Updated Reviewed

Management of the Treatment-Experienced Patient

Poor CD4 Cell Recovery and Persistent Inflammation Despite Viral Suppression

Key Considerations and Recommendations
  • Persistently low CD4 T lymphocyte (CD4) cell counts and immune activation are each associated with increased AIDS- and non-AIDS-related morbidity and mortality among individuals with antiretroviral therapy (ART)-mediated viral suppression.
  • Adding antiretroviral (ARV) drugs to a suppressive ARV regimen (ART intensification) does not improve CD4 cell recovery or reduce immune activation and, therefore, is not recommended (AI).
  • In individuals with viral suppression, switching ARV drug classes does not consistently improve CD4 cell recovery or reduce all relevant markers of immune activation and is not recommended (BIII).
  • Interleukin-2 is not recommended (AI) to increase CD4 cell counts and/or decrease immune activation, because clinical trial data demonstrated no clinical benefit.
  • Other interventions designed to increase CD4 cell counts and/or decrease immune activation are not recommended outside of a clinical trial, because no current interventions have been proven to decrease morbidity or mortality during ART-mediated viral suppression (AII).
  • Efforts to decrease morbidity and mortality during ART-mediated viral suppression should focus on addressing modifiable risk factors for chronic disease (e.g., encouraging smoking cessation, a healthy diet, and regular exercise; treating hypertension and hyperlipidemia).
  • Monitoring markers of immune activation and inflammation is not recommended, because no intervention targeting immune pathways has proven to improve the health of individuals with HIV, and many blood markers that predict morbidity and mortality fluctuate within individuals (AII).
Rating of Recommendations:  A = Strong; B = Moderate; C = Weak
Rating of Evidence:  I = Data from randomized controlled trials; II = Data from well-designed nonrandomized trials or observational cohort studies with long-term clinical outcomes; III = Expert opinion

Despite marked improvements in antiretroviral treatment (ART), morbidity and mortality in individuals with HIV continue to be greater than in the general population, particularly when ART is delayed until advanced disease stages. These morbidities include cardiovascular disease, many non-AIDS cancers, non-AIDS infections, chronic obstructive pulmonary disease, osteoporosis, type 2 diabetes, thromboembolic disease, liver disease, renal disease, neurocognitive dysfunction, and the frailty phenotype.1 Although health-related behaviors and toxicities of antiretroviral (ARV) drugs are important factors, immune characteristics, such as a poor CD4 T lymphocyte (CD4) cell recovery and persistent immune activation and inflammation, likely also contribute to the disease risk among persons with HIV.

Poor CD4 Cell Recovery

As long as ART-mediated viral suppression is maintained, peripheral blood CD4 cell counts in most individuals with HIV will continue to increase for at least a decade. The rate of CD4 cell recovery is typically most rapid in the first 3 months of suppressive ART, followed by more gradual increases over time.2-4  If ART-mediated viral suppression is maintained, most individuals will eventually recover CD4 counts in the normal range (>500 cells/mm3); however, approximately 15% to 20% of individuals who initiate ART at very low CD4 counts (<200 cells/mm3) may plateau at abnormally low CD4 cell counts.3-5  Early initiation of ART in individuals with recent HIV diagnoses likely provides the best opportunity for maximal CD4 cell recovery.6  

Persistently low CD4 cell counts despite ART-mediated viral suppression are associated with increased risk of morbidity and mortality. For example, individuals with HIV who have CD4  counts <200 cells/mm3, despite at least 3 years of suppressive ART, had a 2.6-fold greater risk of mortality than those with higher CD4 cell counts.7  Lower CD4 cell counts during ART-mediated viral suppression are associated with an increased risk of non-AIDS morbidity and mortality,8-11  including cardiovascular disease,12  osteoporosis and fractures,13  liver disease,14  and infection-related cancers.15  The prognostic importance of higher CD4 cell counts likely spans all ranges of CD4 cell counts, though incremental benefits are harder to discern once CD4 counts increase16  to >500 cells/mm3. 

Individuals with poor CD4 cell recovery should be evaluated for modifiable causes of CD4 cell lymphopenia. Concomitant medications should be reviewed, with a focus on those known to decrease white blood cells or, specifically, CD4 cells. If possible, these drugs should be substituted for or discontinued. Untreated coinfections (e.g., hepatitis C virus, HIV-2) and serious medical conditions (e.g., malignancy) also should be considered as possible causes of CD4 lymphopenia, particularly in individuals with consistently declining CD4 cell counts (and percentages) and in those with CD4 counts consistently below 100 cells/mm3

In rare cases, CD4 cell counts actually decline, despite suppressive ART in the absence of an obvious clinical cause. Severe derangements in interleukin (IL)-7-mediated naive T cell homeostasis have been reported in these individuals, although the pathophysiology is likely to be multifactorial.17-19 

Despite strong evidence linking low CD4 cell counts and increased morbidity during ART-mediated viral suppression, no adjunctive therapies that increase CD4 cell count beyond levels achievable with ART alone have been proven to decrease morbidity or mortality. Adding ARV drugs to an already suppressive ARV regimen does not improve CD4 cell recovery,20-25  and does not reduce morbidity or mortality. Therefore, ART intensification is not recommended as a strategy to improve CD4 cell recovery (AI). Similarly, for individuals who are already maintaining viral suppression, switching ARV drug classes also does not consistently improve CD4 cell recovery and is not recommended (BIII).26  

Immune-based therapies also have been investigated as a strategy to increase CD4 cell counts (e.g., IL-2, IL-7, growth hormone). Two large clinical outcome trials, powered to assess impact on clinical endpoints (AIDS and death), evaluated the role of interleukin-2 for improving CD4 cell recovery. IL-2 adjunctive therapy resulted in substantial CD4 cell count increases but with no observable clinical benefit.27 Therefore, IL-2 is not recommended (AI). Given the lack of established clinical benefit to date, other immune-based therapies should not be used except in the context of a clinical trial.

Persistent Immune Activation and Inflammation

HIV infection results in heightened systemic immune activation and inflammation, which predict more rapid CD4 cell decline and progression to AIDS and death, independent of plasma HIV RNA levels.28 Although immune activation declines with suppressive ART, it often persists at abnormal levels in many individuals with HIV maintaining long-term ART-mediated viral suppression—even in those with CD4 cell recovery to normal levels.29, 30  Immune activation and inflammatory markers (e.g., IL-6, D-dimer, high sensitivity C-reactive protein (hs-CRP) also predict mortality and non-AIDS morbidity during ART-mediated viral suppression, including cardiovascular and thromboembolic events, cancer, neurocognitive dysfunction, and frailty.28  A low CD4/CD8 ratio also might reflect this inflammatory state to some degree,31 although it predicts AIDS events far more strongly than non-AIDS morbidity.32  Although individuals with poor CD4 cell recovery (i.e., counts persistently <350 cells/mm3) tend to have greater immune activation and inflammation than those with greater recovery,29  the relationship between innate immune activation and inflammation and morbidity/mortality is largely independent of CD4 cell count.33, 34  Even in individuals with CD4 counts >500 cells/mm3, immune activation and inflammation are associated with increased morbidity and mortality.35, 36  

ART as a Strategy to Reduce Inflammation

Early diagnosis and treatment of HIV is, potentially, an effective strategy to achieve a lower level of persistent immune activation with ART. Most inflammatory markers decline during the first several months of ART and achieve a stable “setpoint” within 1 to 2 years.37, 38 In observational studies, people with HIV who initiated ART during acute HIV infection appeared to achieve a lower immune activation setpoint during ART-mediated viral suppression than those who started ART at later disease stages.39, 40  Indeed, those randomized to the immediate treatment arm of the START trial appeared to achieve a lower early inflammatory setpoint than those who were randomized to delayed therapy.41  Longer-term follow-up of START participants is needed to determine whether such a reduced inflammatory setpoint persists and translates into reduced morbidity and mortality. Collectively, these data reinforce the recommendation to start ART as soon as possible after HIV diagnosis (see Initiation of Antiretroviral Therapy). 

Although earlier initiation of ART appears to consistently reduce the inflammatory setpoint during ART, intensifying or modifying ART after viral suppression is already achieved does not appear to consistently reduce immune activation. For example, adding ARV drugs to an already suppressive ARV regimen (or ART intensification) does not consistently improve immune activation.20-23, 25  Although some studies have suggested that switching an ARV regimen to one with a more favorable lipid profile may improve some markers of immune activation and inflammation,42-44  these studies have limitations and results are not consistent across markers and among studies. Thus, at this time, ART modification cannot be recommended as a strategy to reduce immune activation (BIII)

Other Immune-Based Strategies 

Because persistent immune activation is associated with morbidity and mortality among people with HIV who are virologically suppressed with ART, strategies targeting immune-mediators of inflammation are under investigation. Although the efficacy of canakimumab is not yet proven in people with HIV, the CANTOS trial provided important proof of concept for the causal role of inflammation in the risk of multi-morbidity in people without HIV but with cardiovascular disease. The study demonstrated that treatment with canakimumab, a human monoclonal antibody targeting cytokine IL-1b, a driver of the IL-6 signaling pathway, reduced cardiovascular events and cancer death.45  In people with HIV, canakinumab and the IL-6 inhibitor tocilizumab have been shown to reduce blood levels of markers of inflammation and immune activation.46, 47  Nevertheless, it remains unclear whether the potential risks of these therapies, such as the increased risk of death from sepsis observed in the CANTOS trial, might outweigh any benefits in people with HIV. Therefore, interventions targeting immune mediators of inflammation are not currently recommended for clinical use for the treatment of immune activation in people with HIV (AII).  

Treatments Targeting Traditional Risk Factors and Inflammation

Beyond the well-established clinical benefit for reducing cardiovascular events, hydroxy-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors (or statins) have been shown to improve biomarker levels of inflammation (e.g., hsCRP) and immune activation in the general population.48-50  This premise, and the data suggesting similar benefit among people with HIV, motivated the design of a large clinical trial (REPRIEVE), now fully enrolled, to determine whether pitavastatin reduces cardiovascular events in people with HIV who do not already have a clinical indication for cholesterol-lowering therapy. 51  Although the results will not be known for several years, in addition to identifying the primary cardiovascular outcomes, assessing the impact of pivitastatin on non-cardiovascular events (such as cancer, osteoporotic fractures, and frailty phenotypes) that may be linked to the inflammatory state also will be valuable. Similarly, it remains unclear whether statins might further increase type 2 diabetes risk, which is increased in people with HIV.52  Other commonly used medications with anti-inflammatory properties—like aspirin, angiotensin-converting enzyme inhibitors, methotrexate, and angiotensin receptor blockers53 —have failed to consistently reduce biomarkers of immune activation and/or inflammation in people with HIV in randomized controlled trials and,54-57  as a result, clinical outcome trials specific to this population are not anticipated. 

Treatments Targeting Putative Drivers of the Inflammatory State

Other investigational approaches to reduce the inflammatory state in patients with viral suppression on ART have focused on the presumed root drivers of inflammation, including HIV reactivation from latently infected cells, microbial translocation, and chronic co-infections, particularly cytomegalovirus (CMV).28  Thus far, the only approach targeting these root drivers that has broadly reduced systemic immune activation is treating asymptomatic CMV co-infection,58  an approach that is being pursued further in a prospective larger trial (ACTG A5383). Presently, none of these strategies have been proven to be effective in clinical endpoint trials, so these interventions should be pursued only in the context of clinical trials.

Monitoring Inflammation

In the absence of proven interventions, there is no clear rationale to routinely monitor levels of immune activation and inflammation in treated HIV infection. Furthermore, many of the inflammatory markers that predict morbidity and mortality fluctuate significantly in individuals with HIV. Thus, clinical monitoring with immune activation or inflammatory markers is not currently recommended (AII). The focus of care to reduce chronic non-AIDS morbidity and mortality should be on maintaining ART-mediated viral suppression and addressing strategies to reduce risk factors (e.g., smoking cessation, healthy diet, and exercise) and managing chronic comorbidities, such as hypertension, hyperlipidemia, and diabetes (AII)

References

  1. Deeks SG. HIV Infection, Inflammation, Immunosenescence, and Aging. Annu Rev Med. 2011;62:141-155. Available at: http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=21090961.
  2. Bartlett JA, DeMasi R, Quinn J, Moxham C, Rousseau F. Overview of the effectiveness of triple combination therapy in antiretroviral-naive HIV-1 infected adults. Aids. 2001;15(11):1369-1377. Available at: http://www.ncbi.nlm.nih.gov/pubmed/11504958.
  3. Kelley CF, Kitchen CM, Hunt PW, et al. Incomplete peripheral CD4+ cell count restoration in HIV-infected patients receiving long-term antiretroviral treatment. Clin Infect Dis. 2009;48(6):787-794. Available at: http://www.ncbi.nlm.nih.gov/pubmed/19193107.
  4. Lok JJ, Bosch RJ, Benson CA, et al. Long-term increase in CD4+ T-cell counts during combination antiretroviral therapy for HIV-1 infection. Aids. 2010;24(12):1867-1876. Available at: http://www.ncbi.nlm.nih.gov/pubmed/20467286.
  5. Moore RD, Keruly JC. CD4+ cell count 6 years after commencement of highly active antiretroviral therapy in persons with sustained virologic suppression. Clin Infect Dis. 2007;44(3):441-446. Available at: http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=17205456.
  6. Le T, Wright EJ, Smith DM, et al. Enhanced CD4+ T-cell recovery with earlier HIV-1 antiretroviral therapy. N Engl J Med. 2013;368(3):218-230. Available at: http://www.ncbi.nlm.nih.gov/pubmed/23323898.
  7. Engsig FN, Zangerle R, Katsarou O, et al. Long-term mortality in HIV-positive individuals virally suppressed for >3 years with incomplete CD4 recovery. Clin Infect Dis. 2014;58(9):1312-1321. Available at: http://www.ncbi.nlm.nih.gov/pubmed/24457342.
  8. Lewden C, Bouteloup V, De Wit S, et al. All-cause mortality in treated HIV-infected adults with CD4 >=500/mm3 compared with the general population: evidence from a large European observational cohort collaboration. Int J Epidemiol. 2012;41(2):433-445. Available at: http://www.ncbi.nlm.nih.gov/pubmed/22493325.
  9. Baker JV, Peng G, Rapkin J, et al. CD4+ count and risk of non-AIDS diseases following initial treatment for HIV infection. Aids. 2008;22(7):841-848. Available at: http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=18427202
  10. Achhra AC, Amin J, Law MG, et al. Immunodeficiency and the risk of serious clinical endpoints in a well studied cohort of treated HIV-infected patients. AIDS. 2010;24(12):1877-1886. Available at: http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=20588170.
  11. Smurzynski M, Wu K, Benson CA, Bosch RJ, Collier AC, Koletar SL. Relationship between CD4+ T-cell counts/HIV-1 RNA plasma viral load and AIDS-defining events among persons followed in the ACTG longitudinal linked randomized trials study. J Acquir Immune Defic Syndr. 2010;55(1):117-127. Available at: http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=20622677.
  12. Lichtenstein KA, Armon C, Buchacz K, et al. Low CD4+ T cell count is a risk factor for cardiovascular disease events in the HIV outpatient study. Clin Infect Dis. 2010;51(4):435-447. Available at: http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=20597691.
  13. Yong MK, Elliott JH, Woolley IJ, Hoy JF. Low CD4 count is associated with an increased risk of fragility fracture in HIV-infected patients. J Acquir Immune Defic Syndr. 2011;57(3):205-210. Available at: http://www.ncbi.nlm.nih.gov/pubmed/21522014.
  14. Weber R, Sabin CA, Friis-Moller N, et al. Liver-related deaths in persons infected with the human immunodeficiency virus: the D:A:D study. Arch Intern Med. 2006;166(15):1632-1641. Available at: http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=16908797.
  15. Monforte A, Abrams D, Pradier C, et al. HIV-induced immunodeficiency and mortality from AIDS-defining and non-AIDS-defining malignancies. Aids. 2008;22(16):2143-2153. Available at: http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=18832878
  16. Young J, Psichogiou M, Meyer L, et al. CD4 cell count and the risk of AIDS or death in HIV-Infected adults on combination antiretroviral therapy with a suppressed viral load: a longitudinal cohort study from COHERE. PLoS Med. 2012;9(3):e1001194. Available at: http://www.ncbi.nlm.nih.gov/pubmed/22448150.
  17. Lisco A, Wong CS, Lage SL, et al. Identification of rare HIV-1-infected patients with extreme CD4+ T cell decline despite ART-mediated viral suppression. JCI Insight. 2019;4(8). Available at: https://www.ncbi.nlm.nih.gov/pubmed/30996137.
  18. Schacker TW, Bosch RJ, Bennett K, et al. Measurement of naive CD4 cells reliably predicts potential for immune reconstitution in HIV. J Acquir Immune Defic Syndr. 2010;54(1):59-62. Available at: https://www.ncbi.nlm.nih.gov/pubmed/20182359.
  19. Zeng M, Southern PJ, Reilly CS, et al. Lymphoid tissue damage in HIV-1 infection depletes naive T cells and limits T cell reconstitution after antiretroviral therapy. PLoS Pathog. 2012;8(1):e1002437. Available at: https://www.ncbi.nlm.nih.gov/pubmed/22241988.
  20. Gandhi RT, Zheng L, Bosch RJ, et al. The effect of raltegravir intensification on low-level residual viremia in HIV-infected patients on antiretroviral therapy: a randomized controlled trial. PLoS Med. 2010;7(8). Available at: http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=20711481.
  21. Hatano H, Strain MC, Scherzer R, et al. Increase in 2-Long Terminal Repeat Circles and Decrease in D-dimer After Raltegravir Intensification in Patients With Treated HIV Infection: A Randomized, Placebo-Controlled Trial. J Infect Dis. 2013;208(9):1436-1442. Available at: http://www.ncbi.nlm.nih.gov/pubmed/23975885.
  22. Hunt PW, Shulman NS, Hayes TL, et al. The immunologic effects of maraviroc intensification in treated HIV-infected individuals with incomplete CD4+ T-cell recovery: a randomized trial. Blood. 2013;121(23):4635-4646. Available at: http://www.ncbi.nlm.nih.gov/pubmed/23589670.
  23. Dinoso JB, Kim SY, Wiegand AM, et al. Treatment intensification does not reduce residual HIV-1 viremia in patients on highly active antiretroviral therapy. Proc Natl Acad Sci USA. 2009;106(23):9403-9408. Available at: https://www.ncbi.nlm.nih.gov/pubmed/19470482.
  24. Cuzin L, Trabelsi S, Delobel P, et al. Maraviroc intensification of stable antiviral therapy in HIV-1-infected patients with poor immune restoration: MARIMUNO-ANRS 145 study. J Acquir Immune Defic Syndr. 2012;61(5):557-564. Available at: http://www.ncbi.nlm.nih.gov/pubmed/22986949.
  25. Buzon MJ, Massanella M, Llibre JM, et al. HIV-1 replication and immune dynamics are affected by raltegravir intensification of HAART-suppressed subjects. Nat Med. 2010;16(4):460-465. Available at: http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=20228817.
  26. Martinez E, Larrousse M, Llibre JM, et al. Substitution of raltegravir for ritonavir-boosted protease inhibitors in HIV-infected patients: the SPIRAL study. AIDS. 2010;24(11):1697-1707. Available at: http://www.ncbi.nlm.nih.gov/pubmed/20467288.
  27. Abrams D, Levy Y, Losso MH, et al. Interleukin-2 therapy in patients with HIV infection. N Engl J Med. 2009;361(16):1548-1559. Available at: http://www.ncbi.nlm.nih.gov/pubmed/19828532.
  28. Lederman MM, Funderburg NT, Sekaly RP, Klatt NR, Hunt PW. Residual immune dysregulation syndrome in treated HIV infection. Adv Immunol. 2013;119:51-83. Available at: http://www.ncbi.nlm.nih.gov/pubmed/23886064.
  29. Lederman MM, Calabrese L, Funderburg NT, et al. Immunologic failure despite suppressive antiretroviral therapy is related to activation and turnover of memory CD4 cells. J Infect Dis. 2011;204(8):1217-1226. Available at: http://www.ncbi.nlm.nih.gov/pubmed/21917895.
  30. Hunt PW, Martin JN, Sinclair E, et al. T cell activation is associated with lower CD4+ T cell gains in human immunodeficiency virus-infected patients with sustained viral suppression during antiretroviral therapy. J Infect Dis. 2003;187(10):1534-1543. Available at: http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?cmd=Retrieve&db=PubMed&dopt=Citation&list_uids=12721933
  31. Serrano-Villar S, Sainz T, Lee SA, et al. HIV-infected individuals with low CD4/CD8 ratio despite effective antiretroviral therapy exhibit altered T cell subsets, heightened CD8+ T cell activation, and increased risk of non-AIDS morbidity and mortality. PLoS Pathog. 2014;10(5):e1004078. Available at: https://www.ncbi.nlm.nih.gov/pubmed/24831517.
  32. Trickey A, May MT, Schommers P, et al. CD4:CD8 Ratio and CD8 Count as Prognostic Markers for Mortality in Human Immunodeficiency Virus-Infected Patients on Antiretroviral Therapy: The Antiretroviral Therapy Cohort Collaboration (ART-CC). Clin Infect Dis. 2017;65(6):959-966. Available at: https://pubmed.ncbi.nlm.nih.gov/28903507/.
  33. Hunt PW, Sinclair E, Rodriguez B, et al. Gut Epithelial Barrier Dysfunction and Innate Immune Activation Predict Mortality in Treated HIV Infection. J Infect Dis. 2014. Available at: http://www.ncbi.nlm.nih.gov/pubmed/24755434.
  34. Tenorio AR, Zheng Y, Bosch RJ, et al. Soluble Markers of Inflammation and Coagulation but Not T-Cell Activation Predict Non-AIDS-Defining Morbid Events During Suppressive Antiretroviral Treatment. J Infect Dis. 2014. Available at: http://www.ncbi.nlm.nih.gov/pubmed/24795473.
  35. Tien PC, Choi AI, Zolopa AR, et al. Inflammation and mortality in HIV-infected adults: analysis of the FRAM study cohort. J Acquir Immune Defic Syndr. 2010;55(3):316-322. Available at: https://www.ncbi.nlm.nih.gov/pubmed/20581689.
  36. Baker JV, Sharma S, Grund B, et al. Systemic Inflammation, Coagulation, and Clinical Risk in the START Trial. Open Forum Infect Dis. 2017;4(4):ofx262. Available at: https://www.ncbi.nlm.nih.gov/pubmed/29308409.
  37. Gandhi RT, McMahon DK, Bosch RJ, et al. Levels of HIV-1 persistence on antiretroviral therapy are not associated with markers of inflammation or activation. PLoS Pathog. 2017;13(4):e1006285. Available at: https://pubmed.ncbi.nlm.nih.gov/28426825/.
  38. Wada NI, Jacobson LP, Margolick JB, et al. The effect of HAART-induced HIV suppression on circulating markers of inflammation and immune activation. Aids. 2015;29(4):463-471. Available at: https://pubmed.ncbi.nlm.nih.gov/25630041/.
  39. Hellmuth J, Slike BM, Sacdalan C, et al. Very Early Initiation of Antiretroviral Therapy During Acute HIV Infection Is Associated With Normalized Levels of Immune Activation Markers in Cerebrospinal Fluid but Not in Plasma. J Infect Dis. 2019;220(12):1885-1891. Available at: https://www.ncbi.nlm.nih.gov/pubmed/30668739.
  40. Sereti I, Krebs SJ, Phanuphak N, et al. Persistent, Albeit Reduced, Chronic Inflammation in Persons Starting Antiretroviral Therapy in Acute HIV Infection. Clin Infect Dis. 2017;64(2):124-131. Available at: https://www.ncbi.nlm.nih.gov/pubmed/27737952.
  41. Baker JV, Grund B, Sharma S, et al. Cardiometabolic and pulmonary complications. Presented at: LONG-TERM ELEVATED IL-6 AND D-DIMER AFTER DELAYED ART INITIATION IN THE START TRIAL; 2020. Available at: https://www.croiconference.org/abstract/long-term-elevated-il-6-and-d-dimer-after-delayed-art-initiation-in-the-start-trial/.
  42. Martinez E, D'Albuquerque PM, Llibre JM, et al. Changes in cardiovascular biomarkers in HIV-infected patients switching from ritonavir-boosted protease inhibitors to raltegravir. Aids. 2012;26(18):2315-2326. Available at: http://www.ncbi.nlm.nih.gov/pubmed/23018438.
  43. Lake JE, McComsey GA, Hulgan T, et al. Switch to raltegravir decreases soluble CD14 in virologically suppressed overweight women: the Women, Integrase and Fat Accumulation Trial. HIV Med. 2014;15(7):431-441. Available at: http://www.ncbi.nlm.nih.gov/pubmed/24506429.
  44. Asundi A, Robles Y, Starr T, et al. Immunological and Neurometabolite Changes Associated With Switch From Efavirenz to an Integrase Inhibitor. J Acquir Immune Defic Syndr. 2019;81(5):585-593. Available at: https://www.ncbi.nlm.nih.gov/pubmed/31045650.
  45. Ridker PM, Everett BM, Thuren T, et al. Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease. N Engl J Med. 2017;377(12):1119-1131. Available at: https://www.ncbi.nlm.nih.gov/pubmed/28845751.
  46. Hsue PY, Li D, Ma Y, et al. IL-1beta Inhibition Reduces Atherosclerotic Inflammation in HIV Infection. J Am Coll Cardiol. 2018;72(22):2809-2811. Available at: https://www.ncbi.nlm.nih.gov/pubmed/30497570.
  47. Rodriguez B, Chen Z, Tatsuoka C, et al. IL-6 BLOCKADE DECREASES INFLAMMATION AND INCREASES CD127 EXPRESSION IN HIV INFECTION. Presented at: Conference on Retroviruses and Opportunistic Infections; 2020. Boston, Massachusetts. Available at: https://www.croiconference.org/abstract/il-6-blockade-decreases-inflammation-and-increases-cd127-expression-in-hiv-infection/.
  48. Ridker PM, Danielson E, Fonseca FA, et al. Reduction in C-reactive protein and LDL cholesterol and cardiovascular event rates after initiation of rosuvastatin: a prospective study of the JUPITER trial. Lancet. 2009;373(9670):1175-1182. Available at: https://www.ncbi.nlm.nih.gov/pubmed/19329177.
  49. Ota H, Eto M, Kano MR, et al. Induction of endothelial nitric oxide synthase, SIRT1, and catalase by statins inhibits endothelial senescence through the Akt pathway. Arterioscler Thromb Vasc Biol. 2010;30(11):2205-2211. Available at: https://www.ncbi.nlm.nih.gov/pubmed/20705918.
  50. Jougasaki M, Ichiki T, Takenoshita Y, Setoguchi M. Statins suppress interleukin-6-induced monocyte chemo-attractant protein-1 by inhibiting Janus kinase/signal transducers and activators of transcription pathways in human vascular endothelial cells. Br J Pharmacol. 2010;159(6):1294-1303. Available at: https://www.ncbi.nlm.nih.gov/pubmed/20136831.
  51. Fichtenbaum CJ, Ribaudo HJ, Leon-Cruz J, et al. Patterns of Antiretroviral Therapy Use and Immunologic Profiles at Enrollment in the REPRIEVE Trial. The Journal of Infectious Diseases. 2020;222:S8-S19. Available at: https://academic.oup.com/jid/article/222/Supplement_1/S8/5869455.
  52. Hernandez-Romieu AC, Garg S, Rosenberg ES, Thompson-Paul AM, Skarbinski J. Is diabetes prevalence higher among HIV-infected individuals compared with the general population? Evidence from MMP and NHANES 2009-2010. BMJ Open Diabetes Res Care. 2017;5(1):e000304. Available at: https://www.ncbi.nlm.nih.gov/pubmed/28191320.
  53. Hsue PY, Ribaudo HJ, Deeks SG, et al. Safety and Impact of Low-dose Methotrexate on Endothelial Function and Inflammation in Individuals With Treated Human Immunodeficiency Virus: AIDS Clinical Trials Group Study A5314. Clin Infect Dis. 2019; 68(11):1877-1886. Available at: https://pubmed.ncbi.nlm.nih.gov/30219823/.
  54. O'Brien MP, Hunt PW, Kitch DW, et al. A Randomized Placebo Controlled Trial of Aspirin Effects on Immune Activation in Chronically Human Immunodeficiency Virus-Infected Adults on Virologically Suppressive Antiretroviral Therapy. Open Forum Infect Dis. 2017;4(1):ofw278. Available at: https://www.ncbi.nlm.nih.gov/pubmed/28480270.
  55. Cockerham LR, Yukl SA, Harvill K, et al. A Randomized Controlled Trial of Lisinopril to Decrease Lymphoid Fibrosis in Antiretroviral-Treated, HIV-infected Individuals. Pathog Immun. 2017; 2(3):310-334. Available at: https://www.ncbi.nlm.nih.gov/pubmed/28936485.
  56. Utay NS, Kitch DW, Yeh E, et al. Telmisartan Therapy Does Not Improve Lymph Node or Adipose Tissue Fibrosis More Than Continued Antiretroviral Therapy Alone. J Infect Dis. 2018; 217(11):1770-1781. Available at: https://pubmed.ncbi.nlm.nih.gov/29401318/.
  57. Baker JV, Wolfson J, Collins G, et al. Losartan to reduce inflammation and fibrosis endpoints in HIV disease. AIDS. 2021;35(4):575-583. Available at: https://pubmed.ncbi.nlm.nih.gov/33252490/.
  58. Hunt PW, Martin JN, Sinclair E, et al. Valganciclovir reduces T cell activation in HIV-infected individuals with incomplete CD4+ T cell recovery on antiretroviral therapy. J Infect Dis. 2011; 203(10):1474-1483. Available at: https://pubmed.ncbi.nlm.nih.gov/21502083/.

 

Management of the Treatment-Experienced Patient

Poor CD4 Cell Recovery and Persistent Inflammation Despite Viral Suppression

Key Considerations and Recommendations
  • Persistently low CD4 T lymphocyte (CD4) cell counts and immune activation are each associated with increased AIDS- and non-AIDS-related morbidity and mortality among individuals with antiretroviral therapy (ART)-mediated viral suppression.
  • Adding antiretroviral (ARV) drugs to a suppressive ARV regimen (ART intensification) does not improve CD4 cell recovery or reduce immune activation and, therefore, is not recommended (AI).
  • In individuals with viral suppression, switching ARV drug classes does not consistently improve CD4 cell recovery or reduce all relevant markers of immune activation and is not recommended (BIII).
  • Interleukin-2 is not recommended (AI) to increase CD4 cell counts and/or decrease immune activation, because clinical trial data demonstrated no clinical benefit.
  • Other interventions designed to increase CD4 cell counts and/or decrease immune activation are not recommended outside of a clinical trial, because no current interventions have been proven to decrease morbidity or mortality during ART-mediated viral suppression (AII).
  • Efforts to decrease morbidity and mortality during ART-mediated viral suppression should focus on addressing modifiable risk factors for chronic disease (e.g., encouraging smoking cessation, a healthy diet, and regular exercise; treating hypertension and hyperlipidemia).
  • Monitoring markers of immune activation and inflammation is not recommended, because no intervention targeting immune pathways has proven to improve the health of individuals with HIV, and many blood markers that predict morbidity and mortality fluctuate within individuals (AII).
Rating of Recommendations:  A = Strong; B = Moderate; C = Weak
Rating of Evidence:  I = Data from randomized controlled trials; II = Data from well-designed nonrandomized trials or observational cohort studies with long-term clinical outcomes; III = Expert opinion

Download Guidelines