ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Mitochondrial toxicity of HIV nucleoside reverse transcriptase inhibitors

Mitochondrial toxicity of HIV nucleoside reverse transcriptase inhibitors
Literature review current through: Jan 2024.
This topic last updated: Feb 16, 2022.

INTRODUCTION — Since the introduction of potent antiretroviral therapy (ART), the life expectancy of HIV-infected patients has improved enormously. However, the chronic administration of antiretroviral medications has led to the recognition of long-term complications of these therapies [1,2]. Mitochondrial toxicity is recognized as a major adverse effect of nucleoside analogue treatment and can lead to myopathy, peripheral neuropathy, and hepatic steatosis with lactic acidosis, which can be life-threatening.

The induction of mitochondrial dysfunction by nucleoside reverse transcriptase inhibitors (NRTIs) will be discussed here. The use of NRTIs for the treatment of HIV infection is discussed separately. (See "Selecting antiretroviral regimens for treatment-naïve persons with HIV-1: General approach".)

ROLE OF MITOCHONDRIA — The main function of mitochondria is oxidative phosphorylation in which fatty acids and pyruvate are used as substrates to produce energy in the form of adenosine triphosphate (ATP). Normally there is tight coupling between oxidation and phosphorylation (OXPHOS) that is governed by normal mitochondrial enzymes, such as the cytochrome oxidases and other enzymes of the respiratory chain system. OXPHOS is also responsible for neutralization of free radicals and beta-oxidation of free fatty acids.

Mitochondria also play an important role in the generation of reactive oxygen species (ROS), which are produced physiologically during oxidative phosphorylation. Oxidative stress can occur if there is an imbalance between the production of ROS and cellular antioxidant defenses [3]. Mitochondrial DNA polymerase is a target of oxidative damage [4].

PATHOGENESIS — Nucleoside analogues are effective in inhibiting HIV replication due to their high affinity for the viral enzyme reverse transcriptase (a viral DNA polymerase) [5-7]. However, NRTIs can also bind to other human DNA-polymerases, like DNA polymerase beta (necessary for repair of nuclear DNA) and mitochondrial DNA polymerase gamma, which is exclusively responsible for the replication of mitochondrial DNA (mtDNA).

Mitochondrial DNA polymerase gamma — By inhibiting mtDNA enzyme polymerase-gamma, NRTIs can lead to depletion of mtDNA, resulting in organelle dysfunction and impairment of oxidative phosphorylation (figure 1) [2]. mtDNA encodes 13 mitochondrial polypeptides involved in the respiratory chain that are essential for oxidative phosphorylation (OXPHOS) [8]. A disruption of OXPHOS leads to energy loss (decreased ATP) and an increase in electron leakage from the electron-transport chain, which increases the production of reactive oxygen species (ROS) [9]. ROS, in turn, damage proteins, lipids, and mtDNA, leading to a cascade of further oxidative damage and lipid peroxidation. mtDNA is vulnerable to mutations since it has no introns or histones.

Although the so-called "pol-gamma hypothesis" still remains the most important explanation for the induction of mitochondrial dysfunction by NRTIs, nucleoside analogues can also influence intracellular nucleoside transporters, disturb the delicate balance of the intracellular nucleoside pools, or alter kinetics of phosphorylation of natural nucleosides [10-13]. Furthermore, the acceleration of mtDNA turnover as a compensatory mechanism for mtDNA depletion can lead to persistent accumulation of somatic mutations in mtDNA even after NRTI withdrawal [14]. The impact of all these effects on long-term mitochondrial function is still unclear.

Expression of mitochondrial toxicity may not occur until a specific biologic threshold of mitochondrial dysfunction has been reached. Onset of mitochondrial toxicity may vary depending on the individual cell type since copy numbers of mtDNA vary, as well as cellular dependence on energy production [15]. Mitochondria are found in all cells of the body except for erythrocytes.

Production of lactate — Early effects of mitochondrial toxicity include decreased energy production and increased production of lactate. During glycolysis, glucose is metabolized to pyruvate and energy is produced in the form of ATP. Under normal conditions, pyruvate is taken up by mitochondria and metabolized to acetyl-CoA, which feeds into the Krebs cycle. This pathway is responsible for most of the cell's energy production. In the presence of mitochondrial dysfunction, the metabolism of pyruvate is shifted to lactate, with a decrease in energy production.

A decrease in OXPHOS will impair ATP production, beta oxidation of fatty acids, and neutralization of free radicals leading to oxidative stress. Furthermore, since acetyl-co-A can no longer enter the Krebs cycle, it will alternatively be metabolized into ketone bodies, while lactate and triglycerides will accumulate (figure 2). Since every cell type has a different dependence on OXPHOS, cellular dysfunction will only emerge after the decrease in OXPHOS has dropped below a certain cell-dependent threshold [16,17].

IMPLICATED DRUGS — Eight nucleoside reverse transcriptase inhibitors (NRTIs) are available for treatment of HIV. The most commonly used ones are lamivudine (3TC), emtricitabine (FTC), abacavir (ABC), tenofovir disoproxil fumarate (TDF), and tenofovir alafenamide (TAF). Zidovudine (ZDV), didanosine (ddI), and stavudine (d4T) are less frequently used, and zalcitabine (ddC) was removed from the market in 2006. A discussion on selecting an antiretroviral regimen is presented separately. (See "Selecting an antiretroviral regimen for treatment-experienced patients with HIV who are failing therapy".)

All NRTIs are chain terminators and result in the reversible termination of the viral DNA chain and also (if incorporated) of the mtDNA chain. This point is important since discontinuation of medications may lead to reversibility of mitochondrial toxicity if recognized early.

The NRTIs all have different affinities for mtDNA polymerase gamma, explaining in part the different propensity of the drugs to induce toxicity [18] (see 'Pathogenesis' above). In vitro, the order of strength of inhibiting polymerase gamma for the different NRTIs is: ddC >> ddI > d4T ≥ ZDV >>> TDF = 3TC = FTC = ABC [19-21]. There are insufficient data to speculate about TAF. Other factors that may explain the varying effects of individual drugs on DNA polymerase gamma include differences in cell entry, and transport to the mitochondrial membrane [9].

Some NRTIs (ddC, ddI, and d4T) are particularly associated with a decline in mtDNA content and ultrastructural changes, including swollen or enlarged mitochondria and loss and distortion of their cristae. Due to the significant risk of mitochondrial toxicity associated with ddI and D4T, these drugs are generally avoided for the treatment of HIV, and distribution by the original producer was stopped in September 2017. It is unclear how long generic d4T and ddI will be available thereafter.

Mitochondrial toxicity secondary to drug exposure had been described prior to the development of antiretroviral medications. These drugs included valproic acid, tetracycline, amiodarone, and others. Fialuridine (FIAU) was a nucleoside analogue in clinical development for the treatment of hepatitis B, but the clinical trial was prematurely halted when patients developed evidence of liver failure and hepatic steatosis that was secondary to mitochondrial toxicity [22]. FIAU was efficiently incorporated into internal positions of mtDNA in place of thymidine and led to ultrastructural changes in mitochondria and increased intracellular lipid droplets [23]. Histological findings of liver tissue demonstrated severe accumulation of microvesicular and macrovesicular fat, with minimal necrosis of hepatocytes [22]. Electron microscopy showed abnormal mitochondria and the accumulation of fat in hepatocytes. There was no known repair mechanism to remove the internal FIAU and the damage to mtDNA was permanent [24].

CLINICAL PRESENTATION — The clinical presentation of mitochondrial toxicity depends on the target organ that is involved (table 1). There is little doubt that mitochondrial toxicity is the major cause of NRTI-induced myopathy, neuropathy, lipoatrophy, and lactic acidosis [6,7,25]. Whether mitochondrial toxicity is involved in other clinical manifestations, such as pancreatitis, myelosuppression, cardiomyopathy, tubular dysfunction, or osteopenia, is not as clear [26].

Myopathy — The first reports of a toxic mitochondrial myopathy in HIV-infected patients were related to the use of ZDV [27]. Symptoms of myopathy included proximal muscle weakness, tenderness, and myalgia. The majority of patients have elevated serum creatine phosphokinase levels and may occasionally have elevated lactate levels [28,29]. Occasionally a patient may have electromyographic evidence of proximal muscle myopathy despite normal muscle enzymes [30].

ZDV affects the muscle mitochondria by inhibiting DNA polymerase gamma, resulting in termination of the DNA chain and depletion of muscle mtDNA [27,31]. Histological features include an inflammatory, destructive mitochondrial myopathy with "ragged-red" fibers seen on trichrome stain, indicative of abnormal mitochondria with paracrystalline inclusions [27]. Patients with ubiquitous "ragged-red" fibers also have significant lipid and glycogen accumulation in proximity to abnormal mitochondria, as seen in patients with inherited mitochondrial myopathies [32]. When mitochondria are structurally and functionally abnormal, long-chain fatty acids cannot be effectively utilized, resulting in lipid accumulation within the muscle [32]. Carnitine, which is essential for the transport of long-chain fatty acids into the muscle mitochondria, is depleted in ZDV-treated patients who have significant numbers of "ragged-red" fibers on muscle biopsy [32]. Two patients who had repeat biopsies after discontinuation of ZDV showed marked histologic improvement.

Lipoatrophy — Clinical alterations in adipose tissue and metabolic changes, such as insulin resistance and hyperlipidemia, are observed in patients taking antiretroviral therapy. This constellation of findings is referred to as the "lipodystrophy syndrome". Lipoatrophy, or fat atrophy, is associated with nucleoside reverse transcriptase inhibitor use, particularly d4T [33-36]. As an example, in a large prospective, randomized trial conducted over 96 weeks, a 50 percent dose reduction in d4T induced significantly more lipodystrophy compared with TDF (5.6 versus 0.2 percent) [36].

Several groups have demonstrated profound mtDNA depletion and morphologic alterations in subcutaneous fat biopsies of these patients [37-40]. Some have speculated that lipoatrophy is secondary to adipocyte apoptosis rather than necrosis, because the loss of fat is neither painful nor inflammatory. Furthermore, mitochondria are closely involved in apoptotic pathways, again implying a role for NRTI-induced toxicity [15,41]. Others have suggested that HIV itself may cause mitochondrial dysfunction, either directly or indirectly through the induction of inflammatory cytokines [40].

The epidemiology, clinical features, and treatment of lipoatrophy are discussed elsewhere. (See "Epidemiology, clinical manifestations, and diagnosis of HIV-associated lipodystrophy" and "Treatment of HIV-associated lipodystrophy".)

Hepatic steatosis — Hepatic steatosis and hepatic failure together with lactic acidosis have been reported as rare but serious adverse effects of nucleoside analogue therapy [30,42]. In 1993, eight cases of severe hepatomegaly with diffuse steatosis were reported in HIV-infected patients without AIDS, six of which were fatal [43]. All had received zidovudine for a minimum of six months prior to the onset of gastrointestinal symptoms and mild elevations of transaminases. Case reports of liver failure and hepatic steatosis have since been reported, especially in association with ddI and d4T [44].

Microvesicular hepatic steatosis has been identified in most of these cases, which is related to mitochondrial dysfunction [45]. Decreased mitochondrial oxidation of fatty acids leads to increased esterification of triglycerides and a decreased egress of triglycerides from the liver, which accumulate as small lipid droplets. Prior to the era of potent ART, it was well recognized that other drugs that inhibit mitochondrial oxidation, including valproate, tetracycline, amiodarone, ethanol, and salicylic acid, can lead to these histologic findings [46].

Hyperlactatemia and lactic acidosis — Hyperlactatemia and lactic acidosis are associated with NRTI use and typically occur in the absence of systemic hypoperfusion (called type B lactic acidosis) [47]. (See "Causes of lactic acidosis", section on 'Type B lactic acidosis'.)

Lactic acidosis represents a serious metabolic manifestation of mitochondrial toxicity that can lead to death [47]. Hepatic dysfunction may be an essential prerequisite for the development of lactate accumulation, since the liver is the most important organ for clearance of lactate [48].

Symptoms of hyperlactatemia or lactic acidosis may be nonspecific and include nausea, vomiting, abdominal pain, weight loss, and severe fatigue (table 2). Serum aminotransferases are only mildly elevated in most cases [43].

Lactic acidosis usually follows a minimum of six months of treatment, but may occur precipitously. Due to the nonspecificity of symptoms, hyperlactatemia may not be recognized for weeks to months [49]. Without intervention, lactic acidosis leads to a fatal outcome, most often due to liver failure and cardiac arrhythmias. NRTIs should be stopped promptly, and supportive medication should be initiated. The use of NRTI-sparing regimens is discussed elsewhere. (See "Switching antiretroviral therapy for adults with HIV-1 and a suppressed viral load".)

In the pre-ART era, lactic acidosis was described in patients on monotherapy with high-dose ZDV (1200 mg/day) and ddI with an estimated incidence of 1.2 per 1000 treated patient-years [42]. In the early era of combination therapy, d4T and ddI were most closely associated with this syndrome, sometimes in combination with each other [30,49-52].

In a prospective study in South Africa, 1771 HIV-infected participants were randomly assigned to a nucleoside backbone of d4T/3TC or ddI/AZT plus either efavirenz or lopinavir/ritonavir. Lactic acidosis was defined as a serum lactate concentration >5 mmol/L with an arterial pH <7.35 or a serum bicarbonate concentration <20 mmol/L. Symptomatic hyperlactatemia was defined as serum lactate >2.2 mmol/L with symptoms consistent with increased lactate (eg, nausea, anorexia, fatigue). A total of 13 cases with lactic acidosis (0.7 percent) and 28 cases with symptomatic hyperlactatemia (1.6 percent) were observed and two patients died. The risk of lactic acidosis or symptomatic hyperlactatemia was higher in female patients and among those taking d4T/3TC [53]. These observations were confirmed in similar studies from Botswana, Malawi, and Cambodia [54-56].

Asymptomatic hyperlactatemia (lactate <5 mmol/L) has been described in much higher prevalence (10 to 40 percent) and has also been observed in association with the use of d4T ± ddI [47,57-60]. Hyperlactatemia does not appear to be a constant phenomenon despite continuation of medication; some of these paradoxical observations may be related to incorrect blood sampling.

The natural history of hyperlactatemia is unclear since cases of lactic acidosis have been described where normal lactate levels were documented just prior to onset. In order to interpret the importance of an elevated lactate, decision algorithms have been developed (algorithm 1) [61].

Pancreatitis — The dideoxynucleoside analogues are most closely associated with pancreatitis (ddI>d4T). It has not been clearly established that pancreatitis is related to mitochondrial toxicity. However, concomitant chemical pancreatitis has been described in patients who had developed symptomatic lactic acidosis on NRTI therapy, suggesting a common mechanism [30,51].

Evidence that mitochondrial toxicity may be implicated in pancreatitis also comes from the clinical trial of FIAU for the treatment of hepatitis B. Seven of 15 patients who developed liver failure and hepatic steatosis secondary to mitochondrial toxicity also had biochemical evidence of pancreatitis [22]. All five patients who died had both gross and microscopic evidence of pancreatitis at autopsy.

Bone marrow suppression — The use of ZDV is associated with bone marrow suppression, which may be related to mitochondrial toxicity [34].

Neuropathy — The use of didanosine and stavudine has been linked to the development of peripheral neuropathy.

Nephrotoxicity — Tenofovir, in particular, tenofovir disoproxil fumarate (TDF), can induce proximal tubular dysfunction with subsequent renal failure. (See "Overview of antiretroviral agents used to treat HIV", section on 'Tenofovir' and "Overview of kidney disease in patients with HIV", section on 'Medication nephrotoxicity'.)

Accumulation of tenofovir in proximal tubular cells has been demonstrated in experimental models, with a pivotal role for membrane transporters as OAT1 and MRP4 [62]. Although in one animal study it was suggested that mtDNA content was altered (increased) in isolated proximal tubular cells, mitochondrial dysfunction was not shown. Since tenofovir lacks affinity for DNA polymerase-gamma, it seems very unlikely that this drug can affect mtDNA levels via this pathway. However, in one study, more somatically mutated mtDNA could be demonstrated in association with TDF exposure, suggesting that TDF-associated renal damage was at least in part mitochondrially mediated [63].

Osteopenia/osteoporosis — Although the occurrence of decreased bone mineral density during chronic HIV infection has been associated with the use of antiretroviral treatment, in particular with the use of tenofovir disoproxil fumarate, it has not yet been demonstrated that mitochondrial dysfunction plays any role in this feature.

PATHOLOGY — Electron microscopy demonstrates markedly swollen mitochondria with loss of cristae, matrix dissolution, and scattered vesicular inclusions, consistent with the theory that the mitochondrion is the main target of injury. Other specific findings in various tissues are discussed below.

RISK FACTORS — Not all individuals will develop mitochondrial toxicity, even after several years of exposure to the most toxic drugs (eg, ddC or d4T/ddI combinations). Several studies have tried to identify specific mitochondrial DNA (mtDNA) haplotypes that would make someone more sensitive to the effects of NRTI treatment; however, these reports have been unrevealing due to limitations such as heterogeneous methods and outcomes, limited racial/ethnic groups, and inadequate statistical power [64].

However, based upon observations that have been reported in patients with HIV and inherited mitochondrial diseases, factors that might be important include:

Female sex [49,52,54,56,65,66]

Concomitant use of ribavirin with ddI for the treatment of hepatitis C [67-69]

Increased age [52]

The risk in females may be even higher during pregnancy [70-72]. (See "Antiretroviral selection and management in pregnant individuals with HIV in resource-rich settings".)

DIAGNOSIS — The gold standard for the diagnosis of nucleoside-related mitochondrial toxic effects is a muscle or liver biopsy.

When a patient presents with symptoms consistent with hyperlactatemia or lactic acidosis, a blood sample for lactate should be sent in a fluoride oxalate tube on ice and delivered within four hours to the laboratory for processing. Levels need to be assessed with the strictest of quality control measures since elevated lactate can be an artifact of collection techniques [73]. Therefore, it is critically important to avoid certain factors that can artificially elevate lactate levels. Ideally samples should be drawn without use, or prolonged use, of a tourniquet. Whenever possible, it is also best to monitor levels in patients who have not exercised shortly before sampling.

Patients with pancreatitis (either chemical or clinical) should also have measurements of serum lactate, since these clinical entities have occurred concomitantly. (See 'Pancreatitis' above.)

PATIENT MONITORING — Routine monitoring for lactic acidosis is not warranted since prospective monitoring is not predictive of risk [55,74-77]. Although hyperlactatemia can be a correlate of mitochondrial dysfunction, chronic hyperlactatemia found on routine testing in asymptomatic patients has a poor predictive value [75,77]. As an example, in a prospective study of 253 patients in Malawi using stavudine, hyperlactatemia (>2.2 mg/dl measured with a point-of-care analyzer) was identified on at least one occasion in 210 patients (83 percent), but was only sustained in 65 (26 percent) over the course of one year [55]. Only one patient fulfilled a diagnosis of lactic acidosis.

The role of monitoring mitochondrial DNA (mtDNA) on venous blood has also been explored [78,79]. Although early reports were promising [78], subsequent studies could not confirm the value of monitoring mtDNA measurements in PBMCs and other tissues [40,80-84]. Furthermore, significant variance has been reported among laboratories assessing the same clinical sample [85].

SPECIAL SITUATIONS: IN UTERO EXPOSURE TO NRTIS — Antiretroviral drugs play an essential role in the prevention of mother-to-child-transmission (MTCT) of HIV. In utero exposure to NRTIs has been demonstrated to induce transient anemia, leucopenia, and hyperlactatemia in the newborns, without further sequelae [86-88]. This topic is discussed in detail elsewhere. (See "Safety and dosing of antiretroviral medications in pregnancy", section on 'Mitochondrial toxicity'.)

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: HIV treatment in nonpregnant adults and adolescents".)

SUMMARY AND RECOMMENDATIONS

Mitochondrial toxicity is recognized as a major adverse effect of treatment with nucleoside reverse transcriptase inhibitors (NRTIs) and can lead to myopathy, peripheral neuropathy, and hepatic steatosis with lactic acidosis, which can be life-threatening. (See 'Introduction' above.)

Mitochondria play an important role in the generation of reactive oxygen species (ROS), which are produced physiologically during oxidative phosphorylation. Oxidative stress can occur if there is an imbalance between the production of these reactive metabolites and cellular antioxidant defenses. (See 'Role of mitochondria' above.)

NRTIs are effective in inhibiting HIV replication due to their high affinity for the viral enzyme reverse transcriptase. However, NRTIs can also bind to human DNA-polymerases, like mitochondrial DNA polymerase gamma, which is exclusively responsible for the replication of mitochondrial DNA. By inhibiting this enzyme, NRTIs can cause depletion of mitochondrial DNA, resulting in organelle dysfunction and mitochondrial toxicity. (See 'Pathogenesis' above.)

The various NRTIs have different affinities for mitochondrial DNA polymerase gamma; those with the highest affinity (such as didanosine and stavudine) are associated with the highest risk of mitochondrial toxicity. Due to the significant risk of mitochondrial toxicity associated with didanosine and stavudine use, these drugs are no longer recommended for the treatment of HIV. (See 'Implicated drugs' above.)

The nucleoside analogues lamivudine (3TC), emtricitabine (FTC), abacavir (ABC), and tenofovir (TDF) lack affinity for mitochondrial DNA polymerase gamma; thus, it seems unlikely that these drugs can induce mitochondrial dysfunction of clinical significance. (See 'Implicated drugs' above.)

The clinical presentation of mitochondrial toxicity depends on the target organ that is involved. Mitochondrial toxicity is the major cause of NRTI-induced myopathy, neuropathy, lipoatrophy, and lactic acidosis. Whether mitochondrial toxicity is involved in other clinical manifestations, such as pancreatitis, myelosuppression, cardiomyopathy, proximal tubular dysfunction, or osteopenia, is not as clear. (See 'Clinical presentation' above.)

The gold standard for the diagnosis of nucleoside-related mitochondrial toxic effects is a muscle biopsy in cases of myopathy, or a liver biopsy in cases of suspected lactic acidosis with steatosis. When a patient presents with symptoms consistent with hyperlactatemia or lactic acidosis, a blood sample for lactate should be sent in a fluoride oxalate tube on ice and delivered within four hours to the laboratory for proper processing. (See 'Diagnosis' above.)

Routine monitoring for lactic acidosis is not warranted since prospective monitoring is not predictive of risk. (See 'Patient monitoring' above.)

ACKNOWLEDGMENT — We are saddened by the death of John G Bartlett, MD, who passed away in January 2021. UpToDate gratefully acknowledges Dr. Bartlett's role as section editor on this topic, his tenure as the founding Editor-in-Chief for UpToDate in Infectious Diseases, and his dedicated and longstanding involvement with the UpToDate program.

  1. Carr A, Cooper DA. Adverse effects of antiretroviral therapy. Lancet 2000; 356:1423.
  2. Brinkman K, Kakuda TN. Mitochondrial toxicity of nucleoside analogue reverse transcriptase inhibitors: a looming obstacle for long-term antiretroviral therapy? Curr Opin Infect Dis 2000; 13:5.
  3. Betteridge DJ. What is oxidative stress? Metabolism 2000; 49:3.
  4. Graziewicz MA, Day BJ, Copeland WC. The mitochondrial DNA polymerase as a target of oxidative damage. Nucleic Acids Res 2002; 30:2817.
  5. Lewis W, Dalakas MC. Mitochondrial toxicity of antiviral drugs. Nat Med 1995; 1:417.
  6. Brinkman K, ter Hofstede HJ, Burger DM, et al. Adverse effects of reverse transcriptase inhibitors: mitochondrial toxicity as common pathway. AIDS 1998; 12:1735.
  7. Kakuda TN. Pharmacology of nucleoside and nucleotide reverse transcriptase inhibitor-induced mitochondrial toxicity. Clin Ther 2000; 22:685.
  8. Morris AA, Carr A. HIV nucleoside analogues: new adverse effects on mitochondria? Lancet 1999; 354:1046.
  9. Lewis W, Day BJ, Copeland WC. Mitochondrial toxicity of NRTI antiviral drugs: an integrated cellular perspective. Nat Rev Drug Discov 2003; 2:812.
  10. Apostolova N, Blas-García A, Esplugues JV. Mitochondrial interference by anti-HIV drugs: mechanisms beyond Pol-γ inhibition. Trends Pharmacol Sci 2011; 32:715.
  11. Koczor CA, Lewis W. Nucleoside reverse transcriptase inhibitor toxicity and mitochondrial DNA. Expert Opin Drug Metab Toxicol 2010; 6:1493.
  12. Torres RA, Lewis W. Aging and HIV/AIDS: pathogenetic role of therapeutic side effects. Lab Invest 2014; 94:120.
  13. Selvaraj S, Ghebremichael M, Li M, et al. Antiretroviral therapy-induced mitochondrial toxicity: potential mechanisms beyond polymerase-γ inhibition. Clin Pharmacol Ther 2014; 96:110.
  14. Payne BA, Wilson IJ, Hateley CA, et al. Mitochondrial aging is accelerated by anti-retroviral therapy through the clonal expansion of mtDNA mutations. Nat Genet 2011; 43:806.
  15. Kakuda TN, Brundage RC, Anderson PL, Fletcher CV. Nucleoside reverse transcriptase inhibitor-induced mitochondrial toxicity as an etiology for lipodystrophy. AIDS 1999; 13:2311.
  16. Wallace DC. Mitochondrial genetics: a paradigm for aging and degenerative diseases? Science 1992; 256:628.
  17. Brinkman K, ter Hofstede HJ. Mitochondrial toxicity of nucleoside analogue reverse transcriptase inhibitors: lactic acidosis, risk factors and therapeutic options. AIDS Reviews 1999; 1:141.
  18. Martin JL, Brown CE, Matthews-Davis N, Reardon JE. Effects of antiviral nucleoside analogs on human DNA polymerases and mitochondrial DNA synthesis. Antimicrob Agents Chemother 1994; 38:2743.
  19. Birkus G, Hitchcock MJ, Cihlar T. Assessment of mitochondrial toxicity in human cells treated with tenofovir: comparison with other nucleoside reverse transcriptase inhibitors. Antimicrob Agents Chemother 2002; 46:716.
  20. Johnson AA, Ray AS, Hanes J, et al. Toxicity of antiviral nucleoside analogs and the human mitochondrial DNA polymerase. J Biol Chem 2001; 276:40847.
  21. Lim SE, Copeland WC. Differential incorporation and removal of antiviral deoxynucleotides by human DNA polymerase gamma. J Biol Chem 2001; 276:23616.
  22. McKenzie R, Fried MW, Sallie R, et al. Hepatic failure and lactic acidosis due to fialuridine (FIAU), an investigational nucleoside analogue for chronic hepatitis B. N Engl J Med 1995; 333:1099.
  23. Lewis W, Levine ES, Griniuviene B, et al. Fialuridine and its metabolites inhibit DNA polymerase gamma at sites of multiple adjacent analog incorporation, decrease mtDNA abundance, and cause mitochondrial structural defects in cultured hepatoblasts. Proc Natl Acad Sci U S A 1996; 93:3592.
  24. Medina DJ, Tsai CH, Hsiung GD, Cheng YC. Comparison of mitochondrial morphology, mitochondrial DNA content, and cell viability in cultured cells treated with three anti-human immunodeficiency virus dideoxynucleosides. Antimicrob Agents Chemother 1994; 38:1824.
  25. Cossarizza A, Moyle G. Antiretroviral nucleoside and nucleotide analogues and mitochondria. AIDS 2004; 18:137.
  26. Gardner K, Hall PA, Chinnery PF, Payne BA. HIV treatment and associated mitochondrial pathology: review of 25 years of in vitro, animal, and human studies. Toxicol Pathol 2014; 42:811.
  27. Dalakas MC, Illa I, Pezeshkpour GH, et al. Mitochondrial myopathy caused by long-term zidovudine therapy. N Engl J Med 1990; 322:1098.
  28. Moyle G. Clinical manifestations and management of antiretroviral nucleoside analog-related mitochondrial toxicity. Clin Ther 2000; 22:911.
  29. Gopinath R, Hutcheon M, Cheema-Dhadli S, Halperin M. Chronic lactic acidosis in a patient with acquired immunodeficiency syndrome and mitochondrial myopathy: biochemical studies. J Am Soc Nephrol 1992; 3:1212.
  30. Coghlan ME, Sommadossi JP, Jhala NC, et al. Symptomatic lactic acidosis in hospitalized antiretroviral-treated patients with human immunodeficiency virus infection: a report of 12 cases. Clin Infect Dis 2001; 33:1914.
  31. Arnaudo E, Dalakas M, Shanske S, et al. Depletion of muscle mitochondrial DNA in AIDS patients with zidovudine-induced myopathy. Lancet 1991; 337:508.
  32. Dalakas MC, Leon-Monzon ME, Bernardini I, et al. Zidovudine-induced mitochondrial myopathy is associated with muscle carnitine deficiency and lipid storage. Ann Neurol 1994; 35:482.
  33. Brinkman K, Smeitink JA, Romijn JA, Reiss P. Mitochondrial toxicity induced by nucleoside-analogue reverse-transcriptase inhibitors is a key factor in the pathogenesis of antiretroviral-therapy-related lipodystrophy. Lancet 1999; 354:1112.
  34. Carr A, Miller J, Law M, Cooper DA. A syndrome of lipoatrophy, lactic acidaemia and liver dysfunction associated with HIV nucleoside analogue therapy: contribution to protease inhibitor-related lipodystrophy syndrome. AIDS 2000; 14:F25.
  35. van Griensven J, De Naeyer L, Mushi T, et al. High prevalence of lipoatrophy among patients on stavudine-containing first-line antiretroviral therapy regimens in Rwanda. Trans R Soc Trop Med Hyg 2007; 101:793.
  36. Venter WDF, Kambugu A, Chersich MF, et al. Efficacy and Safety of Tenofovir Disoproxil Fumarate Versus Low-Dose Stavudine Over 96 Weeks: A Multicountry Randomized, Noninferiority Trial. J Acquir Immune Defic Syndr 2019; 80:224.
  37. Walker UA, Bickel M, Lütke Volksbeck SI, et al. Evidence of nucleoside analogue reverse transcriptase inhibitor--associated genetic and structural defects of mitochondria in adipose tissue of HIV-infected patients. J Acquir Immune Defic Syndr 2002; 29:117.
  38. Shikuma CM, Hu N, Milne C, et al. Mitochondrial DNA decrease in subcutaneous adipose tissue of HIV-infected individuals with peripheral lipoatrophy. AIDS 2001; 15:1801.
  39. Nolan D, Hammond E, Martin A, et al. Mitochondrial DNA depletion and morphologic changes in adipocytes associated with nucleoside reverse transcriptase inhibitor therapy. AIDS 2003; 17:1329.
  40. Morse CG, Voss JG, Rakocevic G, et al. HIV infection and antiretroviral therapy have divergent effects on mitochondria in adipose tissue. J Infect Dis 2012; 205:1778.
  41. Green DR, Reed JC. Mitochondria and apoptosis. Science 1998; 281:1309.
  42. Fortgang IS, Belitsos PC, Chaisson RE, Moore RD. Hepatomegaly and steatosis in HIV-infected patients receiving nucleoside analog antiretroviral therapy. Am J Gastroenterol 1995; 90:1433.
  43. Freiman JP, Helfert KE, Hamrell MR, Stein DS. Hepatomegaly with severe steatosis in HIV-seropositive patients. AIDS 1993; 7:379.
  44. Lai KK, Gang DL, Zawacki JK, Cooley TP. Fulminant hepatic failure associated with 2',3'-dideoxyinosine (ddI). Ann Intern Med 1991; 115:283.
  45. Lonergan JT, Behling C, Pfander H, et al. Hyperlactatemia and hepatic abnormalities in 10 human immunodeficiency virus-infected patients receiving nucleoside analogue combination regimens. Clin Infect Dis 2000; 31:162.
  46. Deschamps D, DeBeco V, Fisch C, et al. Inhibition by perhexiline of oxidative phosphorylation and the beta-oxidation of fatty acids: possible role in pseudoalcoholic liver lesions. Hepatology 1994; 19:948.
  47. John M, Mallal S. Hyperlactatemia syndromes in people with HIV infection. Curr Opin Infect Dis 2002; 15:23.
  48. Brinkman K. Editorial response: hyperlactatemia and hepatic steatosis as features of mitochondrial toxicity of nucleoside analogue reverse transcriptase inhibitors. Clin Infect Dis 2000; 31:167.
  49. Falcó V, Rodríguez D, Ribera E, et al. Severe nucleoside-associated lactic acidosis in human immunodeficiency virus-infected patients: report of 12 cases and review of the literature. Clin Infect Dis 2002; 34:838.
  50. Mokrzycki MH, Harris C, May H, et al. Lactic acidosis associated with stavudine administration: a report of five cases. Clin Infect Dis 2000; 30:198.
  51. Miller KD, Cameron M, Wood LV, et al. Lactic acidosis and hepatic steatosis associated with use of stavudine: report of four cases. Ann Intern Med 2000; 133:192.
  52. Lactic Acidosis International Study Group. Risk factors for lactic acidosis and severe hyperlactataemia in HIV-1-infected adults exposed to antiretroviral therapy. AIDS 2007; 21:2455.
  53. Dlamini J, Ledwaba L, Mokwena N, et al. Lactic acidosis and symptomatic hyperlactataemia in a randomized trial of first-line therapy in HIV-infected adults in South Africa. Antivir Ther 2011; 16:605.
  54. Wester CW, Eden SK, Shepherd BE, et al. Risk factors for symptomatic hyperlactatemia and lactic acidosis among combination antiretroviral therapy-treated adults in Botswana: results from a clinical trial. AIDS Res Hum Retroviruses 2012; 28:759.
  55. Chagoma N, Mallewa J, Kaunda S, et al. Longitudinal lactate levels from routine point-of-care monitoring in adult Malawian antiretroviral therapy patients: associations with stavudine toxicities. Trans R Soc Trop Med Hyg 2013; 107:615.
  56. Phan V, Thai S, Choun K, et al. Incidence of treatment-limiting toxicity with stavudine-based antiretroviral therapy in Cambodia: a retrospective cohort study. PLoS One 2012; 7:e30647.
  57. Gérard Y, Maulin L, Yazdanpanah Y, et al. Symptomatic hyperlactataemia: an emerging complication of antiretroviral therapy. AIDS 2000; 14:2723.
  58. Moyle GJ, Datta D, Mandalia S, et al. Hyperlactataemia and lactic acidosis during antiretroviral therapy: relevance, reproducibility and possible risk factors. AIDS 2002; 16:1341.
  59. Boubaker K, Flepp M, Sudre P, et al. Hyperlactatemia and antiretroviral therapy: the Swiss HIV Cohort Study. Clin Infect Dis 2001; 33:1931.
  60. Mocroft A, Phillips AN, Friis-Møller N, et al. Response to antiretroviral therapy among patients exposed to three classes of antiretrovirals: results from the EuroSIDA study. Antivir Ther 2002; 7:21.
  61. Brinkman K. Management of hyperlactatemia: no need for routine lactate measurements. AIDS 2001; 15:795.
  62. Kohler JJ, Hosseini SH, Green E, et al. Tenofovir renal proximal tubular toxicity is regulated by OAT1 and MRP4 transporters. Lab Invest 2011; 91:852.
  63. Samuels R, Bayerri CR, Sayer JA, et al. Tenofovir disoproxil fumarate-associated renal tubular dysfunction: noninvasive assessment of mitochondrial injury. AIDS 2017; 31:1297.
  64. Hart AB, Samuels DC, Hulgan T. The other genome: a systematic review of studies of mitochondrial DNA haplogroups and outcomes of HIV infection and antiretroviral therapy. AIDS Rev 2013; 15:213.
  65. Currier JS. Sex differences in antiretroviral therapy toxicity: lactic acidosis, stavudine, and women. Clin Infect Dis 2007; 45:261.
  66. Boxwell DE, Styrt BA. Lactic acidosis (LA) in patients receiving nucleoside reverse transcriptase inhibitors (NRTIs). 39th Interscience Conference on Antimicrobal Agents and Chemotherapy, San Fransisco, 1999.
  67. Kakuda TN, Brinkman K. Mitochondrial toxic effects and ribavirin. Lancet 2001; 357:1802.
  68. Bani-Sadr F, Carrat F, Pol S, et al. Risk factors for symptomatic mitochondrial toxicity in HIV/hepatitis C virus-coinfected patients during interferon plus ribavirin-based therapy. J Acquir Immune Defic Syndr 2005; 40:47.
  69. McGovern B, Bica I. Risk of HAART therapy in hepatitis C. Hepatology 2002; 35:730.
  70. Use of p24 antigen screening for HIV in primary care. AIDS Read 2001; 11:82.
  71. Mandelbrot L, Kermarrec N, Marcollet A, et al. Case report: nucleoside analogue-induced lactic acidosis in the third trimester of pregnancy. AIDS 2003; 17:272.
  72. Sarner L, Fakoya A. Acute onset lactic acidosis and pancreatitis in the third trimester of pregnancy in HIV-1 positive women taking antiretroviral medication. Sex Transm Infect 2002; 78:58.
  73. Wohl DA, Pilcher CD, Evans S, et al. Absence of sustained hyperlactatemia in HIV-infected patients with risk factors for mitochondrial toxicity. J Acquir Immune Defic Syndr 2004; 35:274.
  74. Brinkman K, Troost N, Schrijnders L, et al. Usefulness of routine lactate measurement to prevent lactic acidosis: evaluation of a protocol (abstract 709). 9th Conference on Retroviruses and Opportunistic Infections, Seattle 2002.
  75. Imhof A, Ledergerber B, Günthard HF, et al. Risk factors for and outcome of hyperlactatemia in HIV-infected persons: is there a need for routine lactate monitoring? Clin Infect Dis 2005; 41:721.
  76. Waiswa M, Byarugaba BB, Ocama P, et al. Hyperlactatemia and concurrent use of antiretroviral therapy among HIV infected patients in Uganda. Afr Health Sci 2012; 12:268.
  77. John M, Moore CB, James IR, et al. Chronic hyperlactatemia in HIV-infected patients taking antiretroviral therapy. AIDS 2001; 15:717.
  78. Côté HC, Brumme ZL, Craib KJ, et al. Changes in mitochondrial DNA as a marker of nucleoside toxicity in HIV-infected patients. N Engl J Med 2002; 346:811.
  79. Mussini C, Pinti M, Bugarini R, et al. Effect of treatment interruption monitored by CD4 cell count on mitochondrial DNA content in HIV-infected patients: a prospective study. AIDS 2005; 19:1627.
  80. Petit C, Mathez D, Barthélémy C, et al. Quantitation of blood lymphocyte mitochondrial DNA for the monitoring of antiretroviral drug-induced mitochondrial DNA depletion. J Acquir Immune Defic Syndr 2003; 33:461.
  81. Hoy JF, Gahan ME, Carr A, et al. Changes in mitochondrial DNA in peripheral blood mononuclear cells from HIV-infected patients with lipoatrophy randomized to receive abacavir. J Infect Dis 2004; 190:688.
  82. Brinkman K. Lipoatrophy and mitochondrial DNA assays: see all, know all? AIDS 2005; 19:91.
  83. Maagaard A, Holberg-Petersen M, Kollberg G, et al. Mitochondrial (mt)DNA changes in tissue may not be reflected by depletion of mtDNA in peripheral blood mononuclear cells in HIV-infected patients. Antivir Ther 2006; 11:601.
  84. Casula M, Weverling GJ, Wit FW, et al. Mitochondrial DNA and RNA increase in peripheral blood mononuclear cells from HIV-1-infected patients randomized to receive stavudine-containing or stavudine-sparing combination therapy. J Infect Dis 2005; 192:1794.
  85. Hammond EL, Sayer D, Nolan D, et al. Assessment of precision and concordance of quantitative mitochondrial DNA assays: a collaborative international quality assurance study. J Clin Virol 2003; 27:97.
  86. Alimenti A, Burdge DR, Ogilvie GS, et al. Lactic acidemia in human immunodeficiency virus-uninfected infants exposed to perinatal antiretroviral therapy. Pediatr Infect Dis J 2003; 22:782.
  87. Giaquinto C, De Romeo A, Giacomet V, et al. Lactic acid levels in children perinatally treated with antiretroviral agents to prevent HIV transmission. AIDS 2001; 15:1074.
  88. Le Chenadec J, Mayaux MJ, Guihenneuc-Jouyaux C, et al. Perinatal antiretroviral treatment and hematopoiesis in HIV-uninfected infants. AIDS 2003; 17:2053.
Topic 3765 Version 21.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟