NAFLD Is Increasingly Affecting Patients With HIV

Publication
Article
ContagionContagion, April 2021 (Vol. 06, No. 02)
Volume 06
Issue 02

The prevalence of this disease in this patient population is much higher than in the general public.

HIV

Nonalcoholic fatty liver disease (NAFLD) is the hepatic manifestation of metabolic syndrome and encompasses a spectrum of disorders, including simple hepatic steatosis (fatty liver), nonalcoholic steatohepatitis (NASH), and cirrhosis. NAFLD is defined by the presence of 5% or greater hepatic steatosis in patients without risk factors of heavy alcohol consumption, viral hepatitis, treatment with steatogenic medications, or genetic causes of hepatic fat deposition.1 NASH is a histological diagnosis characterized by inflammation, ballooning of hepatocytes and fibrosis or nonalcoholic fatty liver that is not associated with hepatocellular damage. Worldwide, the prevalence of NAFLD in the general population is rising and estimated to be 25%.2 This mirrors the rising prevalence of obesity, type 2 diabetes (T2DM), and metabolic syndrome. The disease is most prevalent in the Middle East and South America, whereas it is lowest in Africa. Patients with NAFLD experience increased morbidity and mortality, which are most commonly attributed to cardiovascular disease when compared with matched controls.3 In the United States, NAFLD is the third most common cause of hepatocellular carcinoma, with up to 13% of cases occurring in absence of cirrhosis.4

With longer life expectancy and declining AIDS-associated mortality, non–AIDS-related comorbidities have emerged as diagnostic and treatment problems in persons with HIV (PWH). Chronic liver disease is the second most common cause of non–AIDS-related mortality in PWH.5 Results of a US study of 47,062 PWH from 2006 to 2016 showed that 22% had some form of liver disease. The increased prevalence of liver disease is multifactorial and attributable to NAFLD, alcohol use, and viral hepatitis.6 In a meta-analysis of PWH, the prevalence of NAFLD, biopsy-proven NASH, and fibrosis were 35%, 42% and 22% respectively, which is significantly higher than that of the general population.7

General Pathogenesis and Risk Factors

Insulin resistance (IR) is a key factor in pathogenesis of hepatic steatosis. Dietary factors, including high consumption of fats and sugars, have been linked to NAFLD development.8 IR causes triglycerides to mobilize from the adipose tissue to the liver, whereas IR-associated hyperinsulinemia promotes hepatic de novo lipogenesis.9 Hepatic steatosis is a benign condition that can progress to NASH in certain individuals, and NASH can progress to cirrhosis and liver cancer. Mitochondrial dysfunction plays an important role in dysregulation of the liver’s lipid environment. Enhanced mitochondrial fatty acid oxidation induces production of reactive oxygen species, which results in oxidative stress and development of NASH.10

Pathogenesis and Risk Factors Specific to HIV Infection

There are multiple complex mechanisms by which HIV infection increases the risk of NAFLD. These are incompletely understood and include comorbid risk factors, direct viral effects, and adverse effects of antiretroviral therapy (ART).

Components of metabolic syndrome are more common among PWH. T2DM is 4 times more prevalent in PWH, and dyslipidemia and hypertension are also more common.11–13 Results of a US outpatient study revealed that nearly half of patients were obese at the time of starting ART, and in the next 2 years, 20% moved up to a higher BMI category.14 Achhra et al showed that weight gain in the first year after initiation of ART was associated with increased risk of cardiovascular disease and diabetes.15 NAFLD, which is called lean NAFLD when it affects patients with a BMI of less than 25 kg/m2, occurs in 1 in 4 lean PWH. Lean patients with NAFLD had more metabolic derangements, such as higher triglyceride and ALT levels and lower HDL levels, than lean patients without NAFLD. They had longer duration of HIV infection and higher CD4 lymphocyte counts, and they were more likely to maintain viral suppression. This clinical phenotype may be related to genetic factors, change in gut microbiota, dysfunctional adipose tissue, and improved ability to adapt to an excess intake of calories. However, the exact mechanism is not completely understood.16

Direct viral effects also may contribute to NAFLD. Depletion of CD4+ lymphocytes in the gut causes disruption of the gut epithelial barrier, facilitating microbial translocation into the portal and systemic translocation. This promotes liver fibrosis by activation of hepatic Kupffer cells and induction of proinflammatory cytokines, including tumor necrosis factor, interleukin (IL)-1, and IL-6.17,18 HIV-induced mitochondrial dysfunction results in production of reactive oxygen species, which causes oxidative stress that is known to increase fat accumulation in hepatocytes.19

Finally, certain ARTs may contribute to the pathogenesis of NAFLD. Older nucleoside reverse transcriptase inhibitors, (eg, stavudine, zidovudine, and didanosine) inhibit mitochondrial RNA and have been associated with liver fibrosis. Although these drugs are no longer recommended, associated adverse effects may be irreversible.7,20 Protease inhibitors increase central adiposity, decrease hepatic clearance of very-low-density lipoproteins, increase hepatic triglyceride production, and potentially contribute to NAFLD.21,22 ART in the current era, particularly integrase strand transfer inhibitors, is associated with weight gain with metabolic consequences that are not yet known.23

Diagnosis

The American Association for the Study of Liver Diseases does not recommend screening high-risk groups for NAFLD. However, the European AIDS Clinical Society recommends screening all PWH with metabolic syndrome for NAFLD by ultrasound, evaluation of liver enzymes, and serum fibrosis biomarkers.1,24

The severity of NAFLD is assessed by stratifying the degree of steatosis. Simple hepatic steatosis is generally benign, whereas NASH can progress to cirrhosis, liver failure, and liver cancer. Vodkin et al compared 66 biopsy-proven cases of NAFLD in patients who were infected with HIV versus those who were HIV-negative. They found that those with HIV-associated NAFLD were more likely to have definite steatohepatitis (36.4% vs 62.7%, P=0.027) and had higher scores for noninvasive markers of advanced disease.25 Noninvasive tests have varying degrees of performance in assessing NAFLD in people living with HIV (PLWH).22 Of the imaging modalities, MRI and transient elastography have the best performing characteristics. APRI score (AST: platelet count) and FIB-4 (fibrosis-4 score) can be considered when evaluating patients who are HIV-positive with NAFLD.26 Although liver biopsy is the gold standard for diagnosing and staging NAFLD, it is invasive, prone to sampling errors, and has risk for complications.

Management

Managing NAFLD consists of treating liver disease as well as its associated metabolic comorbidities such as obesity, hyperlipidemia, and diabetes. First-line therapy in NAFLD/NASH consists of lifestyle changes focusing on physical activity and diet. Weight loss of 10% is associated with resolution of steatosis and improvement of fibrosis in most individuals.27 Liver-specific dietary recommendations, such as diet composition, are generally lacking or controversial. Patients with NAFLD must avoid routine or heavy alcohol consumption, and many practitioners recommend complete abstinence, especially for patients with advanced NAFLD.

Bariatric surgery improves comorbid diseases and improves long-term outcomes in obese individuals. A systematic review of studies focusing on pre- and postoperative liver enzymes and histology revealed that bariatric surgery is associated with significant reduction in liver enzymes and histological features of NAFLD, including steatosis, fibrosis, and inflammation. However, further studies are needed before recommending bariatric surgery as a treatment for NAFLD.28

Although there are currently no FDA-approved treatments for NAFLD, multiple agents are under investigation. Pioglitazone has been shown to improve liver histology in patients with and without T2DM and biopsy-proven NASH.29 This effect was also seen among patients with HIV/hepatitis C (HCV) coinfection.30 In individuals with and without HIV, vitamin E use improves steatosis and steatohepatitis but does not improve hepatic fibrosis.31,32

Tesamorelin is an FDA-approved growth hormone (GH)-releasing hormone analog that restores endogenous pulsatile GH secretion and reduces visceral fat in individuals in PWH. A randomized-controlled trial (RCT) of PWH showed that tesamorelin significantly reduced hepatic fat fraction and prevented the progression of fibrosis but did not improve existing fibrosis.33

Obeticholic acid, a farnesoid X receptor agonist, plays a central role in the regulation of bile acids and metabolism. Interim analysis of an RCT revealed that it improved fibrosis and markers of NASH disease activity.34 However, due to safety concerns, it is currently not approved for treatment of NASH.

Maraviroc, a chemokine receptor 5 (CCR5) antagonist, may reduce the incidence of NAFLD as an add-on therapy for individuals with NAFLD and well-controlled HIV infection.35 Cenicriviroc, a chemokine 2 and 5 receptor inhibitor, may also prove to be an effective treatment for NASH.36 Other investigational drugs undergoing evaluation include elafibranor, a peroxisome proliferator-activator, and resmetirom, a thyroid hormone receptor β-agonist.37,38

Conclusion

Addressing NAFLD is an important issue for PWH. Because these individuals are living longer, they are at increased risk of developing chronic diseases such as obesity, T2DM, and NAFLD. PWH with features of metabolic syndrome should be evaluated for NAFLD. As both NAFLD and HIV may lead to increased risk of cardiovascular disease, managing dyslipidemia, hypertension, T2DM, and obesity may improve patients’ longevity and quality of life.39

Zahra Qamar, MD, is a second-year infectious disease fellow at Thomas Jefferson University Hospital in Philadelphia, Pennsylvania.

Steven F. Solga, MD, is an associate professor at the University of Pennsylvania in Philadelphia. Solga has studied and published in the field of fatty liver disease.

Lisa A. Spacek, MD, PhD, FIDSA, is an associate professor and director of HIV Ambulatory Care at Thomas Jefferson University in Philadelphia, Pennsylvania. Spacek’s research focuses on HIV treatment algorithms in resource-limited settings and clinical utility of diagnostics in HIV medicine. She has authored chapters of the Johns Hopkins Antibiotic and HIV guides since 2006.

References

1. 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;67(1):328-357. doi:10.1002/hep.29367

2. Younossi ZM, Koenig AB, Abdelatif D, Fazel Y, Henry L, Wymer M. Global epidemiology of nonalcoholic fatty liver disease-meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology. 2016;64(1):73-84. doi:10.1002/hep.28431

3. Adams LA, Lymp JF, St Sauver J, et al. The natural history of nonalcoholic fatty liver disease: a population-based cohort study. Gastroenterology. 2005;129(1):113-121. doi:10.1053/j.gastro.2005.04.014

4. Mittal S, El-Serag HB, Sada YH, et al. Hepatocellular carcinoma in the absence of cirrhosis in united states veterans is associated with nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol. 2016;14(1):124-131.e1. doi:10.1016/j.cgh.2015.07.019

5. Smith CJ, Ryom L, Weber R, et al; D:A:D Study Group. 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. doi:10.1016/S0140-6736(14)60604-8

6. Paik JM, Henry L, Golabi P, Alqahtani SA, Trimble G, Younossi ZM. Presumed nonalcoholic fatty liver disease among Medicare beneficiaries with HIV, 2006-2016. Open Forum Infect Dis. 2020;7(1):ofz509. doi:10.1093/ofid/ofz509

7. 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. doi:10.1097/QAD.0000000000001504

8. Fan JG, Cao HX. Role of diet and nutritional management in non-alcoholic fatty liver disease. J Gastroenterol Hepatol. 2013;28(suppl 4):81-87. doi:10.1111/jgh.12244

9. Samuel VT, Shulman GI. Mechanisms for insulin resistance: common threads and missing links. Cell. 2012;148(5):852-871. doi:10.1016/j.cell.2012.02.017

10. Begriche K, Massart J, Robin MA, Bonnet F, Fromenty B. Mitochondrial adaptations and dysfunctions in nonalcoholic fatty liver disease. Hepatology. 2013;58(4):1497-1507. doi:10.1002/hep.26226

11. Brown TT, Cole SR, Li X, et al. Antiretroviral therapy and the prevalence and incidence of diabetes mellitus in the multicenter AIDS cohort study. Arch Intern Med. 2005;165(10):1179-1184. doi:10.1001/archinte.165.10.1179

12. Fahme SA, Bloomfield GS, Peck R. Hypertension in HIV-infected adults: novel pathophysiologic mechanisms. Hypertension. 2018;72(1):44-55. doi:10.1161/HYPERTENSIONAHA.118.10893

13. Dubé MP, Stein JH, Aberg JA, et al; Adult AIDS Clinical Trials Group Cardiovascular Subcommittee; HIV Medical Association of the Infectious Disease Society of America. Guidelines for the evaluation and management of dyslipidemia in human immunodeficiency virus (HIV)-infected adults receiving antiretroviral therapy: recommendations of the HIV Medical Association of the Infectious Disease Society of America and the Adult AIDS Clinical Trials Group. Clin Infect Dis. 2003;37(5):613-627. doi:10.1086/378131

14. Tate T, Willig AL, Willig JH, et al. HIV infection and obesity: where did all the wasting go? Antivir Ther (Lond). 2012;17(7):1281-1289. doi:10.3851/IMP2348

15. Achhra AC, Mocroft A, Reiss P, et al; D:A:D Study Group. Short-term weight gain after antiretroviral therapy initiation and subsequent risk of cardiovascular disease and diabetes: the D:A:D study. HIV Med. 2016;17(4):255-268. doi:10.1111/hiv.12294

16. Cervo A, Milic J, Mazzola G, et al. Prevalence, predictors and severity of lean non-alcoholic fatty liver disease in HIV-infected patients. Clin Infect Dis. April 13, 2020. doi:10.1093/cid/ciaa430

17. 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. doi:10.1038/nm1511

18. Sandler NG, Douek DC. Microbial translocation in HIV infection: causes, consequences and treatment opportunities. Nat Rev Microbiol. 2012;10(9):655-666. doi:10.1038/nrmicro2848

19. Pérez-Matute P, Pérez-Martínez L, Blanco JR, Oteo JA. Role of mitochondria in HIV infection and associated metabolic disorders: focus on nonalcoholic fatty liver disease and lipodystrophy syndrome. Oxid Med Cell Longev. July 21, 2013. doi:10.1155/2013/493413

20. 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. doi:10.1086/589294

21. Gervasoni C, Cattaneo D, Filice C, Galli M; Gruppo Italiano Studio-NASH in Malattie Infettive. Drug-induced liver steatosis in patients with HIV infection. Pharmacol Res. 2019;145:104267. doi:10.1016/j.phrs.2019.104267

22. Lake JE, Overton T, Naggie S, et al. Expert panel review on non-alcoholic fatty liver disease in persons with HIV. Clin Gastroenterol Hepatol. October 2020. doi:10.1016/j.cgh.2020.10.018

23. Sax PE, Erlandson KM, Lake JE, et al. Weight gain following initiation of antiretroviral therapy: risk factors in randomized comparative clinical trials. Clin Infect Dis. 2020;71(6):1379-1389. doi:10.1093/cid/ciz999

24. Ryom L, Cotter A, De Miguel R, et al; EACS Governing Board. 2019 update of the European AIDS Clinical Society Guidelines for treatment of people living with HIV version 10.0. HIV Med. 2020;21(10):617-624. doi:10.1111/hiv.12878

25. Vodkin I, Valasek MA, Bettencourt R, Cachay E, Loomba R. Clinical, biochemical and histological differences between HIV-associated NAFLD and primary NAFLD: a case-control study. Aliment Pharmacol Ther. 2015;41(4):368-378. doi:10.1111/apt.13052

26. Lemoine M, Assoumou L, De Wit S, et al; ANRS-ECHAM Group. 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. doi:10.1097/QAI.0000000000001936

27. 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.e5. doi:10.1053/j.gastro.2015.04.005

28. Bower G, Toma T, Harling L, et al. Bariatric surgery and non-alcoholic fatty liver disease: a systematic review of liver biochemistry and histology. Obes Surg. 2015;25(12):2280-2289. doi:10.1007/s11695-015-1691-x

29. Aithal GP, Thomas JA, Kaye PV, et al. Randomized, placebo-controlled trial of pioglitazone in nondiabetic subjects with nonalcoholic steatohepatitis. Gastroenterology. 2008;135(4):1176-1184. doi:10.1053/j.gastro.2008.06.047

30. Matthews L, Kleiner DE, Chairez C, et al. Pioglitazone for hepatic steatosis in hiv/hepatitis C virus coinfection. AIDS Res Hum Retroviruses. 2015;31(10):961-966. doi:10.1089/AID.2015.0093

31. Sanyal AJ, Chalasani N, Kowdley KV, et al. Pioglitazone, vitamin E, or placebo for nonalcoholic steatohepatitis. N Engl J Med. 2010;362(18):1675-1685. doi:10.1056/NEJMoa0907929

32. Sebastiani G, Saeed S, Lebouche B, et al. Vitamin E is an effective treatment for nonalcoholic steatohepatitis in HIV mono-infected patients. AIDS. 2020;34(2):237-244. doi:10.1097/QAD.0000000000002412

33. Stanley TL, Fourman LT, Feldpausch MN, et al. Effects of tesamorelin on non-alcoholic fatty liver disease in HIV: a randomised, double-blind, multicentre trial. Lancet HIV. 2019;6(12):e821-e830. doi:10.1016/S2352-3018(19)30338-8

34. Younossi ZM, Ratziu V, Loomba R, et al. Obeticholic acid for the treatment of non-alcoholic steatohepatitis: interim analysis from a multicentre, randomised, placebo-controlled phase 3 trial. Lancet. 2019;394(10215):2184-2196. doi:10.1016/S0140-6736(19)33041-7

35. Adding MAraViroc &/or METformin for hepatic steatosis in people living with HIV (MAVMET). ClinicalTrials.gov. Updated January 14, 2020. Accessed February 22, 2021. https://clinicaltrials.gov/ct2/show/NCT03129113

36. Anstee QM, Neuschwander-Tetri BA, Wong VWS, et al. Cenicriviroc for the treatment of liver fibrosis in adults with nonalcoholic steatohepatitis: AURORA Phase 3 study design. Contemp Clin Trials. 2020;89:105922. doi:10.1016/j.cct.2019.105922

37. Botta M, Audano M, Sahebkar A, Sirtori CR, Mitro N, Ruscica M. PPAR agonists and metabolic syndrome: an established role? Int J Mol Sci. 2018;19(4):1197. doi:10.3390/ijms19041197

38. A phase 3 study to evaluate the safety and biomarkers of resmetirom (MGL-3196) in non alcoholic fatty liver disease (NAFLD) patients. ClinicalTrials.gov. Updated July 7, 2020. Accessed February 22, 2021. https://clinicaltrials.gov/ct2/show/NCT04197479

39. Dulai PS, Singh S, Patel J, et al. Increased risk of mortality by fibrosis stage in nonalcoholic fatty liver disease: Systematic review and meta-analysis. Hepatology. 2017;65(5):1557-1565. doi:10.1002/hep.29085

Related Videos
© 2024 MJH Life Sciences

All rights reserved.