Assessment of Mitochondrial Toxicity in Human Cells Treated with Tenofovir: Comparison with Other Nucleoside Reverse Transcriptase Inhibitors

ABSTRACT Drug-associated dysfunction of mitochondria is believed to play a role in the etiology of the various adverse symptoms that occur in human immunodeficiency virus (HIV)-infected patients treated with the nucleoside reverse transcriptase inhibitors (NRTIs). Tenofovir, a nucleotide analog recently approved for use in the treatment of HIV infection, was evaluated in vitro for its potential to cause mitochondrial toxicity and was compared to currently used NRTIs. Treatment with tenofovir (3 to 300 μM) for up to 3 weeks produced no significant changes in mitochondrial DNA (mtDNA) levels in human hepatoblastoma (HepG2) cells, skeletal muscle cells (SkMCs), or renal proximal tubule epithelial cells. The potencies of inhibition of mtDNA synthesis by the NRTIs tested were zalcitabine (ddC) > didanosine (ddI) > stavudine > zidovudine (ZDV) > lamivudine = abacavir = tenofovir, with comparable relative effects in the three cell types. Unlike ddC and ddI, tenofovir did not affect cellular expression of COX II and COX IV, two components of the mitochondrial cytochrome c oxidase complex. Lactate production was elevated by less than 20% in HepG2 cells or SkMCs following treatment with 300 μM tenofovir. In contrast, lactate synthesis increased by >200% in the presence of 300 μM ZDV. Thus, treatment of various human cell types with tenofovir at concentrations that greatly exceed those required for it both to have in vitro anti-HIV type 1 activity in peripheral blood mononuclear cells (50% effective concentration, 0.2 μM) and to achieve therapeutically relevant levels in plasma (maximum concentrations in plasma, 0.8 to 1.3 μM) is not associated with mitochondrial toxicity.

[1]  T. Cihlar,et al.  Tenofovir exhibits low cytotoxicity in various human cell types: comparison with other nucleoside reverse transcriptase inhibitors. , 2002, Antiviral research.

[2]  J. Kahn,et al.  Phase I/II Trial of the Pharmacokinetics, Safety, and Antiretroviral Activity of Tenofovir Disoproxil Fumarate in Human Immunodeficiency Virus-Infected Adults , 2001, Antimicrobial Agents and Chemotherapy.

[3]  T. Cihlar,et al.  NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS, 20(4–7), 641–648 (2001) HUMAN RENAL ORGANIC ANION TRANSPORTER 1 (hOAT1) AND ITS ROLE IN THE NEPHROTOXICITY OF ANTIVIRAL NUCLEOTIDE ANALOGS , 2003 .

[4]  C. Perry,et al.  Abacavir , 2000, Drugs.

[5]  G. Moyle Clinical manifestations and management of antiretroviral nucleoside analog-related mitochondrial toxicity. , 2000, Clinical therapeutics.

[6]  K. Miller,et al.  Lactic Acidosis and Hepatic Steatosis Associated with Use of Stavudine: Report of Four Cases , 2000, Annals of Internal Medicine.

[7]  F. Authier,et al.  Cytochrome c oxidase deficiency in the muscle of patients with zidovudine myopathy is segmental and affects both mitochondrial DNA- and nuclear DNA-encoded subunits , 2000, Acta Neuropathologica.

[8]  T. Kakuda,et al.  Pharmacology of nucleoside and nucleotide reverse transcriptase inhibitor-induced mitochondrial toxicity. , 2000, Clinical therapeutics.

[9]  V. Darley-Usmar,et al.  Differential Effects of Antiretroviral Nucleoside Analogs on Mitochondrial Function in HepG2 Cells , 2000, Antimicrobial Agents and Chemotherapy.

[10]  M. Magnani,et al.  Metabolism, mitochondrial uptake and toxicity of 2', 3'-dideoxycytidine. , 1999, The Biochemical journal.

[11]  I. Nelson,et al.  A missense mutation of cytochrome oxidase subunit II causes defective assembly and myopathy. , 1999, American journal of human genetics.

[12]  N. Chandel,et al.  Cells depleted of mitochondrial DNA (p0) yield insight into physiological mechanisms , 1999, FEBS letters.

[13]  J. Sastre,et al.  Zidovudine (AZT) causes an oxidation of mitochondrial DNA in mouse liver , 1999, Hepatology.

[14]  J. Smeitink,et al.  Adverse effects of reverse transcriptase inhibitors: mitochondrial toxicity as common pathway , 1998, AIDS.

[15]  J. Sastre,et al.  AZT treatment induces molecular and ultrastructural oxidative damage to muscle mitochondria. Prevention by antioxidant vitamins. , 1998, The Journal of clinical investigation.

[16]  L. Nijtmans,et al.  Assembly of cytochrome-c oxidase in cultured human cells. , 1998, European journal of biochemistry.

[17]  B. Robbins,et al.  Anti-Human Immunodeficiency Virus Activity and Cellular Metabolism of a Potential Prodrug of the Acyclic Nucleoside Phosphonate 9-R-(2-Phosphonomethoxypropyl)adenine (PMPA), Bis(isopropyloxymethylcarbonyl)PMPA , 1998, Antimicrobial Agents and Chemotherapy.

[18]  J. L. Smith,et al.  Expression of mtDNA and nDNA encoded respiratory chain proteins in chemically and genetically-derived Rho0 human fibroblasts: a comparison of subunit proteins in normal fibroblasts treated with ethidium bromide and fibroblasts from a patient with mtDNA depletion syndrome. , 1997, Biochimica et biophysica acta.

[19]  J. Shapiro,et al.  Zidovudine-induced fatal lactic acidosis and hepatic failure in patients with acquired immunodeficiency syndrome: report of two patients and review of the literature. , 1997, Critical care medicine.

[20]  G. Barlovatz-Meimon,et al.  Cellular and mitochondrial toxicity of zidovudine (AZT), didanosine (ddI) and zalcitabine (ddC) on cultured human muscle cells , 1997, Journal of the Neurological Sciences.

[21]  T. Cihlar,et al.  Incorporation of Selected Nucleoside Phosphonates and Anti-Human Immunodeficiency Virus Nucleotide Analogues into DNA by Human DNA Polymerases α, β and γ , 1997 .

[22]  W. Parker,et al.  Lack of mitochondrial toxicity in CEM cells treated with carbovir. , 1997, Antiviral research.

[23]  C. Perry,et al.  Lamivudine. A review of its antiviral activity, pharmacokinetic properties and therapeutic efficacy in the management of HIV infection. , 1997, Drugs.

[24]  J. Sommadossi,et al.  Effect of nucleoside analogs on neurite regeneration and mitochondrial DNA synthesis in PC-12 cells. , 1997, The Journal of pharmacology and experimental therapeutics.

[25]  R. Schinazi,et al.  Effect of beta-enantiomeric and racemic nucleoside analogues on mitochondrial functions in HepG2 cells. Implications for predicting drug hepatotoxicity. , 1996, Biochemical pharmacology.

[26]  S. Eriksson,et al.  Phosphorylation of the anti-hepatitis B nucleoside analog 1-(2'-deoxy-2'-fluoro-1-beta-D-arabinofuranosyl)-5-iodouracil (FIAU) by human cytosolic and mitochondrial thymidine kinase and implications for cytotoxicity , 1996, Antimicrobial agents and chemotherapy.

[27]  E. Levine,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. , 1996, Proceedings of the National Academy of Sciences of the United States of America.

[28]  R. O. Poyton,et al.  Crosstalk between nuclear and mitochondrial genomes. , 1996, Annual review of biochemistry.

[29]  H. Conjeevaram,et al.  Hepatic failure and lactic acidosis due to fialuridine (FIAU), an investigational nucleoside analogue for chronic hepatitis B. , 1995, The New England journal of medicine.

[30]  J. Cherrington,et al.  Kinetic Interaction of the Diphosphates of 9-(2-phosphonylmethoxyethyl)adenine and Other anti-HIV Active Purine Congeners with HIV Reverse Transcriptase and Human DNA Polymerases α, β and γ , 1995 .

[31]  M. Dalakas,et al.  Mitochondrial toxicity of antiviral drugs , 1995, Nature Medicine.

[32]  R. Schinazi,et al.  Cellular and molecular events leading to mitochondrial toxicity of 1-(2-deoxy-2-fluoro-1-beta-D-arabinofuranosyl)-5-iodouracil in human liver cells. , 1995, The Journal of clinical investigation.

[33]  J. Simpson,et al.  Mammalian DNA polymerases alpha, beta, gamma, delta, and epsilon incorporate fialuridine (FIAU) monophosphate into DNA and are inhibited competitively by FIAU Triphosphate. , 1994, Biochemistry.

[34]  J. L. Martin,et al.  Effects of antiviral nucleoside analogs on human DNA polymerases and mitochondrial DNA synthesis , 1994, Antimicrobial Agents and Chemotherapy.

[35]  C. Tsai,et al.  Comparison of mitochondrial morphology, mitochondrial DNA content, and cell viability in cultured cells treated with three anti-human immunodeficiency virus dideoxynucleosides , 1994, Antimicrobial Agents and Chemotherapy.

[36]  M. Chevallier,et al.  Fulminant hepatitis with severe lactate acidosis in HIV‐infected patients on didanosine therapy , 1994, Journal of internal medicine.

[37]  T. Sprinkle,et al.  Mitochondrial schwannopathy and peripheral myelinopathy in a rabbit model of dideoxycytidine neurotoxicity. , 1994, Laboratory investigation; a journal of technical methods and pathology.

[38]  William,et al.  Zidovudine induces molecular, biochemical, and ultrastructural changes in rat skeletal muscle mitochondria. , 1992, The Journal of clinical investigation.

[39]  J. A. Freedman,et al.  Cytochrome c oxidase: structure, function, and membrane topology of the polypeptide subunits. , 1991, Biochemistry and cell biology = Biochimie et biologie cellulaire.

[40]  W. Grody,et al.  Mitochondrial ultrastructural and molecular changes induced by zidovudine in rat hearts. , 1991, Laboratory investigation; a journal of technical methods and pathology.

[41]  C. H. Chen,et al.  Effect of anti-human immunodeficiency virus nucleoside analogs on mitochondrial DNA and its implication for delayed toxicity. , 1991, Molecular pharmacology.

[42]  S. Dimauro,et al.  Depletion of muscle mitochondrial DNA in AIDS patients with zidovudine-induced myopathy , 1991, The Lancet.

[43]  M. Dalakas,et al.  Mitochondrial myopathy caused by long-term zidovudine therapy. , 1990, The New England journal of medicine.

[44]  K. E. Follis,et al.  Toxicity and efficacy of 2',3'-dideoxycytidine in clinical trials of pigtailed macaques infected with simian retrovirus type 2 , 1989, Antimicrobial Agents and Chemotherapy.

[45]  M. Dalakas,et al.  Reversible axonal neuropathy from the treatment of AIDS and related disorders with 2′,3′‐dideoxycytidine (ddc) , 1989, Muscle & nerve.