Adverse Effects of Highly Active Antiretroviral Therapy in Developing ...

165 downloads 0 Views 233KB Size Report
Sep 6, 2007 - 1Yale School of Medicine, New Haven, Connecticut; 2Department of ... 3Division of Infectious Diseases, Miriam Hospital, Brown University, Providence, ...... mivudine and nevirapine (d4T/3TC/NVP) in an urban district hos-.
REVIEW ARTICLE

HIV/AIDS

Adverse Effects of Highly Active Antiretroviral Therapy in Developing Countries Ramnath Subbaraman,1 Sreekanth Krishna Chaguturu,2 Kenneth H. Mayer,3 Timothy P. Flanigan,3 and Nagalingeswaran Kumarasamy4 1

Yale School of Medicine, New Haven, Connecticut; 2Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston; Division of Infectious Diseases, Miriam Hospital, Brown University, Providence, Rhode Island; and 4YR Gaitonde Centre for AIDS Research and Education, Voluntary Health Services, Chennai, India

3

Recent increases in access to highly active antiretroviral therapy (HAART) have made the management of drug toxicities an increasingly crucial component of human immunodeficiency virus (HIV) care in developing countries. The spectrum of adverse effects related to HAART in developing countries may differ from that in developed countries because of the high prevalence of conditions such as anemia, malnutrition, and tuberculosis and frequent initial presentation with advanced HIV disease. The severity of adverse effects may vary as a result of host genetics and diagnostic delays attributable to inadequate laboratory monitoring. This article reviews current knowledge about toxicities related to HAART in resource-limited regions, which are in the process of rapid treatment scale-up. We conclude that initiating HAART before advanced immunosuppression, titrating doses in single-pill drug combinations to differences in patients’ body weights, providing more intensive laboratory monitoring during the initial months of therapy, and providing access to less-toxic nucleoside reverse-transcriptase inhibitors may decrease the incidence of toxicities related to HAART in resource-limited regions. Although the HIV-AIDS epidemic continues to spread in the developing world [1], reductions in the price of HAART for HIV-infected individuals in resource-constrained regions have made treatment increasingly accessible [2]. HIV care facilities in developing countries have witnessed dramatic decreases in mortality that are similar to those previously recorded in developed countries [3–5]. One by-product of increased access to HAART, however, is that management of antiretroviral drug–related toxicities is becoming an important component of HIV care in developing countries. The spectrum of adverse effects associated with HAART may vary between developed and developing countries for several reasons. First, economic con-

Received 11 August 2006; accepted 8 June 2007; electronically published 6 September 2007. Reprints or correspondence: Dr. N. Kumarasamy, YR Gaitonde Centre for AIDS Research and Education, Voluntary Health Services, Tidel Park Rd., Taramani, Chennai 600113, India ([email protected]). Clinical Infectious Diseases 2007; 45:1093–1101  2007 by the Infectious Diseases Society of America. All rights reserved. 1058-4838/2007/4508-0026$15.00 DOI: 10.1086/521150

straints limit the repertoire of accessible antiretroviral medications, making a handful of drugs responsible for most toxicities in developing countries [6]. Second, prohibitory laboratory monitoring costs may occasionally delay the diagnosis of specific toxicities, thereby increasing their severity. Third, comorbid conditions that are more prevalent in resource-limited regions, such as anemia and malnutrition; initial presentation with advanced immunosuppression; use of concomitant antituberculosis therapy (ATT); and use of herbal medications [7] may influence the incidence of adverse effects. Finally, host genetics may be associated with drug toxicities [8]; this is a relevant issue, because most antiretroviral drugs have been validated in developed countries (primarily in white populations) but are now being widely used in developing countries, where the vast majority of HIV-infected people live. This article reviews research on the adverse effects associated with HAART in the context of rapid treatment scale-up, focusing on drugs predominantly used in resource-constrained regions. Two related challenges in the management of antiretroviral therHIV/AIDS • CID 2007:45 (15 October) • 1093

apy—coadministration of HAART with ATT and immune reconstitution syndrome (IRS)—will also be discussed. NUCLEOSIDE REVERSE-TRANSCRIPTASE INHIBITORS (NRTIS) Stavudine

Next to lamivudine, stavudine is the most commonly used NRTI, because of its relatively low cost [9, 10]. Stavudine is the NRTI that is most often associated with mitochondrial toxicity, which results in high rates of lipoatrophy, peripheral neuropathy, lactic acidosis, and pancreatitis [11]. Peripheral neuropathy. From 10% to 21% of persons exposed to stavudine developed peripheral neuropathy in developed countries [12, 13]. Although symptoms usually resolve after prompt discontinuation of stavudine therapy, persistent symptoms in a subset of patients may be problematic in developing countries, where many persons rely on physical labor for survival and usually do not have disability insurance. Cohort studies from Cameroon, India, and Thailand found peripheral neuropathy rates that were similar to or, surprisingly, lower than those in developed countries [6, 14–16]. It is hard to establish whether these lower rates reflect underascertainment biases (e.g., short follow-up periods and insensitive screening tools). However, 56% of patients in a Malawian cohort developed peripheral neuropathy while receiving stavudine therapy [17]. The authors noted that one-third of their patients had a body weight !60 kg, for which a 30-mg dose of stavudine is recommended; however, only the 40-mg dose was available in the fixed-dose combination (FDC) used in the study. This highlights the necessity of the availability of FDCs with varied doses to minimize toxicity, especially in malnourished patients. Lipodystrophy and metabolic complications. The prevalence of stavudine-associated lipodystrophy in western studies has reached as high as 50%–63% [18–20]. However, many of these studies included patients who also received protease inhibitors (PIs), which independently cause lipodystrophy. The risk has been shown to be greater for those initiating HAART with a low CD4 cell count [21, 22] and a low body mass index (BMI; calculated as the weight in kilograms divided by the square of the height in meters) [23]. Because stavudine-associated lipodystrophy commonly presents as lipoatrophy (i.e., fat loss in the cheeks, arms, and buttocks), malnutrition complicates its diagnosis. Careful assessment is needed to differentiate lipoatrophy from general wasting to prevent unnecessary modifications of therapy. Some data suggest that ethnic variability effects the incidence of lipodystrophy. White race may be an independent risk factor for the development of lipodystrophy [22, 24, 25]. Although a small South Korean cohort had a 3.5% rate of lipodystrophy [26], multiple subsequent east Asian cohorts have shown rates similar to those in western studies [27–29]. Only 17% of pa1094 • CID 2007:45 (15 October) • HIV/AIDS

tients in 2 southeast Asian cohorts developed lipodystrophy, compared with 24.8% of patients in a Rwandan cohort and 46.1% in an Indian cohort [30–33]. The tendency of stavudine-associated lipoatrophy to affect facial features raises concerns that the widespread use of the drug in developing countries may increase stigma and decrease HAART adherence [31]. A study of 410 patients of Chinese ethnicity in Singapore found that lipodystrophy affected social relations for 23% of the patients and mood for 36% [27]. However, !1% of the patients wanted to discontinue therapy because of this toxicity. In contrast, a smaller study suggested that 14% of Brazilian patients considered therapy discontinuation because of this adverse effect [34]. Because toleration of lipodystrophy may be culturally specific, region-specific research may help determine the viability of stavudine use in different countries. Although zidovudine sometimes causes lipodystrophy, stavudine is more strongly associated with this adverse effect [31, 35–37]. Therefore, one approach for reducing the incidence of lipodystrophy would be to substitute zidovudine for stavudine 6–12 months after HAART initiation, when lipodystrophy may begin to develop [35]. This allows antiretroviral roll-out programs to briefly take advantage of the lower cost and better initial tolerability of stavudine. Also, stavudine-containing HAART is associated with resolution of anemia in many patients within 6 months after initiation [38–41]. Because anemia is a relative contraindication for zidovudine use, stavudine therapy could bridge the way to zidovudine therapy by reducing the risk for anemia. Even after lipodystrophy develops, substituting another NRTI for stavudine may result in partial recovery [11, 42–44]. Of all the NRTIs, stavudine therapy has been most strongly associated with dyslipidemia and hyperglycemia in western studies [45–47]. Similar metabolic changes were present in patients in an Indian cohort treated with stavudine and zidovudine and in a cohort of patients of Chinese ethnicity treated with non–PI-containing HAART [27, 31]. As access to HAART increases, patients in developing countries may increasingly face the cardiovascular consequences of altered metabolism. Lactic acidosis. Although relatively infrequent, multiple cohort studies [48–52] and case reports [53–58] from developing countries highlight concerns about timely diagnosis of life-threatening stavudine-induced lactic acidosis, for which women may be at a higher risk [49, 50, 52]. Pilot studies from Haiti and South Africa found that point-of-care testing with handheld devices measuring lactic acid levels (e.g., using finger stick blood samples) facilitated timely diagnosis of hyperlactatemia and prevented unnecessary regimen modifications in patients without increased serum lactate levels [59, 60]. Such devices may be beneficial as HIV care is decentralized to rural

locations, where lactic acid testing in laboratories may be unavailable. Zidovudine

Myelosuppression. Anemia is common in developing countries, particularly among HIV-infected individuals [61–63], and generally worsens with disease progression [64–66]. High background levels of anemia may preclude zidovudine use in many patients. Zidovudine-related anemia usually occurs within 3 months after therapy initiation [6]. Risk factors include high zidovudine dosage, increased treatment duration, low CD4 cell count, and preexisting anemia [65, 67, 68]. Studies from Nigeria, Coˆte d’Ivoire, Haiti, and India have found rates of zidovudine-related anemia of 3%–12% [5, 6, 69–71]. A study from Coˆte d’Ivoire suggested synergistic toxicity between zidovudine and cotrimoxazole. Among 498 patients already receiving cotrimoxazole prophylaxis, the introduction of zidovudine-containing HAART resulted in one-half of the cohort developing severe neutropenia. Complete recovery occurred in nearly all patients after discontinuation of cotrimoxazole therapy, suggesting that this toxicity was attributable to a drug-drug interaction between these 2 myelosuppressive drugs [70]. Among patients who discontinue stavudine therapy because of toxicity, those who cannot substitute zidovudine therapy because of persisting anemia may be left with no options, because these are the least expensive NRTIs in resource-limited regions [9, 10]. This highlights the need for increased access to NRTIs with different toxicity profiles, such as tenofovir and abacavir, in developing countries. NONNUCLEOSIDE REVERSE-TRANSCRIPTASE INHIBITORS (NNRTIS) Nevirapine

Nevirapine is the most commonly used NNRTI in developing countries because of its lower cost, compared with efavirenz [9, 10]. Hypersensitivity rash. Hypersensitivity rash occurred in 16%–20% of patients in studies in developed country [68, 72, 73]. Two US studies that disaggregated data by ethnicity found that mostly persons of Mexican origin and some persons of South American origin were at a higher risk [74, 75]. Data do not suggest a higher risk of rash among other ethnic groups, with most studies in developing countries finding rates similar to or lower than those in developed countries. Cohorts from Haiti, India, Thailand, and Malawi found nevirapine-associated rash rates of 3%–26% [5, 6, 15–17, 76]. Female patients may be at an increased risk for nevirapine-associated rash [76–79]. Initiating patients on a lower lead-in dosage of nevirapine of 200 mg once daily, followed by escalation to the full 200mg twice daily dosage after 2 weeks, helps prevent severe rashes,

such as Stevens-Johnson syndrome [72]. The use of FDCs in developing countries does not enable clinicians to titrate nevirapine therapy initiation. Without appropriate physician education, the use of the full nevirapine dose in FDCs for patients beginning HAART may increase the incidence of life-threatening rashes. Although nevirapine therapy can safely be replaced with efavirenz therapy for those who experience adverse reactions, because there is little evidence of rash cross-toxicity between the 2 drugs [76, 80], this substitution is often precluded by the higher cost of efavirenz in developing countries [10]. Hepatotoxicity. The incidence of drug-related hepatitis in US and European trials has ranged from 1% to 10% [72, 81– 83]. Cohorts from Haiti, Thailand, India, Zambia, and Malawi found similar rates of nevirapine-associated hepatotoxicity, ranging from !1% to 7% [5, 6, 15–17, 84]. A South African study reported a 17% incidence of serious hepatotoxicity (i.e., alanine aminotransferase and aspartate aminotransferase levels 15 times the upper limit of normal) among 385 patients receiving nevirapine-based regimens, compared with no cases of hepatotoxicity among 83 patients receiving efavirenz-based regimens [85]. Female patients with a BMI !18.5 had a 50% incidence of serious hepatotoxicity. Leith et al. [86] argued that the high mean CD4 cell count of 398 cells/mL at treatment initiation in this cohort may explain this elevated hepatitis rate. A retrospective analysis of prior studies found a 12-fold increased risk of severe hepatotoxicity in women with CD4 cell counts 1250 cells/mL, compared with women with CD4 cell counts !250 cells/mL (resulting in a “black box” warning for nevirapine [87]); the risk for men increased at CD4 cell counts 1400 cells/mL [86, 88, 89]. Of note, studies from Thailand and Zambia have not found this association between nevirapine-induced hepatotoxicity and CD4 cell count [84, 90]. The authors of the South African study argued that race and BMI, rather than baseline CD4 cell count, accounted for the high hepatotoxicity rate [91], because studies have found decreased nevirapine clearance in individuals with a low BMI, of black race, or with particular pharmacogenetic profiles [92, 93]. Thai ethnicity may also be a risk factor for nevirapine-associated hepatotoxicity [89]. Additional research is needed to clarify the complex relationship between race/ethnicity, baseline CD4 cell count, and the risk of nevirapine-induced hepatitis. A Thai study found that 17 (18.6%) of 91 patients receiving nevirapine therapy developed serious hepatitis [94], which may be explained by the high prevalence of hepatitis B virus (HBV) and hepatitis C virus (HCV) coinfection in this cohort. Similar to findings in western studies [95], HBV-infected patients in this study had a higher hepatotoxicity rate (57.4%) than did HBV-uninfected patients, and patients with HCV had a hepatotoxicity rate of 72.2%. Conversely, long-term nevirapine use HIV/AIDS • CID 2007:45 (15 October) • 1095

may increase the rate of progression to cirrhosis in HBV-HCV coinfected patients [96]. The authors of the Thai study, therefore, suggest that nevirapine use might be contraindicated in regions where HBV-HCV coinfection screening is unavailable and where background prevalence is 110% [94]. Prevention of nevirapine-associated liver toxicity, therefore, requires attention to multiple factors prior to HAART initiation, including female sex, high CD4 cell count at initiation, low BMI, low albumin level [85], HBV-HCV coinfection, use of ATT, and, possibly, race/ethnicity. Efavirenz

Neuropsychiatric disorders. Neuropsychiatric disorders are the most concerning adverse effects associated with efavirenz therapy with regard to tolerability and adherence. In western cohorts, one-half of patients have these symptoms at initiation of efavirenz therapy, but these symptoms usually resolve within 1 month [97]. People of African descent with a variant of hepatic enzyme CYP2B6 may experience slower clearance of efavirenz from plasma and increased neurotoxicity [98–100]. A study from Haiti supports this data on ethnic differences. The study found that 46 (10%) of 452 patients discontinued efavirenz therapy because of persistent neurotoxicity [5]; this rate is higher than that found in US studies [97]. A study from Coˆte d’Ivoire also found a high neurotoxicity rate (69%) after initiation of efavirenz therapy [101, 102]. In contrast to the Haitian study, these symptoms resolved in most patients by the third month of therapy, with only 1 patient of 808 patientmonths of follow-up discontinuing therapy because of neurotoxicity. Additional research clarifying the influence of ethnicity on efavirenz plasma levels may lead to dose adjustments, which may decrease rates of neurotoxicity in particular populations. OVERLAPPING TOXICITIES OF HAART AND ATT Dean et al. [103] highlighted the problem of overlapping toxicities when they found a high incidence (54%) of adverse events in a cohort of 188 patients coinfected with HIV and tuberculosis. These toxicities (table 1), which occurred at a Table 1.

higher rate than in previous control groups of HIV-infected patients treated with ATT in the pre-HAART era, led to interruption of HAART or ATT regimens in one-third of patients [103]. Nevirapine has multiple overlapping toxicities with ATT drugs, especially rifampicin [104]. A Thai study indicated that patients receiving ATT while receiving nevirapine therapy had a 7.4-fold increased risk of developing hepatitis and a 3-fold increased risk of developing a rash [105]. One small cohort found a high incidence (55%) of peripheral neuropathy in patients receiving both stavudine and isoniazid therapies [106], and another study found peripheral neuropathy to be the most common toxicity in a cohort of patients coinfected with HIV and tuberculosis [103]. Patients receiving both drugs should be closely monitored and should receive supplemental pyridoxine therapy to reduce the risk of isoniazidrelated neurotoxicity. Finally, as patients begin to experience failure of first-line, NNRTI-based regimens (with tuberculosis being the most common infection marking clinical failure), overlapping toxicities associated with ATT and PIs, such as hepatitis, will become increasingly important in developing countries. IRS IRS, a paradoxical worsening of clinical status after HAART initiation, is increasingly recognized as an adverse consequence of antiretroviral therapy [107–111]. Developing countries may have a higher incidence of IRS as a result of a higher burden of opportunistic infections and frequent therapy initiation in patients with low CD4 cell counts, both of which are risk factors for IRS [112–116]. For example, HIV-infected patients receiving care from Medicins Sans Frontieres programs in sub-Saharan Africa, southeast Asia, and Central America had a median CD4 cell count of 48 cells/mL at HAART initiation, placing many at a high risk for IRS [117]. Unlike in developed countries, tuberculosis is the most common pathogen involved in IRS in resource-limited countries. Indian and Thai studies found IRS rates of 15% and 13%, respectively, among patients coinfected with HIV and tuberculosis after the initiation of HAART [118, 119]. Such cases are difficult to differentiate from cases of multidrug-resistant

Overlapping toxicities associated with antiretroviral and antituberculosis drugs.

Toxicity

Antiretroviral drugs

Antituberculosis drugs

Hepatitis

Nevirapine, protease inhibitors

Rifampicin, isoniazid, pyrazinamide, ethionamide

Rash

Nevirapine, efavirenz, abacavir

Rifampicin, isoniazid, quinolones

Anemia, neutropenia Nausea, vomiting

Zidovudine Zidovudine, ritonavir, indinavir

Rifampicin, isoniazid Rifampicin, pyrazinamide, quinolones, ethionamide

Peripheral neuropathy CNS symptoms

Stavudine, didanosine, zalcitabine Efavirenz

Isoniazid, ethambutol, cycloserine Streptomycin, quinolones, cycloserine

1096 • CID 2007:45 (15 October) • HIV/AIDS

tuberculosis, as highlighted in 2 Indian case reports [120, 121]. Other reported presentations of IRS that may be more common in developing countries include exacerbations of leprosy [122, 123], leishmaniasis [124, 125], and Mycobacterium bovis infection (acquired through bacille Calmette-Gue´rin vaccination) [126]. CONCLUSIONS This review illuminates a few common trends in HAART-related toxicities that are relevant to developing countries. First, initiation of antiretroviral therapy at advanced stages of AIDS has implications beyond the obvious risk of morbidity and mortality due to opportunistic infections. Low CD4 cell count at treatment initiation is a risk factor for multiple adverse effects, including stavudine-induced peripheral neuropathy [127, 128], lipodystrophy [21, 22], and lactic acidosis [11, 129]; zidovudine-induced myelosuppression [67]; didanosine-induced pancreatitis [130]; and IRS [113]. Moreover, the high burden of opportunistic infection in patients with low CD4 cell counts increases overlapping toxicities between HAART and opportunistic infection treatments—a problem of particular concern for patients receiving ATT. Therefore, earlier HAART initiation, before the development of a low CD4 cell count and opportunistic infection, may reduce the incidence of adverse effects. Increased access to HIV testing services, with the aim of engaging more patients in long-term follow-up, may help achieve this goal. Second, although fixed-dose combinations of HAART are highly effective and increase adherence [14, 16], they may lead to increased toxicity when used improperly. Roll-out programs should ensure that FDCs are available in doses that are appropriate for a patient’s body weight, as well as allow for a run-in period of a lower dose of nevirapine. Third, most adverse effects can be ascertained through an appropriate clinical examination for specific symptoms and signs, including neuropsychiatric problems (due to efavirenzrelated toxicity), fatigue with conjunctival pallor (due to zidovudine-related anemia), and peripheral wasting (due to stavudine-related lipodystrophy). Implementation of protocols for regular clinical screening of patients, especially during the initial months of therapy, may help detect toxicities earlier. As suggested by a Kenyan study, early detection may also be facilitated by training family or community volunteers to identify toxicities [131]. In addition, facilities for laboratory monitoring of specific toxicities are a crucial component of scale-up of antiretroviral therapy. Current World Health Organization guidelines recommend liver enzyme and hemoglobin investigations only when patients are symptomatic [9]; however, because most cases of nevirapine-related hepatitis and zidovudine-induced anemia occur during the initial months of therapy, more intensive laboratory monitoring during this time may prevent severe toxicity.

Finally, by reducing antiretroviral drug options, toxicities may have a significant socioeconomic impact on low-income patients in developing countries. In studies from Haiti and South India, adverse effects were the primary reason for therapy modification [5, 132]. Currently, most government roll-out programs in resource-constrained regions provide few or no second-line drugs [133, 134]. Therefore, increased access to less-toxic first-line drugs and less-expensive second-line drugs is needed to cope with this issue. Specifically, patients will benefit if government programs ensure access to tenofovir or abacavir therapy for the small subset of patients who experience adverse reactions associated with both stavudine and zidovudine therapies, as well as with PIs for patients who cannot tolerate both nevirapine and efavirenz. Although few data are available on tenofovir, abacavir, and PI use in developing countries, these medications can be anticipated to have their own specific benefits and complications in resource-limited regions. A multisite trial in Africa found tenofovir therapy to be associated with a 1.3% rate of significant nephrotoxicity, which was comparable to other regimens [135]. Because abacavir therapy may cause a hypersensitivity rash, abacavir use may possibly complicate diagnosis of nevirapineinduced rash in patients initiating both medications. As previously noted, PI use may be problematic in patients receiving ATT, not only because of overlapping hepatotoxicity, but also because of drug-drug interactions with rifampicin [136]. Despite the problems associated with toxicities, the distribution of HAART in developing countries should not be discouraged, especially when these life-saving medications remain unavailable to the majority of patients in need [2]. However, excellent clinical follow-up is simultaneously required to manage the morbidity associated with HAART. Acknowledgments We thank the staff of the YRG Centre for AIDS Research and Education for their generous facilitation of this study. Financial support. Fogarty-Ellison Overseas Fellowship in Global Health and Clinical Research (3 D43 TW000237-13S1 to R.S.) and LifespanTufts-Brown Center for AIDS Research (5P30AI042853-09 to K.H.M.). Potential conflicts of interest. K.H.M. has a research grant from Gilead Sciences. All other authors: no conflicts.

References 1. Joint United Nations Programme on HIV/AIDS (UNAIDS), World Health Organization. AIDS epidemic update: December 2005. Geneva: UNAIDS, 2005. Available at: http://www.unaids.org/epi/2005/doc/ EPIupdate2005_pdf_en/epi-update2005_en.pdf. Accessed 24 February 2006. 2. Joint United Nations Programme on HIV/AIDS, World Health Organization (WHO). Progress on global access to HIV antiretroviral therapy: an update on 3 by 5 June 2005. Geneva: WHO, 2005. Available at: http://www.who.int/hiv/pub/progressreports/3by5%20Pro gress%20Report_E_light.pdf. Accessed 3 July 2006. 3. Kumarasamy N, Solomon S, Chaguturu SK, et al. The changing natural history of HIV disease: before and after the introduction of

HIV/AIDS • CID 2007:45 (15 October) • 1097

4.

5. 6.

7. 8.

9.

10.

11.

12.

13.

14.

15.

16.

17.

18.

19.

20.

21.

generic antiretroviral therapy in southern India. Clin Infect Dis 2005; 41:1525–8. Badri M, Wilson D, Wood R. Effect of highly active antiretroviral therapy on incidence of tuberculosis in South Africa: a cohort study. Lancet 2002; 359:2059–64. Severe P, Leger P, Charles M, et al. Antiretroviral therapy in a thousand patients with AIDS in Haiti. N Engl J Med 2005; 353:2325–34. Kumarasamy N, Lai A, Cecelia AJ, et al. Toxicities and adverse events following generic HAART in south Indian HIV-infected individuals [abstract P189]. In: Proceedings of the 7th International Congress on Drug Therapy and HIV Infection (Glasgow, United Kingdom). 2004. Mills E, Foster BC, van Heeswijk R, et al. Impact of African herbal medicines on antiretroviral metabolism. AIDS 2005; 19:95–7. Hetherington S, Cutrell A, Edwards M, Spreen W, Steel H. Risk factor analysis of hypersensitivity reactions to abacavir: retrospective analysis of 25 clinical trials [abstract 527]. In: Proceedings of the 1st International AIDS Society Conference on HIV Pathogenesis and Treatment (Buenos Aires, Argentina). 2001. World Health Organization (WHO). Scaling up antiretroviral therapy in resource-limited settings. Geneva: WHO, 2004. Available at: http: //www.who.int/hiv/pub/prev_care/en/arvrevision2003en.pdf. Accessed 3 July 2006. Management Sciences for Health, Rational Pharmaceutical Management Plus Program. Review of antiretroviral therapy guidelines in select countries of Africa and the Caribbean: a challenge for optimizing treatment and product supply. 2005. Available at: http:// www.who.int/3by5/amds/en/Standard_Treatment_Guidelines-Final .pdf. Accessed 3 July 2006. McComsey G, Lonergan JT. Mitochondrial dysfunction: patient monitoring and toxicity management. J Acquir Immune Defic Syndr 2004; 37(Suppl 1):S30–5. Moyle GJ, Sadler M. Peripheral neuropathy with nucleoside antiretrovirals: risk factors, incidence and management. Drug Safety 1998; 19:481–94. Scarsella A, Coodley G, Shalit P, et al. Stavudine-associated peripheral neuropathy in zidovudine-naive patients: effect of stavudine exposure and antiretroviral experience. Adv Ther 2002; 19:1–8. Laurent C, Kouanfack C, Koulla-Shiro S, et al. Effectiveness and safety of a generic fixed-dose combination of nevirapine, stavudine, and lamivudine in HIV-1–infected adults in Cameroon: open-label multicentre trial. Lancet 2004; 364:29–34. Anekthananon T, Ratanasuwan W, Techasathit W, Sonjai A, Suwanagool S. Safety and efficacy of a simplified fixed-dose combination of stavudine, lamivudine and nevirapine (GPO-VIR) for the treatment of advanced HIV-infected patients: a 24-week study. J Med Assoc Thai 2004; 87:760–7. Pujari SN, Patel AK, Naik E, et al. Effectiveness of generic fixed-dose combinations of highly active antiretroviral therapy for treatment of HIV infection in India. J Acquir Immune Defic Syndr 2004; 37: 1566–9. van Oosterhout JJ, Bodasing N, Kumwenda JJ, et al. Evaluation of antiretroviral therapy results in a resource-poor setting in Blantyre, Malawi. Trop Med Int Health 2005; 10:464–70. Saint-Marc T, Partisani M, Poizot-Martin I, et al. A syndrome of peripheral fat wasting (lipodystrophy) in patients receiving long-term nucleoside analogue therapy. AIDS 1999; 13:1659–67. Heath KV, Hogg RS, Chan KJ, et al. Lipodystrophy-associated morphological, cholesterol and triglyceride abnormalities in a populationbased HIV/AIDS treatment database. AIDS 2001; 15:231–9. Bernasconi E, Boubaker K, Junghans C, et al. Abnormalities of body fat distribution in HIV-infected persons treated with antiretroviral drugs: the Swiss HIV cohort study. J Acquir Immune Defic Syndr 2002; 31:50–5. Arpadi SM, Cuff PA, Horlick M, Wang J, Kotler DP. Lipodystrophy in HIV-infected children is associated with high viral load and low CD4+ -lymphocyte count and CD4+-lymphocyte percentage at base-

1098 • CID 2007:45 (15 October) • HIV/AIDS

22.

23.

24.

25.

26.

27.

28.

29.

30.

31.

32.

33.

34.

35.

36. 37.

38.

39.

line and use of protease inhibitors and stavudine. J Acquir Immune Defic Syndr 2001; 27:30–4. Lichtenstein KA, Delaney KM, Armon C, et al. Incidence of and risk factors for lipoatrophy (abnormal fat loss) in ambulatory HIV-1–infected patients. J Acquir Immune Defic Syndr 2003; 32:48–56. McComsey G, Maa JF. Host factors may be more important than choice of antiretrovirals in the development of lipoatrophy. AIDS Read 2003; 13:539, 542, 559. Lichtenstein KA, Ward DJ, Moorman AC, et al. Clinical assessment of HIV-associated lipodystrophy in an ambulatory population. AIDS 2001; 15:1389–98. Mallal SA, John M, Moore CB, James IR, McKinnon EJ. Contribution of nucleoside analogue reverse transcriptase inhibitors to subcutaneous fat wasting in patients with HIV infection. AIDS 2000; 14: 1309–16. Chang KH, Kim JM, Song YG, Hong SK, Lee HC, Lim SK. Does race protect an oriental population from developing lipodystrophy in HIVinfected individuals on HAART? J Infect 2002; 44:33–8. Paton NI, Earnest A, Ng YM, Karim F, Aboulhab J. Lipodystrophy in a cohort of human immunodeficiency virus-infected Asian patients: prevalence, associated factors, and psychological impact. Clin Infect Dis 2002; 35:1244–9. Ho TT, Chan KC, Wong KH, Lee SS. Indinavir-associated facial lipodystrophy in HIV-infected patients. AIDS Patient Care STDS 1999; 13:11–6. Fraser HE, Ishihara M, Miller JE, et al. Prevalence survey of lipodystrophy in HIV-positive patients in Japan [abstract 017]. In: Program and abstracts of the 2nd International Workshop on Adverse Drug Reactions and Lipodystrophy (Singapore). Singapore: International Medical Press, 2000. Puttawong S, Prasithsirikul W, Vadcharavivad S. Prevalence of lipodystrophy in Thai HIV-infected patients. J Med Assoc Thai 2004; 87: 605–11. Pujari SN, Dravid A, Naik E, et al. Lipodystrophy and dyslipidemia among patients taking first-line, World Health Organization– recommended highly active antiretroviral therapy regimens in western India. J Acquir Immune Defic Syndr 2005; 39:199–202. Tin EE, Bowonwatanuwong C, Desakorn V, Wilairatana P, Krudsood S, Pitisuttithum P. The efficacy and adverse effects of GPO-VIR (stavudine+lamivudine+nevirapine) in treatment-naive adult HIV patients. Southeast Asian J Trop Med Public Health 2005; 36:362–9. van Griensven J, De Naeyer L, Mushi T, Obarijoro S, Gazille C, Zachariah R. How common is lipodystrophy after at least 1 year of WHO first line antiretroviral treatment in Kigali, Rwanda [abstract WEPE0140]? In: Program and abstracts of the 16th International AIDS Conference (Toronto, Canada). Toronto, Canada: International AIDS Society, 2006. Fernandes AP, Sanches R, Mill J, Castelar L, Donadi E. Facing lipodystrophy [abstract TUPEO869]. In: Program and abstracts of the 16th International AIDS Conference (Toronto, Canada). Toronto, Canada: International AIDS Society, 2006. Saghayam S, Chaguturu SK, Kumarasamy N, Solomon S, Mayer KH, Wanke C. Lipoatrophy is the predominant presentation of HIV-associated lipodystrophy in southern India. Clin Infect Dis 2004; 38: 1646–7. Nolan D, Mallal S. The role of nucleoside reverse transcriptase inhibitors in the fat redistribution syndrome. J HIV Ther 2004; 9:34–40. van der Valk M, Casula M, Weverlingz GJ, et al. Prevalence of lipoatrophy and mitochondrial DNA content of blood and subcutaneous fat in HIV-1–infected patients randomly allocated to zidovudine- or stavudine-based therapy. Antivir Ther 2004; 9:385–93. Subbaraman R, Singh S, Cecelia AJ, et al. Resolution of anemia with use of highly active antiretroviral therapy (HAART) among HIVinfected patients in southern India [abstract THPE0113]. In: Program and abstracts of the 16th International AIDS Conference (Toronto, Canada). Toronto, Canada: International AIDS Society, 2006. Berhane K, Karim R, Cohen MH, et al. Impact of highly active an-

40.

41.

42.

43.

44.

45.

46.

47.

48.

49.

50.

51.

52.

53.

54.

tiretroviral therapy on anemia and relationship between anemia and survival in a large cohort of HIV-infected women: women’s interagency HIV study. J Acquir Immune Defic Syndr 2004; 37:1245–52. Moore RD, Forney D. Anemia in HIV-infected patients receiving highly active antiretroviral therapy. J Acquir Immune Defic Syndr 2002; 29:54–7. Moyle G, Sawyer W, Law M, Amin J, Hill A. Changes in hematologic parameters and efficacy of thymidine analogue-based, highly active antiretroviral therapy: a meta-analysis of six prospective, randomized, comparative studies. Clin Ther 2004; 26:92–7. McComsey GA, Paulsen DM, Lonergan JT, et al. Improvements in lipoatrophy, mitochondrial DNA levels and fat apoptosis after replacing stavudine with abacavir or zidovudine. AIDS 2005; 19:15–23. Carr A, Workman C, Smith DE, et al. Abacavir substitution for nucleoside analogs in patients with HIV lipoatrophy: a randomized trial. JAMA 2002; 288:207–15. Moyle GJ, Baldwin C, Langroudi B, Mandalia S, Gazzard BG. A 48week, randomized, open-label comparison of three abacavir-based substitution approaches in the management of dyslipidemia and peripheral lipoatrophy. J Acquir Immune Defic Syndr 2003; 33:22–8. Galli M, Ridolfo AL, Adorni F, et al. Body habitus changes and metabolic alterations in protease inhibitor-naive HIV-1–infected patients treated with two nucleoside reverse transcriptase inhibitors. J Acquir Immune Defic Syndr 2002; 29:21–31. Gallant JE, Staszewski S, Pozniak AL, et al. Efficacy and safety of tenofovir DF vs stavudine in combination therapy in antiretroviralnaive patients: a 3-year randomized trial. JAMA 2004; 292:191–201. Sobieszczyk ME, Hoover DR, Anastos K, et al. Prevalence and predictors of metabolic syndrome among HIV-positive and negative women [abstract WEPE0147]. In: Program and abstracts of the 16th International AIDS Conference (Toronto, Canada). Toronto, Canada: International AIDS Society, 2006. Hosseinipour M, Ingiliz P, Kanyama C, et al. Clinical features of lactic acidosis in Malawi [abstract CDB0665]. In: Program and abstracts of the 16th International AIDS Conference (Toronto, Canada). Toronto, Canada: International AIDS Society, 2006. Bolhaar M, Karstaedt A. A significant increase in incidence of lactic acidosis and symptomatic hyperlactaemia in women on HAART in Soweto [abstract WEPE0169]. In: Program and abstracts of the 16th International AIDS Conference (Toronto, Canada). Toronto, Canada: International AIDS Society, 2006. Kimani DK, File´n F, Nderitu M, Van Engelgem I, Suleh A, Zachariah R. Characteristics and outcomes of patients with symptomatic hyperlactatemia, on a first-line antiretroviral regimen of stavudine, lamivudine and nevirapine (d4T/3TC/NVP) in an urban district hospital setting in Kenya [abstract WEPE0150]. In: Program and abstracts of the 16th International AIDS Conference (Toronto, Canada). Toronto, Canada: International AIDS Society, 2006. Mwebaze PS, Castelnuovo B, Byakwaga H, et al. Nucleoside-associated lactic acidosis in HIV-1–infected African patients [abstract WEPE 0155]. In: Program and abstracts of the 16th International AIDS Conference (Toronto, Canada). Toronto, Canada: International AIDS Society, 2006. Luke C, Mfenyana G, Pawinski R, Moosa Y, Easterbrook PJ. High incidence of lactic acidosis (LA) at an antiretroviral (ARV) treatment site in Kwazulu-Natal (KZN), South Africa [abstract CDB0702]. In: Program and abstracts of the 16th International AIDS Conference (Toronto, Canada). Toronto, Canada: International AIDS Society, 2006. Sheng WH, Hsieh SM, Lee SC, et al. Fatal lactic acidosis associated with highly active antiretroviral therapy in patients with advanced human immunodeficiency virus infection in Taiwan. Int J STD AIDS 2004; 15:249–53. Lai HY, Chen JH, Tsai PP, Ho MW, Shen WC. Hepatic steatosis and pancreatitis associated with the use of stavudine in a patient with HIV infection. AJR Am J Roentgenol 2004; 183:1605–7.

55. Cho BC, Han SH, Choi SH, et al. A case of lactic acidosis caused by stavudine in an AIDS patient. Korean J Intern Med 2004; 19:66–9. 56. Marra A, Lewi D, Lanzoni V, et al. Lactic acidosis and antiretroviral therapy: a case report and literature review. Braz J Infect Dis 2000; 4:151–5. 57. Cornejo-Juarez P, Sierra-Madero J, Volkow-Fernandez P. Metabolic acidosis and hepatic steatosis in two HIV-infected patients on stavudine (d4T) treatment. Arch Med Res 2003; 34:64–9. 58. Patel AK, Patel K, Patel J. Lactic acidosis in HIV-1 infected patients receiving antiretroviral therapy. J Assoc Physicians India 2004; 52: 666–9. 59. Ivers LC, Mukherjee JS. Point of care testing for antiretroviral therapy–related lactic acidosis in resource-poor settings. AIDS 2006; 20: 779–80. 60. Mathee S, Abrahams M, Jackson N, et al. Point of care lactate testing is an effective measure in supporting the WHO first-line regimen in settings with a high incidence of stavudine-related toxicity [abstract MOPE0085]. In: Program and abstracts of the 16th International AIDS Conference (Toronto, Canada). Toronto, Canada: International AIDS Society, 2006. 61. Erhabor O, Ejele OA, Nwauche CA, Buseri FI. Some haematological parameters in human immunodeficiency virus (HIV) infected Africans: the Nigerian perspective. Niger J Med 2005; 14:33–8. 62. O’Brien ME, Kupka R, Msamanga GI, Saathoff E, Hunter DJ, Fawzi WW. Anemia is an independent predictor of mortality and immunologic progression of disease among women with HIV in Tanzania. J Acquir Immune Defic Syndr 2005; 40:219–25. 63. van Lettow M, West CE, van der Meer JW, Wieringa FT, Semba RD. Low plasma selenium concentrations, high plasma human immunodeficiency virus load and high interleukin-6 concentrations are risk factors associated with anemia in adults presenting with pulmonary tuberculosis in Zomba district, Malawi. Eur J Clin Nutr 2005; 59: 526–32. 64. Belperio PS, Rhew DC. Prevalence and outcomes of anemia in individuals with human immunodeficiency virus: a systematic review of the literature. Am J Med 2004; 116(Suppl 7A):27S–43S. 65. Sullivan PS, Hanson DL, Chu SY, Jones JL, Ward JW. Epidemiology of anemia in human immunodeficiency virus (HIV)-infected persons: results from the multistate adult and adolescent spectrum of HIV disease surveillance project. Blood 1998; 91:301–8. 66. Wills TS, Nadler JP, Somboonwit C, et al. Anemia prevalence and associated risk factors in a single-center ambulatory HIV clinical cohort. AIDS Read 2004; 14:305, 310, 313–5. 67. Richman DD, Fischl MA, Grieco MH, et al. The toxicity of azidothymidine (AZT) in the treatment of patients with AIDS and AIDSrelated complex: a double-blind, placebo-controlled trial. N Engl J Med 1987; 317:192–7. 68. Carr A, Cooper D. Adverse effects of antiretroviral therapy. Lancet 2000; 356:1423–30. 69. Idoko JA, Akinsete L, Abalaka AD, et al. A multicentre study to determine the efficacy and tolerability of a combination of nelfinavir (VIRACEPT), zalcitabine (HIVID) and zidovudine in the treatment of HIV infected Nigerian patients. West Afr J Med 2002; 21:83–6. 70. Moh R, Danel C, Sorho S, et al. Haematological changes in adults receiving a zidovudine-containing HAART regimen in combination with cotrimoxazole in Coˆte d’Ivoire. Antivir Ther 2005; 10:615–24. 71. Shah I. Adverse effects of antiretroviral therapy in HIV-1 infected children. J Trop Pediatr 2006; 52:244–8. 72. Pollard RB, Robinson P, Dransfield K. Safety profile of nevirapine, a nonnucleoside reverse transcriptase inhibitor for the treatment of human immunodeficiency virus infection. Clin Ther 1998; 20: 1071–92. 73. Warren KJ, Boxwell DE, Kim NY, Drolet BA. Nevirapine-associated Stevens-Johnson syndrome. Lancet 1998; 351:567. 74. Derisi M, Ballard C, Abulhosn K, Colwell B, Barber E, Matthews WC. Sulfa-associated rash and race are risk factors for non-nucleoside reverse transcriptase inhibitor-associated rash [abstract 61]. In: Ab-

HIV/AIDS • CID 2007:45 (15 October) • 1099

75.

76.

77.

78.

79. 80.

81.

82.

83.

84.

85.

86.

87.

88.

89.

90.

91. 92.

stracts of the 7th Conference on Retroviruses and Opportunistic Infections (San Francisco, CA). San Francisco: Foundation for Retrovirology and Human Health, 2000. Gangar M, Arias G, O’Brien JG, Kemper CA. Frequency of cutaneous reactions on rechallenge with nevirapine and delavirdine. Ann Pharmacother 2000; 34:839–42. Ananworanich J, Moor Z, Siangphoe U, et al. Incidence and risk factors for rash in Thai patients randomized to regimens with nevirapine, efavirenz or both drugs. AIDS 2005; 19:185–92. Antinori A, Baldini F, Girardi E, et al. Female sex and the use of antiallergic agents increase the risk of developing cutaneous rash associated with nevirapine therapy. AIDS 2001; 15:1579–81. Mazhude C, Jones S, Murad S, Taylor C, Easterbrook P. Female sex but not ethnicity is a strong predictor of non-nucleoside reverse transcriptase inhibitor-induced rash. AIDS 2002; 16:1566–8. Bersoff-Matcha SJ, Miller WC, Aberg JA, et al. Sex differences in nevirapine rash. Clin Infect Dis 2001; 32:124–9. Soriano V, Dona C, Barreiro P, Gonzalez-Lahoz J. Is there crosstoxicity between nevirapine and efavirenz in subjects developing rash? AIDS 2000; 14:1672–3. Martinez E, Blanco JL, Arnaiz JA, et al. Hepatotoxicity in HIV-1–infected patients receiving nevirapine-containing antiretroviral therapy. AIDS 2001; 15:1261–8. Stern JO, Robinson PA, Love J, Lanes S, Imperiale MS, Mayers DL. A comprehensive hepatic safety analysis of nevirapine in different populations of HIV infected patients. J Acquir Immune Defic Syndr 2003; 34(Suppl 1):S21–33. Wit FW, Weverling GJ, Weel J, Jurriaans S, Lange JM. Incidence of and risk factors for severe hepatotoxicity associated with antiretroviral combination therapy. J Infect Dis 2002; 186:23–31. Cantrell R, Chi B, Mulenga L, et al. Incidence and predictors of hepatotoxicity among patients receiving nevirapine (NVP)– containing antiretroviral therapy (ART) in Zambia [abstract WEPE0172]. In: Program and abstracts of the 16th International AIDS Conference (Toronto, Canada). Toronto, Canada: International AIDS Society, 2006. Sanne I, Mommeja-Marin H, Hinkle J, et al. Severe hepatotoxicity associated with nevirapine use in HIV-infected subjects. J Infect Dis 2005; 191:825–9. Leith J, Piliero P, Storfer S, Mayers D, Hinzmann R. Appropriate use of nevirapine for long-term therapy. J Infect Dis 2005; 192:545–6; author reply, 546. Viramune [package insert]. Ridgefield, CT: Boehringer Ingelheim, 2005. Available at: http://www.bidocs.com/renetnt:/Prescribing+In formation/PIs/Viramune/Viramune.pdf. Accessed 3 July 2006. Jamisse L, Balkus J, Hitti J, et al. Nevirapine-associated hepatic toxicity among HIV-1–seropositive pregnant women in Mozambique [abstract WEPE0173]. In: Program and abstracts of the 16th International AIDS Conference (Toronto, Canada). Toronto, Canada: International AIDS Society, 2006. Storfer S, Leith J, Piliero P, Hall D. Analysis of hepatic events within the 2NN study: Controlling for ethnicity and CD4+ count at initiation of nevirapine therapy [abstract PE9.6/2]. In: Program and abstracts of the 10th European AIDS Conference (Dublin, Ireland). 2005. Phanuphak N, Apornpong T, Intarasuk S, Teeratakulpisarn S, Phanuphak P. Toxicities from nevirapine in HIV-infected males and females, including pregnant females with various CD4 cell counts [abstract 21]. In: Program and abstracts of the 12th Conference on Retroviruses and Opportunistic Infections (Boston, MA). Alexandria, VA: Foundation for Retrovirology and Human Health, 2005. Sanne I, Rousseau F, Bartlett JA, Lederman MM. Reply to Leith et al. J Infect Dis 2005; 192:546–7. de Maat MM, Huitema AD, Mulder JW, Meenhorst PL, van Gorp EC, Beijnen JH. Population pharmacokinetics of nevirapine in an unselected cohort of HIV-1–infected individuals. Br J Clin Pharmacol 2002; 54:378–85.

1100 • CID 2007:45 (15 October) • HIV/AIDS

93. Haas DW, Bartlett JA, Andersen JW, et al. Pharmacogenetics of nevirapine-associated hepatotoxicity: an adult AIDS clinical trials group collaboration. Clin Infect Dis 2006; 43:783–6. 94. Law WP, Dore GJ, Duncombe CJ, et al. Risk of severe hepatotoxicity associated with antiretroviral therapy in the HIV-NAT cohort, Thailand, 1996–2001. AIDS 2003; 17:2191–9. 95. Sulkowski MS, Thomas DL, Mehta SH, Chaisson RE, Moore RD. Hepatotoxicity associated with nevirapine or efavirenz-containing antiretroviral therapy: role of hepatitis C and B infections. Hepatology 2002; 35:182–9. 96. Pineda JA, Macias J. Progression of liver fibrosis in patients coinfected with hepatitis C virus and human immunodeficiency virus undergoing antiretroviral therapy. J Antimicrob Chemother 2005; 55:417–9. 97. Molina JM, Ferchal F, Rancinan C, et al. Once-daily combination therapy with emtricitabine, didanosine, and efavirenz in human immunodeficiency virus–infected patients. J Infect Dis 2000; 182: 599–602. 98. Barrett JS, Joshi AS, Chai M, Ludden TM, Fiske WD, Pieniaszek HJ Jr. Population pharmacokinetic meta-analysis with efavirenz. Int J Clin Pharmacol Ther 2002; 40:507–19. 99. Pfister M, Labbe L, Hammer SM, et al. Population pharmacokinetics and pharmacodynamics of efavirenz, nelfinavir, and indinavir: adult AIDS clinical trial group study 398. Antimicrob Agents Chemother 2003; 47:130–7. 100. Haas DW, Ribaudo HJ, Kim RB, et al. Pharmacogenetics of efavirenz and central nervous system side effects: an adult AIDS clinical trials group study. AIDS 2004; 18:2391–400. 101. Danel C, Moh R, Messou E, et al. Short-term tolerance of efavirenz in HIV-infected african adults participating in the TRIVACAN ANRS 1269 trial, Abidjan, Coˆte d’Ivoire [abstract 53]. In: Program and abstracts of the 2nd International AIDS Conference on HIV Pathogenesis and Treatment (Paris, France). 2003. 102. Boyle BA. Issues in antiretroviral toxicity. AIDS Read 2003; 13:459, 463–4, 468, 469, 479. 103. Dean GL, Edwards SG, Ives NJ, et al. Treatment of tuberculosis in HIV-infected persons in the era of highly active antiretroviral therapy. AIDS 2002; 16:75–83. 104. Swaminathan S, Luetkemeyer A, Srikantiah P, Lin R, Charlebois E, Havlir DV. Antiretroviral therapy and TB. Trop Doct 2006; 36:73–9. 105. Manosuthi W, Chumpathat N, Chaovavanich A, Sungkanuparph S. Safety and tolerability of nevirapine-based antiretroviral therapy in HIV-infected patients receiving fluconazole for cryptococcal prophylaxis: a case-control study. BMC Infect Dis 2005; 5:67. 106. Breen RA, Lipman MC, Johnson MA. Increased incidence of peripheral neuropathy with co-administration of stavudine and isoniazid in HIV-infected individuals. AIDS 2000; 14:615. 107. Shelburne SA 3rd, Hamill RJ, Rodriguez-Barradas MC, et al. Immune reconstitution inflammatory syndrome: emergence of a unique syndrome during highly active antiretroviral therapy. Medicine (Baltimore) 2002; 81:213–27. 108. Buckingham SJ, Haddow LJ, Shaw PJ, Miller RF. Immune reconstitution inflammatory syndrome in HIV-infected patients with mycobacterial infections starting highly active anti-retroviral therapy. Clin Radiol 2004; 59:505–13. 109. French MA, Price P, Stone SF. Immune restoration disease after antiretroviral therapy. AIDS 2004; 18:1615–27. 110. Hirsch HH, Kaufmann G, Sendi P, Battegay M. Immune reconstitution in HIV-infected patients. Clin Infect Dis 2004; 38:1159–66. 111. Sungkanuparph S, Vibhagool A, Mootsikapun P, Chetchotisakd P, Tansuphaswaswadikul S, Bowonwatanuwong C. Opportunistic infections after the initiation of highly active antiretroviral therapy in advanced AIDS patients in an area with a high prevalence of tuberculosis. AIDS 2003; 17:2129–31. 112. Shelburne SA, Visnegarwala F, Darcourt J, et al. Incidence and risk factors for immune reconstitution inflammatory syndrome during highly active antiretroviral therapy. AIDS 2005; 19:399–406.

113. Cheng VC, Yuen KY, Chan WM, Wong SS, Ma ES, Chan RM. Immunorestitution disease involving the innate and adaptive response. Clin Infect Dis 2000; 30:882–92. 114. French MA, Lenzo N, John M, et al. Immune restoration disease after the treatment of immunodeficient HIV-infected patients with highly active antiretroviral therapy. HIV Med 2000; 1:107–15. 115. Lortholary O, Fontanet A, Memain N, et al. Incidence and risk factors of immune reconstitution inflammatory syndrome complicating HIVassociated cryptococcosis in France. AIDS 2005; 19:1043–9. 116. Michailidis C, Pozniak AL, Mandalia S, Basnayake S, Nelson MR, Gazzard BG. Clinical characteristics of IRIS syndrome in patients with HIV and tuberculosis. Antivir Ther 2005; 10:417–22. 117. Tassie JM, Szumilin E, Calmy A, Goemaere E; Medecins Sans Frontieres. Highly active antiretroviral therapy in resource-poor settings: the experience of Medecins Sans Frontieres. AIDS 2003; 17:1995–7. 118. Kumarasamy N, Chaguturu S, Mayer KH, et al. Incidence of immune reconstitution syndrome in HIV/tuberculosis-coinfected patients after initiation of generic antiretroviral therapy in India. J Acquir Immune Defic Syndr 2004; 37:1574–6. 119. Manosuthi W, Kiertiburanakul S, Phoorisri T, Sungkanuparph S. Immune reconstitution inflammatory syndrome of tuberculosis among HIV-infected patients receiving antituberculous and antiretroviral therapy. J Infect 2006; 53:357–63. 120. Vallabhaneni S, Kumarasamy N, Wing EJ, Flanigan TP. A diagnostic dilemma in a patient receiving antiretroviral and antituberculosis therapy. AIDS Read 2005; 15:72–5. 121. Swaminathan S, Padmapriyadarsini C, Narendran G, Thomas BE, Anand S, Rajyalakshmi A. Immune reconstitution syndrome following initiation of antiretroviral therapy in a patient with HIV infection and multidrug-resistant tuberculosis. Indian J Chest Dis Allied Sci 2005; 47:299–304. 122. Couppie P, Abel S, Voinchet H, et al. Immune reconstitution inflammatory syndrome associated with HIV and leprosy. Arch Dermatol 2004; 140:997–1000. 123. Lawn SD, Wood C, Lockwood DN. Borderline tuberculoid leprosy: an immune reconstitution phenomenon in a human immunodeficiency virus–infected person. Clin Infect Dis 2003; 36:e5–6. 124. Ridolfo AL, Gervasoni C, Antinori S, et al. Post-kala-azar dermal leishmaniasis during highly active antiretroviral therapy in an AIDS patient infected with leishmania infantum. J Infect 2000; 40:199–202. 125. Blanche P, Gombert B, Rivoal O, Abad S, Salmon D, Brezin A. Uveitis due to Leishmania major as part of HAART-induced immune restitution syndrome in a patient with AIDS. Clin Infect Dis 2002; 34: 1279–80.

126. Sharp MJ, Mallon DF. Regional bacillus Calmette-Gue´rin lymphadenitis after initiating antiretroviral therapy in an infant with human immunodeficiency virus type 1 infection. Pediatr Infect Dis J 1998; 17:660–2. 127. Moore RD, Wong WM, Keruly JC, McArthur JC. Incidence of neuropathy in HIV-infected patients on monotherapy versus those on combination therapy with didanosine, stavudine and hydroxyurea. AIDS 2000; 14:273–8. 128. Lichtenstein KA, Armon C, Baron A, et al. Modification of the incidence of drug-associated symmetrical peripheral neuropathy by host and disease factors in the HIV outpatient study cohort. Clin Infect Dis 2005; 40:148–57. 129. John M, Mallal S. Hyperlactatemia syndromes in people with HIV infection. Curr Opin Infect Dis 2002; 15:23–9. 130. Moore RD, Keruly JC, Chaisson RE. Incidence of pancreatitis in HIVinfected patients receiving nucleoside reverse transcriptase inhibitor drugs. AIDS 2001; 15:617–20. 131. Ilako F, Kimura M, Wanjiku L, Marston B. Assessment of common adverse drug reactions and complications of ART using community volunteers in Kibera, Nairobi, Kenya [abstract WEKC203]. In: Program and abstracts of the 16th International AIDS Conference (Toronto, Canada). Toronto, Canada: International AIDS Society, 2006. 132. Kumarasamy N, Vallabhaneni S, Cecelia AJ, et al. Reasons for modification of generic highly active antiretroviral therapeutic regimens among patients in southern India. J Acquir Immune Defic Syndr 2006; 41:53–8. 133. National AIDS Control Organization of India (NACO). National guidelines for implementation of antiretroviral therapy. New Delhi: NACO, 2004. Available at: http://www.nacoonline.org/guidelines/ ART_Guidelines.pdf. Accessed 3 July 2006. 134. National Department of Health of South Africa. National antiretroviral treatment guidelines. South Africa: Minuteman Press, 2004. Available at: http://www.doh.gov.za/docs/factsheets/guidelines/art guide04-f.html. Accessed 3 July 2006. 135. Reid A, Sto¨hr W, Walker S, Ssali F, Munderi P, Gilks C. Glomerular dysfunction and associated risk factors following initiation of ART in adults with HIV infection in Africa [abstract THAB0105]. In: Program and abstracts of the 16th International AIDS Conference (Toronto, Canada). Toronto, Canada: International AIDS Society, 2006. 136. Burman WJ, Jones BE. Treatment of HIV-related tuberculosis in the era of effective antiretroviral therapy. Am J Respir Crit Care Med 2001; 164:7–12.

HIV/AIDS • CID 2007:45 (15 October) • 1101