Skip to main content

Advertisement

Log in

HIV-Associated NAFLD: Disease Burden and Management

  • Fatty Liver Disease (V Ajmera, Section Editor)
  • Published:
Current Hepatology Reports Aims and scope Submit manuscript

Abstract

Purpose of Review

Highly potent anti-retroviral therapy (ART) for the treatment of human immunodeficiency virus (HIV) has led to dramatic improvements in quality of life and lifespan in persons living with HIV (PLWH). PLWH, however, are suffering from other comorbid conditions, including non-alcoholic fatty liver disease (NAFLD). This review summarizes the epidemiology and pathophysiology of NAFLD in PLWH and explores unique diagnostic and treatment considerations in this population.

Recent Findings

Though it is well established that there is a high prevalence of NAFLD in PLWH, the mechanisms underlying NAFLD in this population are just beginning to be explored. Traditional NAFLD risk factors, including insulin resistance, visceral adiposity, and genetics, have been consistently linked with NAFLD in PLWH. In addition, HIV-related factors including mitochondrial dysfunction, microbiome alterations, and direct effects of the virus and of ART may play a role.

Summary

Given the burden of NAFLD in PLWH, further studies are necessary to investigate mechanisms specific to HIV with which to target therapies.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

Abbreviations

HIV:

Human immunodeficiency virus

NAFLD:

Non-alcoholic fatty liver disease

NASH:

Non-alcoholic steatohepatitis

ART:

Anti-retroviral therapy

PLWH:

Persons living with HIV

References

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. UNAIDS. Fact Sheet - World AIDS Day 2018.; 2018.

  2. Smith CJ, Ryom L, Weber R, et al. Trends in underlying causes of death in people with HIV from 1999 to 2011 (D:A:D): A multicohort collaboration. Lancet. 2014;384(9939):241–248. https://doi.org/10.1016/S0140-6736(14)60604-8.

  3. Serrano-Villar S, Gutiérrez F, Miralles C, et al. Human immunodeficiency virus as a chronic disease: Evaluation and management of nonacquired immune deficiency syndrome-defining conditions. Open Forum Infect Dis. 2016;3(2). https://doi.org/10.1093/ofid/ofw097.

  4. Price JC, Thio CL. Liver disease in the HIV-infected individual. Clin Gastroenterol Hepatol. 2010;8(12):1002–1012. https://doi.org/10.1016/j.cgh.2010.08.024.

  5. Kleiner DE, Makhlouf HR. Histology of Nonalcoholic Fatty Liver Disease and Nonalcoholic Steatohepatitis in Adults and Children. Clin Liver Dis. 2016;20(2):293–312. https://doi.org/10.1016/j.cld.2015.10.011.

  6. Lackner C. Hepatocellular ballooning in nonalcoholic steatohepatitis: The pathologist’s perspective. Expert Rev Gastroenterol Hepatol. 2011;5(2):223–231. https://doi.org/10.1586/egh.11.8.

  7. Wong RJ, Aguilar M, Cheung R, et al. Nonalcoholic steatohepatitis is the second leading etiology of liver disease among adults awaiting liver transplantation in the United States. Gastroenterology. 2015;148(3):547–555. https://doi.org/10.1053/j.gastro.2014.11.039.

  8. Cholankeril G, Wong RJ, Hu M, et al. Liver Transplantation for Nonalcoholic Steatohepatitis in the US: Temporal Trends and Outcomes. Dig Dis Sci. 2017;62(10):2915–2922. https://doi.org/10.1007/s10620-017-4684-x.

  9. Younossi Z, Koenig A, Abdelatif D, Fazel Y, Henry L, Wymer M. Global Epidemiology of Non-Alcoholic Fatty Liver Disease-Meta-Analytic Assessment of Prevalence, Incidence and Outcomes. Hepatology. 2015;64(1):73–84. https://doi.org/10.1002/hep.28431.

  10. Hadigan C, Liebau J, Andersen R, Holalkere NS, Sahani D V. Magnetic resonance spectroscopy of hepatic lipid content and associated risk factors in HIV infection. J Acquir Immune Defic Syndr. 2007;46(3):312–317. https://doi.org/10.1097/QAI.0b013e3181568cc2.

  11. Moreno-Torres A, Domingo P, Pujol J, Blanco-Vaca F, Arroyo JA, Sambeat MA. Liver triglyceride content in HIV-1-infected patients on combination antiretroviral therapy studied with 1H-MR spectroscopy. Antivir Ther. 2007;12(2):195–203.

  12. Guaraldi G, Squillace N, Stentarelli C, et al. Nonalcoholic Fatty Liver Disease in HIV-Infected Patients Referred to a Metabolic Clinic: Prevalence, Characteristics, and Predictors. Clin Infect Dis. 2008;47(2):250–257. https://doi.org/10.1086/589294.

  13. Crum-Cianflone N, Dilay A, Collins G, et al. Nonalcoholic fatty liver disease among HIVinfected persons. J Acquir Immune Defic Syndr. 2009;50(5):464–473. https://doi.org/10.1097/QAI.0b013e318198a88a.

  14. Nishijima T, Gatanaga H, Shimbo T, et al. Traditional but not HIV-related factors are associated with nonalcoholic fatty liver disease in asian patients with HIV-1 infection. PLoS One. 2014;9(1). https://doi.org/10.1371/journal.pone.0087596.

  15. Price JC, Seaberg EC, Latanich R, et al. Risk Factors for Fatty Liver in the Multicenter AIDS Cohort Study. Am J Gastroenterol. 2014;109(5):695–704. https://doi.org/10.1038/ajg.2014.32.

  16. Macías J, González J, Tural C, et al. Prevalence and factors associated with liver steatosis as measured by transient elastography with controlled attenuation parameter in HIVinfected patients. AIDS. 2014;28(9):1279–1287. https://doi.org/10.1097/QAD.0000000000000248.

  17. Lui G, Wong VWS, Wong GLH, et al. Liver fibrosis and fatty liver in Asian HIV-infected patients. Aliment Pharmacol Ther. 2016;44(4):411–421. https://doi.org/10.1111/apt.13702.

  18. Lombardi R, Lever R, Smith C, et al. Liver test abnormalities in patients with HIV monoinfection: Assessment with simple noninvasive fibrosis markers. Ann Gastroenterol. 2017;30(3):349–356. https://doi.org/10.20524/aog.2017.0141.

  19. Vuille-Lessard É, Lebouché B, Lennox L, et al. Nonalcoholic fatty liver disease diagnosed by transient elastography with controlled attenuation parameter in unselected HIV monoinfected patients. AIDS. 2016;30(17):2635–2643. https://doi.org/10.1097/QAD.0000000000001241.

  20. Price JC, Dodge JL, Ma Y, et al. Controlled attenuation parameter and magnetic resonance spectroscopy-measured liver steatosis are discordant in obese HIV-infected adults. AIDS. 2017;31(15):2119–2125. https://doi.org/10.1097/QAD.0000000000001601.

  21. Kardashian A, Ma Y, Scherzer R, et al. Sex differences in the association of HIV infection with hepatic steatosis. AIDS. 2017;31(3):365–373. https://doi.org/10.1097/QAD.0000000000001334.

  22. Perazzo H, Cardoso SW, Yanavich C, et al. Predictive factors associated with liver fibrosis and steatosis by transient elastography in patients with HIV mono-infection under long-term combined antiretroviral therapy. J Int AIDS Soc. 2018;21(11):e25201. https://doi.org/10.1002/jia2.25201.

  23. Lemoine M, Assoumou L, De Wit S, et al. Diagnostic accuracy of noninvasive markers of steatosis, NASH, and liver fibrosis in HIV-Monoinfected individuals at risk of nonalcoholic fatty liver disease (NAFLD): Results from the ECHAM study. J Acquir Immune Defic Syndr. 2019;80(4):e86–e94. https://doi.org/10.1097/QAI.0000000000001936.

  24. •• Maurice JB, Patel A, Scott AJ, Patel K, Thursz M, Lemoine M. Prevalence and risk factors of nonalcoholic fatty liver disease in HIV-monoinfection. AIDS. 2017;31(11):1621–1632. https://doi.org/10.1097/QAD.0000000000001504. Important meta-analysis examining the prevalence of and risk factors for NAFLD and NASH using both non-invasive techniques and liver biopsy.

  25. Lemoine M, Barbu V, Girard PM, et al. Altered hepatic expression of SREBP-1 and PPARγ is associated with liver injury in insulin-resistant lipodystrophic HIV-infected patients. AIDS. 2006;20(3):387–395. https://doi.org/10.1097/01.aids.0000206503.01536.11.

  26. Mohammed SS, Aghdassi E, Salit IE, et al. HIV-positive patients with nonalcoholic fatty liver disease have a lower body mass index and are more physically active than HIVnegative patients. J Acquir Immune Defic Syndr. 2007;45(4):432–438. https://doi.org/10.1097/QAI.0b013e318074efe3.

  27. Ingiliz P, Valantin MA, Duvivier C, et al. Liver damage underlying unexplained transaminase elevation in human immunodeficiency virus-1 mono-infected patients on antiretroviral therapy. Hepatology. 2009;49(2):436–442. https://doi.org/10.1002/hep.22665.

  28. Sterling RK, Smith PG, Brunt EM. Hepatic steatosis in human immunodeficiency virus: A prospective study in patients without viral hepatitis, diabetes, or alcohol abuse. J Clin Gastroenterol. 2013;47(2):182–187. https://doi.org/10.1097/MCG.0b013e318264181d.

  29. • Morse CG, McLaughlin M, Matthews L, et al. Nonalcoholic steatohepatitis and hepatic fibrosis in HIV-1-monoinfected adults with elevated aminotransferase levels on antiretroviral therapy. Clin Infect Dis. 2015;60(10):1569–1578. https://doi.org/10.1093/cid/civ101; This study of 62 HIV-monoinfected individuals who underwent liver biopsy abnormal liver enzymes demonstrated that the majority had clinically significant liver pathology with NASH being the most common histologic abnormality.

  30. • Vodkin I, Valasek MA, Bettencourt R, Cachay E, Loomba R. Clinical, biochemical and histological differences between HIV-associated NAFLD and primary NAFLD: A casecontrol study. Aliment Pharmacol Ther. 2015;41(4):368–378. https://doi.org/10.1111/apt.13052; This retrospective case-control study of 33 HIV+ patients with NAFLD and 33 HIV- patients with NAFLD found that the HIV+ group had increased liver disease severity and higher NASH prevalence on biopsy as compared to the HIV- group.

  31. Iogna Prat L, Roccarina D, Lever R, et al. Etiology and severity of liver disease in HIV-positive patients with suspected NAFLD: Lessons from a cohort with available liver biopsies. J Acquir Immune Defic Syndr. 2019;80(4):474–480. https://doi.org/10.1097/QAI.0000000000001942.

  32. Buzzetti E, Pinzani M, Tsochatzis EA. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism. 2016;65(8):1038–1048. https://doi.org/10.1016/j.metabol.2015.12.012.

  33. Marchesini G, Brizi M, Morselli-Labate AM, et al. Association of nonalcoholic fatty liver disease with insulin resistance. Am J Med. 1999;107(5):450–455. https://doi.org/10.1016/S0002-9343(99)00271-5.

  34. Odegaard JI, Ricardo-Gonzalez RR, Red Eagle A, et al. Alternative M2 Activation of Kupffer Cells by PPARδ Ameliorates Obesity-Induced Insulin Resistance. Cell Metab. 2008;7(6):496–507. https://doi.org/10.1016/j.cmet.2008.04.003.

  35. Matsuzawa N, Takamura T, Kurita S, et al. Lipid-induced oxidative stress causes steatohepatitis in mice fed an atherogenic diet. Hepatology. 2007;46(5):1392–1403. https://doi.org/10.1002/hep.21874.

  36. Pessayre D, Fromenty B. NASH: a mitochondrial disease. J Hepatol. 2005;42(6):928–940. https://doi.org/10.1016/j.jhep.2005.03.004.

  37. Ricci C, Pastukh V, Leonard J, et al. Mitochondrial DNA damage triggers mitochondrialsuperoxide generation and apoptosis. Am J Physiol Physiol. 2007;294(2):C413c422. https://doi.org/10.1152/ajpcell.00362.2007.

  38. Remmen H Van, Richardson A. Oxidative damage to mitochondria and aging. Exp Gerontol. 2001;36(7):957–968. https://doi.org/10.1016/S0531-5565(01)00093-6.

  39. Tilg H, Cani PD, Mayer EA. Gut microbiome and liver diseases. Gut. 2016;65(12):2035–2044. https://doi.org/10.1136/gutjnl-2016-312729.

  40. Abdel-Misih SRZ, Bloomston M. Liver Anatomy. Surg Clin North Am. 2010;90(4):643–653. https://doi.org/10.1016/j.suc.2010.04.017.

  41. Cani PD, Amar J, Iglesias MA, et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes. 2007;56(7):1761–1772. https://doi.org/10.2337/db06-1491.

  42. Lemoine M, Lacombe K, Bastard JP, et al. Metabolic syndrome and obesity are the cornerstones of liver fibrosis in HIV-monoinfected patients. AIDS. 2017;31(14):1955–1964. https://doi.org/10.1097/QAD.0000000000001587.

  43. Gutierrez AD, Balasubramanyam A. Dysregulation of glucose metabolism in HIV patients: Epidemiology, mechanisms, and management. Endocrine. 2012;41(1):1–10. https://doi.org/10.1007/s12020-011-9565-z.

  44. Nou E, Lo J, Grinspoon SK. Inflammation, immune activation, and cardiovascular disease in HIV. AIDS. 2016;30(10):1495–1509. https://doi.org/10.1097/QAD.0000000000001109.

  45. Armah KA, McGinnis K, Baker J, et al. HIV status, burden of comorbid disease, and biomarkers of inflammation, altered coagulation, and monocyte activation. Clin Infect Dis. 2012;55(1):126–136. https://doi.org/10.1093/cid/cis406.

  46. Grunfeld C, Pang M, Doerrler W, Shigenaga JK, Jensen P, Feingold KR. Lipids, lipoproteins, triglyceride clearance, and cytokines in human immunodeficiency virus infection and the acquired immunodeficiency syndrome. J Clin Endocrinol Metab. 1992;74(5):1045–1052. https://doi.org/10.1210/jcem.74.5.1373735.

  47. Brenchley JM, Price DA, Schacker TW, et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med. 2006;12(12):1365–1371. https://doi.org/10.1038/nm1511.

  48. Dillon SM, Frank DN, Wilson CC. The gut microbiome and HIV-1 pathogenesis: A twoway street. AIDS. 2016;30(18):2737–2751. https://doi.org/10.1097/QAD.0000000000001289.

  49. Lin W, Weinberg EM, Chung RT. Pathogenesis of accelerated fibrosis in HIV/HCV coinfection. J Infect Dis. 2013;207(1):S13–S18. https://doi.org/10.1093/infdis/jis926.

  50. Mosoian A, Zhang L, Hong F, et al. Frontline Science: HIV infection of Kupffer cells results in an amplified proinflammatory response to LPS. J Leukoc Biol. 2017;101(5):1983–1090. https://doi.org/10.1189/jlb.3hi0516-242r.

  51. Miro O, Lopez S, Martinez E, et al. Mitochondrial Effects of HIV Infection on the Peripheral Blood Mononuclear Cells of HIV-Infected Patients Who Were Never Treated with Antiretrovirals. Clin Infect Dis. 2004;39(5):710–716. https://doi.org/10.1086/423176.

  52. Miura T, Goto M, Hosoya N, et al. Depletion of mitochondrial DNA in HIV-1-infected patients and its amelioration by antiretroviral therapy. J Med Virol. 2003;70(4):497–505. https://doi.org/10.1002/jmv.10423.

  53. Wei Y, Rector RS, Thyfault JP, Ibdah JA. Nonalcoholic fatty liver disease and mitochondrial dysfunction. World J Gastroenterol. 2008;66(3):936–952. https://doi.org/10.3748/wjg.14.193.

  54. Kakuda TN, Brundage RC, Anderson PL, Fletcher C V. Nucleoside reverse transcriptase inhibitor-induced mitochondrial toxicity as an etiology for lipodystrophy. AIDS. 1999;13(16):2311–2312. https://doi.org/10.1097/00002030-199911120-00019.

  55. Blanco F, Barreiro P, Ryan P, et al. Risk factors for advanced liver fibrosis in HIVinfected individuals: Role of antiretroviral drugs and insulin resistance. J Viral Hepat. 2011;18(1):11–16. https://doi.org/10.1111/j.1365-2893.2009.01261.x.

  56. Behrens G, Dejam A, Schmidt H, et al. Impaired glucose tolerance, beta cell function and lipid metabolism in HIV patients under treatment with protease inhibitors. AIDS. 1999;13(10):F63–F70. https://doi.org/10.1097/00002030-199907090-00001.

  57. Liang JS, Distler O, Cooper DA, et al. HIV protease inhibitors protect apolipoprotein B from degradation by the proteasome: A potential mechanism for protease inhibitorinduced hyperlipidemia. Nat Med. 2001;7(12):1327–1331. https://doi.org/10.1038/nm1201-1327.

  58. Petit JM, Duong M, Florentin E, et al. Increased VLDL-apoB and IDL-apoB production rates in nonlipodystrophic HIV-infected patients on a protease inhibitor-containing regimen. J Lipid Res. 2003;44(9):1692–1700. https://doi.org/10.1194/jlr.m300041-jlr200.

  59. Bonjoch A, Pou C, Pérez-Álvarez N, et al. Switching the third drug of antiretroviral therapy to maraviroc in aviraemic subjects: A pilot, prospective, randomized clinical trial. J Antimicrob Chemother. 2013;68(6):1382–1387. https://doi.org/10.1093/jac/dks539.

  60. Norwood J, Turner M, Bofill C, et al. Weight gain in persons with HIV switched from efavirenz-based to integrase strand transfer inhibitor-based regimens. J Acquir Immune Defic Syndr. 2017;76(5):527–531. https://doi.org/10.1097/QAI.0000000000001525.

  61. Browning JD. Statins and hepatic steatosis: Perspectives from the Dallas heart study. Hepatology. 2006;44(2):466–471. https://doi.org/10.1002/hep.21248.

  62. Browning JD, Szczepaniak LS, Dobbins R, et al. Prevalence of hepatic steatosis in an urban population in the United States: Impact of ethnicity. Hepatology. 2004;40(6):1387–1395. https://doi.org/10.1002/hep.20466.

  63. Poynard T, Ratziu V, Naveau S, et al. The diagnostic value of biomarkers (SteatoTest) for the prediction of liver steatosis. Comp Hepatol. 2005;4:10. https://doi.org/10.1186/1476-5926-4-10.

  64. Bedogni G, Bellentani S, Miglioli L, et al. The Fatty Liver Index: a simple and accurate predictor of hepatic steatosis in the general population. BMC Gastroenterol. 2006;6:33. https://doi.org/10.1186/1471-230X-6-33.

  65. Machado M V., Cortez-Pinto H. Non-invasive diagnosis of non-alcoholic fatty liver disease. A critical appraisal. J Hepatol. 2013;58(5):1007–1019. https://doi.org/10.1016/j.jhep.2012.11.021.

  66. Hernaez R, Lazo M, Bonekamp S, et al. Diagnostic accuracy and reliability of ultrasonography for the detection of fatty liver: A meta-analysis. Hepatology. 2011;54(3):1082–1090. https://doi.org/10.1002/hep.24452.

  67. de Lédinghen V, Vergniol J, Foucher J, Merrouche W, le Bail B. Non-invasive diagnosis of liver steatosis using controlled attenuation parameter (CAP) and transient elastography. Liver Int. 2012;31(4):848–855. https://doi.org/10.1111/j.1478-3231.2012.02820.x.

  68. Karlas T, Petroff D, Sasso M, et al. Individual patient data meta-analysis of controlled attenuation parameter (CAP) technology for assessing steatosis. J Hepatol. 2017;66(5):1022–1030. https://doi.org/10.1016/j.jhep.2016.12.022.

  69. Imajo K, Kessoku T, Honda Y, et al. Magnetic Resonance Imaging More Accurately Classifies Steatosis and Fibrosis in Patients With Nonalcoholic Fatty Liver Disease Than Transient Elastography. Gastroenterology. 2016;150(3):626–637. https://doi.org/10.1053/j.gastro.2015.11.048.

  70. Castera L, Friedrich-Rust M, Loomba R. Noninvasive Assessment of Liver Disease in Patients With Nonalcoholic Fatty Liver Disease. Gastroenterology. 2019;156(5):1264–1281. https://doi.org/10.1053/j.gastro.2018.12.036.

  71. Morse CG, McLaughlin M, Proschan M, et al. Transient elastography for the detection of hepatic fibrosis in HIV-monoinfected adults with elevated aminotransferases on antiretroviral therapy. AIDS. 2015;29(17):2297–2302 https://doi.org/10.1097/QAD.0000000000000841.

  72. •• Chalasani N, Younossi Z, Lavine JE, et al. The diagnosis and management of nonalcoholic fatty liver disease: Practice guidance from the American Association for the Study of Liver Diseases. Hepatology. 2018. https://doi.org/10.1002/hep.29367; Important evidence-based consensus guidelines issued by the American Association for the Study of Liver Diseases on the management of NAFLD.

  73. Ryom L, Boesecke C, Bracchi M, et al. Highlights of the 2017 European AIDS Clinical Society (EACS) Guidelines for the treatment of adult HIV-positive persons version 9.0. HIV Med. 2018. https://doi.org/10.1111/hiv.12600.

  74. European AIDS Clinical Society. European AIDS Clinical Society Guifelines 9.0. Uganda Refug Response - Mon Snapshot Oct 2017. 2017.

  75. Vilar-Gomez E, Martinez-Perez Y, Calzadilla-Bertot L, et al. Weight loss through lifestyle modification significantly reduces features of nonalcoholic steatohepatitis. Gastroenterology. 2015;149(2):367–378. https://doi.org/10.1053/j.gastro.2015.04.005.

  76. European Association for the Study of the Liver (EASL), European Association for the Study of Diabetes (EASD), European Association for the Study of Obesity (EASO). EASL-EASD-EASO Clinical Practice Guidelines for the management of non-alcoholic fatty liver disease. J Hepatol. 2016;64(6):1388–1402. https://doi.org/10.1016/j.jhep.2015.11.004; •• Important evidence-based consensus guidelines issued by the European Association for the Study of Liver Disease on the management of NAFLD.

  77. Suzuki A, Lindor K, Saver JS, et al. Effect of changes on body weight and lifestyle in nonalcoholic fatty liver disease. J Hepatol. 2005;43(6):1060–1066. https://doi.org/10.1016/j.jhep.2005.06.008.

  78. Lazo M, Solga SF, Horska A, et al. Effect of a 12-month intensive lifestyle intervention on hepatic steatosis in adults with type 2 diabetes. Diabetes Care. 2010;33(10):2156–2163. https://doi.org/10.2337/dc10-0856.

  79. Ryan MC, Itsiopoulos C, Thodis T, et al. The Mediterranean diet improves hepatic steatosis and insulin sensitivity in individuals with non-alcoholic fatty liver disease. J Hepatol. 2013;59(1):138–143. https://doi.org/10.1016/j.jhep.2013.02.012..

  80. Kistler KD, Brunt EM, Clark JM, Diehl AM, Sallis JF, Schwimmer JB. Physical activity recommendations, exercise intensity, and histological severity of nonalcoholic fatty liver disease. Am J Gastroenterol. 2011;106(3):460–468. https://doi.org/10.1038/ajg.2010.488.

  81. Eckard C, Cole R, Lockwood J, et al. Prospective histopathologic evaluation of lifestyle modification in nonalcoholic fatty liver disease: A randomized trial. Therap Adv Gastroenterol. 2013;6(4):249–259. https://doi.org/10.1177/1756283X13484078.

  82. Hashida R, Kawaguchi T, Bekki M, et al. Aerobic vs. resistance exercise in non-alcoholic fatty liver disease: A systematic review. J Hepatol. 2017;66(1):142–152. https://doi.org/10.1016/j.jhep.2016.08.023.

  83. Zhang H-J, He J, Pan L-L, et al. Effects of Moderate and Vigorous Exercise on Nonalcoholic Fatty Liver Disease. JAMA Intern Med. 2016;176(8):1074–1082. https://doi.org/10.1001/jamainternmed.2016.3202.

  84. Keating SE, Hackett DA, George J, Johnson NA. Exercise and non-alcoholic fatty liver disease: A systematic review and meta-analysis. J Hepatol. 2012;57(1):157–166. https://doi.org/10.1016/j.jhep.2012.02.023.

  85. Keating SE, Hackett DA, Parker HM, et al. Effect of aerobic exercise training dose on liver fat and visceral adiposity. J Hepatol. 2015;63(1):174–182. https://doi.org/10.1016/j.jhep.2015.02.022.

  86. Thoma C, Day CP, Trenell MI. Lifestyle interventions for the treatment of non-alcoholic fatty liver disease in adults: A systematic review. J Hepatol. 2012;56(1):255–266. https://doi.org/10.1016/j.jhep.2011.06.010.

  87. Sanyal AJ, Chalasani N, Kowdley K V., et al. Pioglitazone, Vitamin E, or Placebo for Nonalcoholic Steatohepatitis. N Engl J Med. 2010;362(18):1675–1685. https://doi.org/10.1056/NEJMoa0907929.

  88. Schürks M, Glynn RJ, Rist PM, Tzourio C, Kurth T. Effects of vitamin E on stroke subtypes: Meta-analysis of randomised controlled trials. BMJ. 2010;341:c5702. https://doi.org/10.1136/bmj.c5702.

  89. Klein EA, Thompson IM, Tangen CM, et al. Vitamin E and the risk of prostate cancer: The selenium and vitamin E cancer prevention trial (SELECT). JAMA - J Am Med Assoc. 2011;306(14):1549–1556. https://doi.org/10.1001/jama.2011.1437.

  90. Cusi K, Orsak B, Bril F, et al. Long-term pioglitazone treatment for patients with nonalcoholic steatohepatitis and prediabetes or type 2 diabetes mellitus a randomized trial. Ann Intern Med. 2016;165(5):305–315. https://doi.org/10.7326/M15-1774.

  91. Pop LM, Lingvay I, Yuan Q, Li X, Adams-Huet B, Maalouf NM. Impact of pioglitazone on bone mineral density and bone marrow fat content. Osteoporos Int. 2017;28(11):3261–3269. https://doi.org/10.1007/s00198-017-4164-3.

  92. Liao HW, Saver JL, Wu YL, Chen TH, Lee M, Ovbiagele B. Pioglitazone and cardiovascular outcomes in patients with insulin resistance, pre-diabetes and type 2 diabetes: A systematic review and meta-analysis. BMJ Open. 2017;7(1):e013927. https://doi.org/10.1136/bmjopen-2016-013927.

  93. Hampp C, Pippins J. Pioglitazone and bladder cancer: FDA’s assessment. Pharmacoepidemiol Drug Saf. 2017;26(2):117–118. https://doi.org/10.1002/pds.4154.

  94. • Friedman SL, Ratziu V, Harrison SA, et al. A randomized, placebo-controlled trial of cenicriviroc for treatment of nonalcoholic steatohepatitis with fibrosis. Hepatology. 2018;67(5):1754–1767. https://doi.org/10.1002/hep.29477; This randomized control trial conducted in 289 subjects with NASH found that there was a decrease in fibrosis stage in those on cenicriviroc, a CCR5/CCR2 antagonist, compared to placebo.

  95. Tacke F. Cenicriviroc for the treatment of non-alcoholic steatohepatitis and liver fibrosis. Expert Opin Investig Drugs. 2018;27(3):301–311. https://doi.org/10.1080/13543784.2018.1442436.

  96. Klibanov OM, Williams SH, Iler CA. Cenicriviroc, an orally active CCR5 antagonist for the potential treatment of HIV infection. Curr Opin Investig Drugs. 2010;11(8):940–950.

  97. Thompson M, Saag M, Dejesus E, et al. A 48-week randomized phase 2b study evaluating cenicriviroc versus efavirenz in treatment-naive HIV-infected adults with C-C chemokine receptor type 5-tropic virus. AIDS. 2016;30(6):869–878. https://doi.org/10.1097/QAD.0000000000000988.

  98. Konikoff FM, Leikin-Frenkel A, Goldiner I, et al. Biliary and systemic effects of fatty acid bile acid conjugates. Eur J Gastroenterol Hepatol. 2003;15(6):649–655. https://doi.org/10.1097/00042737-200306000-00012.

  99. Safadi R, Konikoff FM, Mahamid M, et al. The Fatty Acid-Bile Acid Conjugate Aramchol Reduces Liver Fat Content in Patients With Nonalcoholic Fatty Liver Disease. Clin Gastroenterol Hepatol. 2014;12(12):2085–2091. https://doi.org/10.1016/j.cgh.2014.04.038.

  100. • Ajmera VH, Cachay E, Ramers C, et al. Novel MRI assessment of treatment response in HIV-associated NAFLD : a randomized trial of an SCD 1 inhibitor (ARRIVE Trial). Hepatology. 2019. https://doi.org/10.1002/hep.30674; This double-blind, randomized, placebo-controlled trial of 50 patients with HIV-associated NAFLD found no significant reduction in hepatic fat or change body fat and muscle composition with 12 weeks of aramchol.

  101. Stanley TL, Feldpausch MN, Oh J, et al. Effect of tesamorelin on visceral fat and liver fat in HIV-infected patients with abdominal fat accumulation: A randomized clinical trial. JAMA - J Am Med Assoc. 2014;312(4):380–390. https://doi.org/10.1001/jama.2014.8334.

  102. Calza L, Colangeli V, Borderi M, et al. Improvement in liver steatosis after the switch from a ritonavir-boosted protease inhibitor to raltegravir in HIV-infected patients with non-alcoholic fatty liver disease. Infect Dis (Auckl). 2019;51(8):593–601. https://doi.org/10.1080/23744235.2019.1629008.

  103. Federico A, Dallio M, Godos J, Loguercio C, Salomone F. Targeting gut-liver axis for the treatment of nonalcoholic steatohepatitis: Translational and clinical evidence. Transl Res. 2016;167(1):116–124. https://doi.org/10.1016/j.trsl.2015.08.002.

  104. Loguercio C, Federico A, Tuccillo C, et al. Beneficial effects of a probiotic VSL#3 on parameters of liver dysfunction in chronic liver diseases. J Clin Gastroenterol. 2005;39(6):540–543.

  105. Gangarapu V, Ince AT, Baysal B, et al. Efficacy of rifaximin on circulating endotoxins and cytokines in patients with nonalcoholic fatty liver disease. Eur J Gastroenterol Hepatol. 2015;27(7):840–845. https://doi.org/10.1097/MEG.0000000000000348.

  106. Tenorio AR, Chan ES, Bosch RJ, et al. Rifaximin has a marginal impact on microbial translocation, T-cell activation and inflammation in HIV-positive immune non-responders to antiretroviral therapy - ACTG A5286. J Infect Dis. 2015;211(5):780–790. https://doi.org/10.1093/infdis/jiu515.

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jennifer C. Price.

Ethics declarations

Conflict of Interest:

Alyson Kaplan declares no potential conflicts of interest.

Jennifer C. Price reports grants from Gilead Sciences and Merck and ownership interest (spouse) in Abbvie, Bristol-Myers Squibb, Johnson and Johnson, and Merck, and is an advisory board member of Surrozon.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

This article is part of the Topical Collection on Fatty Liver Disease

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kaplan, A., Price, J.C. HIV-Associated NAFLD: Disease Burden and Management. Curr Hepatology Rep 18, 482–491 (2019). https://doi.org/10.1007/s11901-019-00501-0

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11901-019-00501-0

Keywords

Navigation