque[1] among well-controlled ART-treated HIV patients. In this study, sCD163, a marker of monocyte/macrophage activation not previously measured in EC, was markedly increased compared to chronically HIV-1infected, ART-treated patients and HIV-negative controls, underscoring the potential contribution that monocytes/macrophages might play in the observed findings. Furthermore, sCD14 was also elevated, but surprisingly, CRP, a marker of generalized inflammation and CVD[13] was not increased in this group, despite the presence of significant coronary artery disease.

It is possible that sustained prolonged low-level viral replication in EC might directly contribute to endothelial damage[14] or lead to sustained T-cell and monocyte activation, which in turn contribute to increased arterial inflammation[15]. Although all EC in our study had undetectable viral loads over a prolonged period of time, there is residual low-level viremia and persistent cell integrated HIV-1DNA[3, 4, 10], replication competent virus can be isolated, and viral evolution has been demonstrated[16, 17]. Another possibility is that highly effective HIV-specific immune responses in EC[3, 18, 19], while critical to control viral replication, may result in chronic immune activation, accelerating atherosclerosis[20].

Our study included a small number of EC but this group represents < 1% of the total HIV population[21, 22], and we specifically selected a relatively younger group without any cardiovascular history, in whom there was no prior evidence of ”viral blips” over a long period of longitudinal follow-up. Our study is preliminary and underpowered to correlate immune activation with the degree of coronary atherosclerosis. Anticipated differences in the %CD38+HLA-DR+ CD8+ between the EC and chronic HIV-1 groups were observed but did not reach statistical significance due to sample sizes.

Taken together, our data suggests that HIV-1 elite controllers, despite excellent virologic and immunologic control and no confounding ART, have significant coronary atherosclerosis. The precise interplay between immune activation, highly effective HIV specific T-cell responses, low level viral replication and CVD need to be fully elucidated in larger studies of EC, in which highly sensitive assays of persistent low level viremia, below standard clinical detection limits, might also be related to atherosclerotic indices.

Acknowledgments

We wish to thank the participants of this study, the Nursing and Bionutrition Staff of the MGH and MIT GCRC and the members of the International HIV Controllers Study (www.hivcontrollers.org).

Funding was received from Bristol Myers Squibb, Inc., NIH R01 095123 (Dr. Grinspoon), NIH K24 DK064545 (Dr. Grinspoon), NIH K23 HL092792 (Dr. Lo), F32 HL088991 (Dr. Lo), NIH NS040237 (Dr. Williams) and M01 RR01066-25S1, the Bill and Melinda Gates Foundation (Dr. Pereyra). Funding sources had no role in the design of the study, data analysis or the writing of the manuscript.

Disclosures: Dr. Grinspoon received research funding for the coronary and immune function data on the non-elite controller women reported in this manuscript through an investigator-initiated research grant from Bristol Myers Squibb, Inc.

References

  • 1.Burdo TH, Lo J, Abbara S, Wei J, DeLelys ME, Preffer F, et al. Soluble CD163, a novel marker of activated macrophages, is elevated and associated with noncalcified coronary plaque in HIV-infected patients. J Infect Dis. 2011;204:1227–1236. doi: 10.1093/infdis/jir520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Kaplan RC, Sinclair E, Landay AL, Lurain N, Sharrett AR, Gange SJ, et al. T cell activation and senescence predict subclinical carotid artery disease in HIV-infected women. J Infect Dis. 2011;203:452–463. doi: 10.1093/infdis/jiq071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Pereyra F, Palmer S, Miura T, Block BL, Wiegand A, Rothchild AC, et al. Persistent low-level viremia in HIV-1 elite controllers and relationship to immunologic parameters. J Infect Dis. 2009;200:984–990. doi: 10.1086/605446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Hatano H, Delwart EL, Norris PJ, Lee TH, Dunn-Williams J, Hunt PW, et al. Evidence for persistent low-level viremia in individuals who control human immunodeficiency virus in the absence of antiretroviral therapy. J Virol. 2009;83:329–335. doi: 10.1128/JVI.01763-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Hunt PW, Brenchley J, Sinclair E, McCune JM, Roland M, Page-Shafer K, et al. Relationship between T cell activation and CD4+ T cell count in HIV-seropositive individuals with undetectable plasma HIV RNA levels in the absence of therapy. J Infect Dis. 2008;197:126–133. doi: 10.1086/524143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Pereyra F, Jia X, McLaren PJ, Telenti A, de Bakker PI, Walker BD, et al. The major genetic determinants of HIV-1 control affect HLA class I peptide presentation. Science. 2010;330:1551–1557. doi: 10.1126/science.1195271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Pereyra F, Addo MM, Kaufmann DE, Liu Y, Miura T, Rathod A, et al. Genetic and immunologic heterogeneity among persons who control HIV infection in the absence of therapy. J Infect Dis. 2008;197:563–571. doi: 10.1086/526786. [DOI] [PubMed] [Google Scholar]
  • 8.Lo J, Abbara S, Shturman L, Soni A, Wei J, Rocha-Filho JA, et al. Increased prevalence of subclinical coronary atherosclerosis detected by coronary computed tomography angiography in HIV-infected men. AIDS. 2010;24:243–253. doi: 10.1097/QAD.0b013e328333ea9e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Buzon MJ, Massanella M, Llibre JM, Esteve A, Dahl V, Puertas MC, et al. HIV-1 replication and immune dynamics are affected by raltegravir intensification of HAART-suppressed subjects. Nat Med. 2010;16:460–465. doi: 10.1038/nm.2111. [DOI] [PubMed] [Google Scholar]
  • 10.Buzon MJ, Seiss K, Weiss R, Brass AL, Rosenberg ES, Pereyra F, et al. Inhibition of HIV-1 integration in ex vivo-infected CD4 T cells from elite controllers. J Virol. 2011;85:9646–9650. doi: 10.1128/JVI.05327-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Boufassa F, Saez-Cirion A, Lechenadec J, Zucman D, Avettand-Fenoel V, Venet A, et al. CD4 dynamics over a 15 year-period among HIV controllers enrolled in the ANRS French observatory. PLoS One. 2011;6:e18726. doi: 10.1371/journal.pone.0018726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Hsue PY, Hunt PW, Schnell A, Kalapus SC, Hoh R, Ganz P, et al. Role of viral replication, antiretroviral therapy, and immunodeficiency in HIV-associated atherosclerosis. AIDS. 2009;23:1059–1067. doi: 10.1097/QAD.0b013e32832b514b. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Ridker PM, Rifai N, Rose L, Buring JE, Cook NR. Comparison of C-Reactive Protein and Low-Density Lipoprotein Cholesterol Levels in the Prediction of First Cardiovascular Events. N Engl J Med. 2002;347:1557–1565. doi: 10.1056/NEJMoa021993. [DOI] [PubMed] [Google Scholar]
  • 14.Torriani FJ, Komarow L, Parker RA, Cotter BR, Currier JS, Dube MP, et al. Endothelial function in human immunodeficiency virus-infected antiretroviral-naive subjects before and after starting potent antiretroviral therapy: The ACTG (AIDS Clinical Trials Group) Study 5152s. J Am Coll Cardiol. 2008;52:569–576. doi: 10.1016/j.jacc.2008.04.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Subramanian S, Tawakol A, Burdo TH, Abbara S, Wei J, Vijayakumar J, et al. Arterial Inflammation in HIV-Infected Patients. JAMA. 2012 doi: 10.1001/jama.2012.6698. In Press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.O'Connell KA, Brennan TP, Bailey JR, Ray SC, Siliciano RF, Blankson JN. Control of HIV-1 in elite suppressors despite ongoing replication and evolution in plasma virus. J Virol. 2010;84:7018–7028. doi: 10.1128/JVI.00548-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Blankson JN, Bailey JR, Thayil S, Yang HC, Lassen K, Lai J, et al. Isolation and characterization of replication-competent human immunodeficiency virus type 1 from a subset of elite suppressors. J Virol. 2007;81:2508–2518. doi: 10.1128/JVI.02165-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Hersperger AR, Pereyra F, Nason M, Demers K, Sheth P, Shin LY, et al. Perforin expression directly ex vivo by HIV-specific CD8 T-cells is a correlate of HIV elite control. PLoS Pathog. 2010;6:e1000917. doi: 10.1371/journal.ppat.1000917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Migueles SA, Osborne CM, Royce C, Compton AA, Joshi RP, Weeks KA, et al. Lytic granule loading of CD8+ T cells is required for HIV-infected cell elimination associated with immune control. Immunity. 2008;29:1009–1021. doi: 10.1016/j.immuni.2008.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Hsue PY, Deeks SG, Hunt PW. Immunologic basis of cardiovascular disease in HIV-infected adults. J Infect Dis. 2012;205(Suppl 3):S375–382. doi: 10.1093/infdis/jis200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Deeks SG, Walker BD. Human immunodeficiency virus controllers: mechanisms of durable virus control in the absence of antiretroviral therapy. Immunity. 2007;27:406–416. doi: 10.1016/j.immuni.2007.08.010. [DOI] [PubMed] [Google Scholar]
  • 22.Lambotte O, Boufassa F, Madec Y, Nguyen A, Goujard C, Meyer L, et al. HIV controllers: a homogeneous group of HIV-1-infected patients with spontaneous control of viral replication. Clin Infect Dis. 2005;41:1053–1056. doi: 10.1086/433188. [DOI] [PubMed] [Google Scholar]

RESOURCES