Skip to main content
Log in

Didanosine

A Review of its Antiviral Activity, Pharmacokinetic Properties and Therapeutic Potential in Human Immunodeficiency Virus Infection

  • Drug Evaluation
  • Published:
Drugs Aims and scope Submit manuscript

Abstract

Synopsis

Didanosine is a dideoxynucleoside analogue which undergoes intracellular conversion to the putative active triphosphate metabolite. The active metabolite appears to inhibit viral reverse tran-scriptase and terminate the proviral DNA, and produces virustatic inhibition of actively replicating human immunodeficiency virus (HIV) at clinically relevant concentrations.

In phase I studies didanosine had beneficial effects on various surrogate markers of clinical efficacy and also improved clinical manifestations of HIV infection, with a 21-month survival rate of 80% in patients with acquired immune deficiency syndrome (AIDS) and 93% in patients with AIDS-related complex (ARC) in I study. Didanosine also improved CD4+ cell counts in a phase II/III trial in patients previously treated with zidovudine, whereas cell counts declined in patients continuing zidovudine therapy. However, the effects of didanosine on clinical end-points (disease progression, survival, HIV encephalopathy) remain to be established. Peripheral neuropathy and pancreatitis are the predominant dose-limiting adverse events and didanosine therapy should be withdrawn in patients developing signs or symptoms of pancreatitis and during acute treatment of Pneumocystis carinii pneumonia. However, at currently recommended clinical dosages didanosine is generally well tolerated with minimal haematological toxicity.

Thus, in a therapeutic area with few treatment options, didanosine offers a welcome alternative for patients intolerant of, or resistant to, zidovudine. There are a number of clinical trials in progress evaluating didanosine alone or in combination with other antiviral agents, and these results are awaited with considerable interest.

Pharmacodynamic Properties

Didanosine is a dideoxynucleoside analogue which undergoes intracellular phosphorylation to the putative active metabolite 2’,3’-dideoxyadenosine 5’-triphosphate (ddATP). Didanosine inhibits replication of the human immunodeficiency virus (HIV), probably via inhibition of viral reverse transcriptase and termination of the proviral DNA chain by ddATP, but is not virucidal per se.

Didanosine has dose-proportional activity against HIV in a range of in vitro models, with approximately 100% inhibition of viral replication observed at drug concentrations which are both achievable in humans and not associated with significant in vitro cytotoxicity (≈ 20 μmol/ L). In contrast to zidovudine, didanosine had minimal effects on bone marrow progenitor cells at clinically relevant concentrations, and improved the viability and function of polymorpho-nuclear leucocytes from patients with HIV infection. In vitro synergism against HIV has been observed between didanosine and various other antiviral agents including zidovudine and riba-virin. Cross-resistance to didanosine has not been observed in zidovudine-resistant clinical isolates but a decrease in HIV sensitivity has been reported after prolonged didanosine therapy.

Pharmacokinetic Properties

The pharmacokinetic properties of didanosine have been studied in patients with acquired immune deficiency syndrome (AIDS) or AIDS-related complex (ARC). Didanosine is acid labile and in clinical trials, was usually given orally on an empty stomach, either in a citrate/phosphate buffer solution or immediately after antacid administration. Under these conditions didanosine in doses up to 10.2 mg/kg had an oral bioavailability of approximately 40%, although interpatient variation was extensive. The relationship between dose and maximum plasma didanosine concentration (Cmax) was linear in this dosage range, with a Cmax of 24 μmol/L occurring approximately 0.6 hours after didanosine 10.2 mg/kg orally. At higher oral dosages, bioavailability was reduced to approximately 20%, possibly because of incomplete absorption or a saturable absorption mechanism. A buffered chewable/dispersible tablet is also available and has approximately 20 to 25% increased bioavailability compared with other formulations. The area under the plasma didanosine concentration-time curve has been correlated with reductions in p24 antigen levels and neuropsychological improvement in patients.

The plasma half-life of didanosine is short (0.6 to 1.4 hours) but the putative active metabolite ddATP has an intracellular half-life of more than 12 hours. Steady-state volume of distribution is dose-independent and approximates the volume of total body water. Didanosine concentrations in the CSF were 21% of plasma concentrations 1 hour after an intravenous infusion but were detectable in only 3 CSF samples from 20 children receiving oral didanosine 20 to 180 mg/m2. Animal studies suggest concomitant probenecid may increase didanosine CSF penetration. Didanosine undergoes extensive metabolism to ddATP, uric acid and/or purine metabolites. Didanosine clearance was dose-independent and 35 to 60% of a dose was recovered unchanged in the urine, most within the first 8 hours.

The pharmacokinetic properties of didanosine are similar after single and multiple dose administration and appear comparable in adults, children and pregnant women. Didanosine crosses the placenta. Half-life was increased approximately 3-fold in patients with renal failure but didanosine was removed by dialysis, with the half-life approximating that in patients with normal renal function

Clinical Efficacy

In phase I studies didanosine had beneficial effects on surrogate markers of HIV infection which are considered to serve as indicators of clinical efficacy. CD4+ cell counts increased, the CD4+: CD8+ ratio improved and serum viral p24 antigen levels decreased. Constitutional symptoms (fatigue, anorexia, reduced energy) improved, with resolution of oral hairy leucoplakia and peripheral neuropathy, decreased organomegaly, improvements in other haematological parameters, and increased cognitive function observed in individual studies. 21-month survival rate was 80% in patients with AIDS and 93% in patients with ARC in 1 study. The results from these studies probably underestimate the efficacy of didanosine as most evaluated a large range of dosages, with little efficacy at the lowest dosages and marked toxicity at the highest dosages. Patients with ARC, receiving adequate therapeutic dosages of didanosine, and not previously treated with zidovudine, were more likely to respond.

Interim analysis of a phase II/III trial showed CD4+ cell counts were increased at weeks 12 and 24 in patients receiving didanosine 500 or 750 mg/day, whereas they decreased in patients receiving zidovudine 600 mg/day; however, these patients had previously received long term zidovudine therapy, which probably precluded any further significant response to this agent. Preliminary results indicate that combination of didanosine with zidovudine or interferon-α also improves surrogate markers of infection.

However, none of these studies have as yet provided adequate data on the effects of didanosine treatment on the primary clinical endpoints of HIV infection, that is, disease progression and patient survival.

Tolerability

The spectrum of adverse events associated with didanosine differs from that produced by zidovudine; in particular, didanosine haematological toxicity is minimal. In phase I trials, peripheral neuropathy and pancreatitis were the major dose-limiting adverse effects associated with didanosine therapy but both were relatively infrequent at dosages less than 13 mg/kg/day. Both usually resolved upon didanosine withdrawal and the drug was reintroduced at a lower dosage without recurrence in some patients. Pancreatitis was occasionally fatal (less than 0.35% incidence in more than 10 000 patients receiving didanosine in phase II trials or the expanded access programme) and the risk of developing pancreatitis appears to correlate with advanced HIV disease, previous history of pancreatitis, poor clinical status, and concomitant use of alcohol or medications also associated with pancreatitis. Dose-limiting hepatitis, seizures, retinal atrophy, heart failure, QT prolongation, optic neuritis, rash and hypocalcaemia have also been reported.

Other adverse events occurring in patients receiving didanosine have included haematological abnormalities, laboratory abnormalities, and mild nonspecific symptoms such as headache, dry mouth, insomnia, irritability and restlessness. However, it is difficult to attribute these to didanosine as HIV infection is associated with a variety of general symptoms and abnormalities. Diarrhoea and hypokalaemia were observed in some patients but were generally attributed to the buffer solution administered with didanosine.

Dosage and Administration

Didanosine is usually administered every 12 hours and should be taken on an empty stomach, either as the buffered tablets, concomitantly with a citrate/phosphate buffer solution (pH 7 to 8) or antacid solution, or immediately after an antacid dose, to reduce acid hydrolysis. Dosage is adjusted according to bodyweight in adults and body surface area in children. Dosage recmmendations are not available for children aged less than 6 months. In patients with renal failure, didanosine dosage should be reduced 2- to 3-fold and the drug administered at the end of haemo-dialysis. Dosage reduction should also be considered in patients with hepatic impairment. Drugs with absorption dependent on gastric acidity should be given at least 2 hours before didanosine. Didanosine should be used with extreme caution in patients with an increased risk of pancreatitis and should be withdrawn if signs and symptoms of pancreatitis develop and during acute treatment of Pneumocystis carinii pneumonia.

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

Access this article

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

Instant access to the full article PDF.

Similar content being viewed by others

References

  • Ahluwalia G, Cooney DA, Mitsuya H, Fridland A, Flora KP, et al. Initial studies on the cellular pharmacology of 2′, 3′-dideox-yinosine, an inhibitor of HIV infectivity. Biochemical Pharmacology 36: 3797–3800, 1987

    Article  PubMed  CAS  Google Scholar 

  • Ahluwalia G, Johnson MA, Fridland A, Cooney DA, Broder S, et al. Cellular pharmacology of the anti-HIV agent 2′, 3′-di-deoxyadenosine. Proceedings of the American Academy of Cancer Research 29: 349P, 1988

    Google Scholar 

  • Anderson BD, Wygant MB, Xiang TX, Waugh WA, Stella VJ. Preformulation solubility and kinetic studies of 2′, 3′-dideox-ynucleosides: potential anti-AIDS agents. International Journal of Pharmaceutics 45: 27–37, 1988

    Article  CAS  Google Scholar 

  • Aoki S, Yarchoan R, Thomas RV, Pluda JM, Marczyk K, et al. Quantitative analysis of HIV-1 proviral DNA in peripheral blood mononuclear cells from patients with AIDS or ARC: decrease of proviral DNA content following treatment with 2′ 3′-dideoxyinosine (ddI). AIDS Research and Human Retro-viruses 6: 1331–1339, 1990

    CAS  Google Scholar 

  • Ashorn P, Moss B, Weinstein JN, Chaudhary VK, FitzGerald DJ, et al. Elimination of infectious human immunodeficiency virus from human T-cell cultures by synergistic action of CD4-Pseudomonas exotoxin and reverse transcriptase inhibitors. Proceedings of the National Academy of Sciences of the United States of America 87: 8889–8893, 1990

    Article  PubMed  CAS  Google Scholar 

  • Bach MC. Clinical response to dideoxyinosine in patients with HIV infection resistant to zidovudine. Correspondence. New England Journal of Medicine 323: 275, 1990

    PubMed  CAS  Google Scholar 

  • Badr MZ. Effects of the anti-AIDS drug dideoxyinosine on hepatic glycolysis in the perfused rat liver: role of intracellular calcium stores. Biochemical Pharmacology 41: 146–148, 1991

    Article  PubMed  CAS  Google Scholar 

  • Balis FM, Pizzo PA, Butler KM, Hawkins ME, Brouwers P, et al. Clinical pharmacology of 2′, 3′-dideoxyinosine in human immunodeficiency virus-infected children. Journal of Infectious Diseases 165: 99–104, 1992

    Article  PubMed  CAS  Google Scholar 

  • Balzarini J, Lee C-K, Herdewijn P, De Clercq E. Mechanism of the potentiating effect of ribavirin on the activity of 2′, 3′-di-deoxyinosine against human immunodeficiency virus. Journal of Biological Chemistry 266: 21509–21514, 1991a

    PubMed  CAS  Google Scholar 

  • Balzarini J, Lee C-K, Schols D, De Clercq E. 1-β-D-ribofuranosyl-1, 2, 4-triazole-3-carboxamide (ribavirin) and 5-ethynyl-1-β-D-ribofuranosylimidazole-4-carboxamide (EICAR) markedly potentiate the inhibitory effect of 2′, 3′-dideoxyinosine on human immunodeficiency virus in peripheral blood lymphocytes. Biochemical and Biophysical Research Communications 178: 563–569, 1991b

    Article  PubMed  CAS  Google Scholar 

  • Balzarini J, Naesens L, Robins MJ, De Clercq E. Potentiating effect of ribavirin on the in vitro and in vivo antiretrovirus activities of 2′, 3′-dideoxyinosine and 2′, 3′-dideoxy-2, 6-diami-nopurine riboside. Journal of Acquired Immune Deficiency Syndromes 3: 1140–1147, 1990

    PubMed  CAS  Google Scholar 

  • Barre-Sinoussi F, Chermann JC, Rey F, Nugeyre MT, Chamaret S, et al. Isolation of a T-lymphotrophic retrovirus from a patient at risk for acquired immune deficiency syndrome (AIDS). Science 220: 868–871, 1983

    Article  PubMed  CAS  Google Scholar 

  • Beijnen JH, Meenhorst PL, Rosing H, van Gijn R, Los G, et al. Analysis of 2′, 3′-dideoxyinosine (ddI) in plasma by isocratic high performance liquid chromatography with ultraviolet detection. Journal of Drug Development 3: 127–133, 1990

    Google Scholar 

  • Beskid G, Eskin B, Cleeland R, Siebelist J, Cappetta AJ, et al. Antibacterial activity of 2′, 3′-dideoxyadenosine in vivo and in vitro. Antimicrobial Agents and Chemotherapy 19: 424–428, 1981

    Article  PubMed  CAS  Google Scholar 

  • Bogner JR, Goebel F-D. Lymphocyte subsets as surrogate markers in antiretroviral therapy. Infection 19(Suppl. 2): S103–S108, 1991

    Article  PubMed  Google Scholar 

  • Bouvet E, Casalino E, Prevost MH, Vachon F. Fatal case of 2′, 3′-dideoxyinosine-associated pancreatitis. Correspondence. Lancet 336: 1515, 1990

    Article  PubMed  CAS  Google Scholar 

  • Butler KM, Husson RN, Balis FM, Brouwers P, Eddy J, et al. Dideoxyinosine in children with symptomatic human immunodeficiency virus infection. New England Journal of Medicine 324: 137–144, 1991

    Article  PubMed  CAS  Google Scholar 

  • Carbon C, Pinching A. Update on the ALPHA trial. Abstract. Paris 11 March, 1992

  • Chang SW, Katz MH, Hernandez SR. The new AIDS case definition. Implications for San Francisco. Editorial. Journal of the American Medical Association 267: 973–975, 1992

    Article  PubMed  CAS  Google Scholar 

  • Clavel F, Guetard D, Brun-Vezinet F, Chamaret S, Rey MA, et al. Isolation of a new human retrovirus from West African patients with AIDS. Science 233: 343–346, 1986

    Article  PubMed  CAS  Google Scholar 

  • Collier AC, Fischl MA, Kaplan LD, Northfeld D, Skolnik PR, et al. Effects of combination therapy with zidovudine ZDV and didanosine DDI on surrogate markers. Abstract 464P. VII International Conference on AIDS: Science Challenging AIDS, Florence June 16–21, 1991

    Google Scholar 

  • Connolly KJ, Allan JD, Fitch H, Jackson-Pope L, McLaren C, et al. Phase I study of 2′- 3′-dideoxyinosine administered orally twice daily to patients with AIDS or AIDS-related complex and hematologic intolerance to zidovudine. American Journal of Medicine 91: 471–478, 1991

    Article  PubMed  CAS  Google Scholar 

  • Cooley TP, Kunches LM, Saunders CA, Perkins CJ, Kelley SL, et al. Treatment of AIDS and AIDS-related complex with 2′, 3′-dideoxyinosine given once daily. Reviews of Infectious Diseases 12(Suppl. 5): S552–S560, 1990

    Article  PubMed  Google Scholar 

  • Cotton P. HIV surrogate markers weighed. Journal of the American Medical Association 265: 1357–1362, 1991a

    Article  PubMed  CAS  Google Scholar 

  • Cotton P. Race joins host of unanswered questions on early HIV therapy. Journal of the American Medical Association 265: 1065–1066, 1991b

    Article  PubMed  CAS  Google Scholar 

  • Dalgleish AG, Beverly PCL, Clapham PR, Crawford DH, Greaves MF, et al. The CD4 (T4) antigen is an essential component of the receptor for the AIDS retrovirus. Nature 312: 763–767, 1984

    Article  PubMed  CAS  Google Scholar 

  • De Jong MD, Borleffs JCC. Didanosine and heart failure. Correspondence. Lancet 339: 806–807, 1992

    Article  PubMed  Google Scholar 

  • Dornsife RE, St Clair MH, Huang AT, Panella TJ, Koszalka GW, et al. Anti-human immunodeficiency virus synergism by zidovudine (3′-azidothymidine) and didanosine (dideoxyinosine) contrasts with their additive inhibition of normal human marrow progenitor cells. Antimicrobial Agents and Chemotherapy 35: 322–328, 1991

    Article  PubMed  CAS  Google Scholar 

  • Drusano GL, Yuen GJ, Lambert JS, Seidlin M, Dolin R, et al. Relationship between dideoxyinosine exposure, CD4 counts, and p24 antigen levels in human immunodeficiency virus infection. A phase I trial. Annals of Internal Medicine 116: 562–566, 1992

    PubMed  CAS  Google Scholar 

  • Du D-L, Volpe DA, Grieshaber CK, Murphy Jr MJ. In vitro mye-lotoxicity of 2′, 3′-dideoxynucleosides on human hematopoietic progenitor cells. Experimental Hematology 18: 832–836, 1990

    PubMed  CAS  Google Scholar 

  • Editorial. Bristol-Myers Squibb’s Videx (ddI) recommended for approval in treating AIDS in adults and children intolerant or resistant to Retrovir (AZT). FDC Reports — The Pink Sheet: 3–4, 22 Jul, 1991

  • Faulds D, Heel RC. Ganciclovir. A review of its antiviral activity, pharmacokinetic properties and therapeutic efficacy in cyto-megalovirus infections. Drugs 39: 597–638, 1990

    Article  PubMed  CAS  Google Scholar 

  • Galinsky RE, Flaharty KK, Hoesterey BL, Anderson BD. Pro-benicid enhances central nervous system uptake of 2′, 3′-di-deoxyinosine by inhibiting cerebrospinal fluid efflux. Journal of Pharmacology and Experimental Therapeutics 257: 972–978, 1991

    PubMed  CAS  Google Scholar 

  • Gallo JM, Finco TS, Swagler AR, Mehta MU, Viswanathan CT, et al. Pharmacokinetic evaluation of drug interactions with anti-HIV drugs, II: effect of 2′, 3′-dideoxyinosine (ddI) on zidovudine kinetics in monkeys. AIDS Research and Human Retro-viruses 8: 277–283, 1992

    Article  CAS  Google Scholar 

  • Gao Q, Gu Z, Parniak MA, Li X, Wainberg MA. In vitro selection of variants of human immunodeficiency virus type 1 resistant to 3′-azido-3′-deoxythymidine and 2′, 3′-dideoxyino-sine. Journal of Virology 66: 12–19, 1992

    PubMed  CAS  Google Scholar 

  • Goldsmith MF. Therapeutic options expand, take new directions. Journal of the American Medical Association 266: 761–763, 1991

    Article  PubMed  CAS  Google Scholar 

  • Goldspiel BR. Status of dideoxyinosine in clinical trials. American Journal of Hospital Pharmacy 48: 1298–1299, 1991

    PubMed  CAS  Google Scholar 

  • Grady T, Powers RE, Saluja AK, Lerch MM, Steer ML. Dideoxyinosine, an AZT analogue, does not induce pancreatitis in rats. Abstract. Gastroenterology 100: A826, 1991

    Google Scholar 

  • Guyader M, Emerman M, Sonigo P, Clavel F, Montagnier L, et al. Genome organization and transactivation of the human immunodeficiency virus type 2. Nature 326: 662–669, 1987

    Article  PubMed  CAS  Google Scholar 

  • Hao Z, Cooney DA, Hartman NR, Perao CF, Fridland A, et al. Factors determining the activity of 2′, 3′-dideoxynucleosides in suppressing human immunodeficiency virus in vitro. Molecular Pharmacology 34: 431–435, 1988

    PubMed  CAS  Google Scholar 

  • Hartman NR, Ahluwalia GS, Cooney DA, Mitsuya H, Kageyama S, et al. Inhibitors of IMP dehydrogenase stimulate the phos-phorylation of the anti-human immunodeficiency virus nu-cleosides 2′, 3′-dideoxyadenosine and 2′, 3′-dideoxyinosine. Molecular Pharmacology 40: 118–124, 1991

    PubMed  CAS  Google Scholar 

  • Hartman NR, Yarchoan R, Pluda JM, Thomas RV, Marczyk KS, et al. Pharmacokinetics of 2′, 3′-dideoxyadenosine and 2′, 3′-di-deoxyinosine in patients with severe human immunodeficiency virus infection. Clinical Pharmacology and Therapeutics 47: 647–654, 1990

    Article  PubMed  CAS  Google Scholar 

  • Hartman NR, Yarchoan R, Pluda JM, Thomas RV, Wyvill KM, et al. Pharmacokinetics of 2′, 3′-dideoxyinosine in patients with severe human immunodeficiency infection II. The effects of different oral formulations and the presence of other medications. Clinical Pharmacology and Therapeutics 50: 278–285, 1991

    Article  PubMed  CAS  Google Scholar 

  • Hayashi S, Fine RL, Chou T-C, Currens MJ, Broder S, et al. In vitro inhibition of the infectivity and replication of human immunodeficiency virus type 1 by combination of antiretro-viral 2′, 3′-dideoxynucleosides and virus-binding inhibitors. Antimicrobial Agents and Chemotherapy 34: 82–88, 1990

    Article  PubMed  CAS  Google Scholar 

  • Heagy W, Crumpacker C, Lopez PA, Finberg RW. Inhibition of immune functions by antiviral drugs. Journal of Clinical Investigation 87: 1916–1924, 1991

    Article  PubMed  CAS  Google Scholar 

  • Jacobson MA, Bacchetti P, Kolokathis A, Chaisson RE, Szabo S, et al. Surrogate markers for survival in patients with AIDS and AIDS related complex treated with zidovudine. British Medical Journal 302: 73–78, 1991

    Article  PubMed  CAS  Google Scholar 

  • Johnson MA, Ahluwalia G, Connelly MC, Cooney DA, Broder S, et al. Metabolic pathways for the activation of the antire-troviral agent 2′, 3′-dideoxyadenosine, in human lymphoid cells. Journal of Biological Chemistry 263: 15354–15357, 1988

    PubMed  CAS  Google Scholar 

  • Johnson MA, Fridland A. Phosphorylation of 2′, 3′-dideoxyino-sine by cytosolic 5′-nucleotidase of human lymphoid cells. Molecular Pharmacology 36: 291–295, 1989

    PubMed  CAS  Google Scholar 

  • Johnson VA, Merrill DP, Videler JA, Chou T-C, Byington RE, et al. Two-drug combinations of zidovudine, didanosine, and recombinant interferon-α A inhibit replication of zidovudine-resistant human immunodeficiency virus type 1 synergistically in vitro. Journal of Infectious Diseases 164: 646–655, 1991

    Article  PubMed  CAS  Google Scholar 

  • Kaneshima H, Shih C-C, Namikawa R, Rabin L, Outzen H, et al. Human immunodeficiency virus infection of human lymph nodes in the SCID-hu mouse. Proceedings of the National Academy of Sciences USA 88: 4523–4527, 1991

    Article  CAS  Google Scholar 

  • Katlama C, Tubiana R, Rosenheim M, Valantin MA, Caumes E, et al. Dideoxyinosine-associated hypokalaemia. Correspondence. Lancet 337: 183, 1991

    Article  PubMed  CAS  Google Scholar 

  • Keilbaugh SA, Prusoff WH, Simpson MV. The PC 12 cell as a model for studies of the mechanism of induction of peripheral neuropathy by anti-HIV-1 dideoxynucleoside analogs. Biochemical Pharmacology 42: R5–R8, 1991

    Article  PubMed  CAS  Google Scholar 

  • Kieburtz KD, Seidlin M, Lambert JS, Dolin R, Reichman R, et al. Extended follow-up of peripheral neuropathy in patients with AIDS and AIDS-related complex treated with dideoxyi-nosine. Journal of Acquired Immune Deficiency Syndromes 5: 60–64, 1992

    PubMed  CAS  Google Scholar 

  • Knupp CA, Dixon RM, Graziano F, Dunkle LM, Barbhaiya RH. A pharmacokinetic interaction study between didanosine and ranitidine in HIV seropositive patients. Abstract 757. 31st In-terscience Conference on Antimicrobial Agents and Chemotherapy, Chicago, pp 18, Sep 29–Oct 2, 1991a

    Google Scholar 

  • Knupp CA, Shyu WC, Dolin R, Valentine FT, McLaren C, et al. Pharmacokinetics of didanosine in patients with acquired immunodeficiency syndrome or acquired immunodeficiency syndrome-related complex. Clinical Pharmacology and Therapeutics 49: 523–525, 1991b

    Article  PubMed  CAS  Google Scholar 

  • Kovacs JA, Boenning C, Davey RT, Walker RJ, Polis MA, et al. Phase I-II trial of dideoxyinosine with interferon-α in the treatment of HIV infection. Abstract. Clinical Research 39: 220A, 1991

    Google Scholar 

  • Lafeuillade A, Aubert L, Chaffanjon P, Quilichini R. Optic neuritis associated with dideoxyinosine. Correspondence. Lancet 337: 615–616, 1991

    Article  PubMed  CAS  Google Scholar 

  • Lagakos SW, Hoth DF. Surrogate markers in AIDS: where are we? where are we going? Annals of Internal Medicine 116: 599–601, 1992

    PubMed  CAS  Google Scholar 

  • Lai KK, Gang DL, Zawacki JK, Cooley TP. Fulminant hepatic failure associated with 2′, 3′-dideoxyinosine (ddI). Annals of Internal Medicine 115: 283–284, 1991

    PubMed  CAS  Google Scholar 

  • Lambert JS, Seidlin M, Reichman RC, Plank CS, Laverty M, et al. 2′, 3′-dideoxyinosine (ddI) in patients with the acquired immunodeficiency syndrome or AIDS-related complex. A phase I trial. New England Journal of Medicine 322: 1333–1340, 1990

    Article  PubMed  CAS  Google Scholar 

  • Langtry HD, Campoli-Richards DM. Zidovudine. A review of its pharmacodynamic and pharmacokinetic properties, and therapeutic efficacy. Drugs 37: 408–450, 1989

    Article  PubMed  CAS  Google Scholar 

  • Larder BA, Chesebro B, Richman DD. Susceptibilities of zido-vudine-susceptible and -resistant human immunodeficiency virus isolates to antiviral agents determined by using a quantitative plaque reduction assay. Antimicrobial Agents and Chemotherapy 34: 436–441, 1990

    Article  PubMed  CAS  Google Scholar 

  • Larder BA, Darby G, Richman DD. HIV with reduced sensitivity to zidovudine isolated during prolonged therapy. Science 243: 1731–1734, 1989

    Article  PubMed  CAS  Google Scholar 

  • Levy JA, Hoffman AD, Kramer SM, Landis JA, Shimbukuro JM, et al. Isolation of lymphocytopathic retroviruses from San Francisco patients with AIDS. Science 225: 840–842, 1984

    Article  PubMed  CAS  Google Scholar 

  • McGowan JJ, Tomaszewski JE, Cradock J, Hoth D, Grieshaber CK, et al. Overview of the preclinical development of an an-tiretroviral drug, 2′, 3′-dideoxyinosine. Reviews of Infectious Diseases 12(Suppl. 5): S513–S521, 1990

    Article  PubMed  CAS  Google Scholar 

  • Merigan TC. Treatment of AIDS with combinations of antiretro-viral agents. American Journal of Medicine 90(Suppl. 4A): 8S–17S, 1991

    Article  PubMed  CAS  Google Scholar 

  • Metroka CE, McMechan MF, Andrada R, Laubenstein LJ, Jacobus DP. Failure of prophylaxis with dapsone in patients taking dideoxyinosine. Correspondence. New England Journal of Medicine 325: 737, 1991

    PubMed  CAS  Google Scholar 

  • Mitsuya H, Broder S. Inhibition of infectivity and replication of HIV-2 and SIV in helper T-cells by 2′, 3′-dideoxynucleosides in vitro. AIDS Research and Human Retroviruses 4: 107–113, 1988

    Article  PubMed  CAS  Google Scholar 

  • Mitsuya H, Broder S. Inhibition of the in vitro infectivity and cytopathic effect of human T-lymphotrophic virus type III/ lymphadenopathy-associated virus (HTLV-III/LAV) by 2′, 3′-dideoxynucleosides. Proceedings of the National Academy of Sciences USA 83: 1911–1915, 1986

    Article  CAS  Google Scholar 

  • Mitsuya H, Jarret RF, Matsukura M, Di Marzo Veronese F, DeVico AL, et al. Long-term inhibition of human T-lymphotrophic virus type III/lymphadenopathy-associated virus (human immunodeficiency virus) DNA synthesis and RNA expression in T cells protected by 2′, 3′-dideoxynucleosides in vitro. Proceedings of the National Academy of Sciences of the United States of America 84: 2033–2037, 1987

    Article  PubMed  CAS  Google Scholar 

  • Mitsuya H, Yarchoan R, Kageyama S, Broder S. Targeted therapy of human immunodeficiency virus-related disease. FASEB Journal 5: 2369–2381, 1991

    PubMed  CAS  Google Scholar 

  • Molina J-M, Groopman JE. Bone marrow toxicity of dideoxyinosine. Correspondence. New England Journal of Medicine 321: 1478, 1989

    PubMed  CAS  Google Scholar 

  • Myers MW. New antiretroviral agents in the clinic. Reviews of Infectious Diseases 12: 944–950, 1990

    Article  PubMed  CAS  Google Scholar 

  • O’Marro SD, Armstrong JA, Asunion C, Gueverra L, Ho M. The effect of combinations of ampligen and zidovudine or dideox-yinosine against human immunodeficiency viruses in vitro. Antiviral Research 17: 169–177, 1992

    Article  PubMed  Google Scholar 

  • Pai SM, Shukla UA, Grasela TH, Knupp CA, Dolin R, et al. Population pharmacokinetic analysis of didanosine 2′, 3′-di-deoxyinosine) plasma concentrations obtained in phase I clinical trials in patients with AIDS or AIDS-related complex. Journal of Clinical Pharmacology 32: 242–247, 1992

    PubMed  CAS  Google Scholar 

  • Pedersen C, Sandström E, Sand Petersen C, Norkrans G, Gerstoft J, et al. The efficacy of inosine pranobex in preventing the acquired immunodeficiency syndrome in patients with human immunodeficiency virus infection. New England Journal of Medicine 322: 1757–1763, 1990

    Article  PubMed  CAS  Google Scholar 

  • Perno CF, Calió R, Rocchi G, Balzarini J, Broder S, et al. In vitro modulation of the activity of anti-HIV drugs in monocytes by GM-CSF and other cytokines. Abstract 105. Antiviral Research (Suppl. 1): 94, 1990a

    Google Scholar 

  • Perno CF, Cooney DA, Currens MJ, Rocchi G, Johns DG, et al. Ability of anti-HIV agents to inhibit HIV replication in mon-ocyte/macrophages or U937 monocytoid cells under conditions of enhancement by GM-CSF or anti-HIV antibody. AIDS Research and Human Retroviruses 6: 1051–1055, 1990b

    PubMed  CAS  Google Scholar 

  • Pons JC, Boubon MC, Taburet AM, Singlas E, Chambrin V, et al. Fetoplacental passage of 2′, 3′-dideoxyinosine. Correspondence. Lancet 337: 732, 1991

    Article  PubMed  CAS  Google Scholar 

  • Popovic M, Sarngadharan MG, Read E, Gallo R. Detection, isolation and continuous production of a cytopathic retrovirus (HTLV-III) from patients with AIDS and pre-AIDS. Science 224: 479–500, 1984

    Article  Google Scholar 

  • Ragni MV, Amato D, Dafni U, Korvic J, Merigan et al. Phase I/II study of combination zidovudine and didanosine in HIV positive asymptomatic patients. Abstract. Paris 11 March, 1992

  • Rathbun RC, Martin III ES, Pithey C, Matthew EB. Increases in CD4 counts are transient after initiation of ddI Abstract 25. Pharmacotherapy 11: 268, 1991

    Google Scholar 

  • Ray GF, Mason WD, Badr MZ. Pharmacokinetics of the anti-AIDS drug 2′, 3′-dideoxyinosine in the rat. Drug Metabolism and Disposition 18: 654–658, 1990

    PubMed  CAS  Google Scholar 

  • Richman DD. Dideoxynucleotides are less inhibitory in vitro against human immunodeficiency virus type 2 (HIV-2) than against HIV-1. Antimicrobial Agents and Chemotherapy 31: 1879–1881, 1987

    Article  PubMed  CAS  Google Scholar 

  • Richman DD. Susceptibility to nucleoside analogues of zidovu-dine-resistant isolate of human immunodeficiency virus. American Journal of Medicine 88(Suppl. 5B): 8S–10S, 1990a

    Article  PubMed  CAS  Google Scholar 

  • Richman DD. Zidovudine resistance of human immunodeficiency virus. Reviews of Infectious Diseases 12(Suppl. 5): S507–S512, 1990b

    Article  PubMed  Google Scholar 

  • Roilides E, Venzon D, Pizzo PA, Rubin M. Effects of antiretroviral dideoxynucleosides on polyrnorphonuclear leukocyte function. Antimicrobial Agents and Chemotherapy 34: 1672–1677, 1990

    Article  PubMed  CAS  Google Scholar 

  • Rozencweig M, McLaren C, Beltangady M, Ritter J, Canetta R, et al. Overview of phase I trials of 2′, 3′-dideoxyinosine (ddI) conducted on adult patients. Reviews of Infectious Diseases 12(Suppl. 5): S570–S575, 1990

    Article  PubMed  Google Scholar 

  • Russell JW, Klunk LJ. Comparative pharmacokinetics of new anti-HIV agents: 2′, 3′-dideoxyadenosine and 2′, 3′-dideoxyinosine. Biochemical Pharmacology 38: 1385–1388, 1989

    Article  PubMed  CAS  Google Scholar 

  • Scadden DT, Zon LI, Groopman JE. Pathophysiology and management of HIV-associated hematologic disorders. Blood 74: 1455–1463, 1989

    PubMed  CAS  Google Scholar 

  • Schinazi RF, Sommadossi J-P, Saalmann V, Cannon DL, Xie M-Y, et al. Activities of 3′-azido-3′-deoxythymidine nucleotide dimers in primary lymphocytes infected with human immunodeficiency virus type I. Antimicrobial Agents and Chemotherapy 34: 1061–1067, 1990

    Article  PubMed  CAS  Google Scholar 

  • Shirasaka T, Murakami K, Ford Jr H, Kelley JA, Yoshioka H, et al. Lipophilic halogenated congeners of 2′, 3′-dideoxypurine nucleosides active against human immunodeficiency virus in vitro. Proceedings of the National Academy of Sciences of the United States of America 87: 9426–9430, 1990

    Article  PubMed  CAS  Google Scholar 

  • Shyu WC, Knupp CA, Pittman KA, Dunkle L, Barbhaiya RH. Food-induced reduction in bioavailability of didanosine. Clinical Pharmacology and Therapeutics 50: 503–507, 1991

    Article  PubMed  CAS  Google Scholar 

  • Simpson MV, Chin CD, Keilbaugh SA, Lin T-S, Prusoff WH. Studies on the inhibition of mitochondrial DNA replication by 3′-azido-3′-deoxythymidine and other dideoxynucleoside analogs which inhibit HIV-1 replication. Biochemical Pharmacology 38: 1033–1036, 1989

    Article  PubMed  CAS  Google Scholar 

  • Singlas E, Parent O, Borsa-Lebas F, Taburet A-M, Humbert G, et al. Pharmacokinetics of dideoxyinosine (ddI) in patients with normal and impaired renal function; influence of haemodialysis. Abstract 1352. Interscience Conference on Antimicrobial Agents and Chemotherapy, Chicago, 1991

    Google Scholar 

  • Sloand EM, Pitt E, Chiarello RJ, Nemo GJ. HIV testing. State of the art. Journal of the American Medical Association 266: 2861–2866, 1991

    Article  PubMed  CAS  Google Scholar 

  • St Clair MH, Martin JL, Tudor-Williams G, Bach MC, Vavro CL, et al. Resistance to ddI and sensitivity to AZT induced by a mutation in HIV-1 reverse transcriptase. Science 253: 1557–1559, 1991

    Article  PubMed  CAS  Google Scholar 

  • Tay LK, Papp EA, Timoszyk J. Metabolism of 14C-2′, 3′-dideox-yinosine by the in situ perfused rat liver preparation. Bio-pharmaceutics and Drug Disposition 12: 285–297, 1991

    Article  CAS  Google Scholar 

  • Warner WL. Toxicology and pharmacology of adenine in animals and man. Transfusion 17: 326–332, 1977

    Article  PubMed  CAS  Google Scholar 

  • Wells KH, Poiesz BJ. Biology of retroviruses: detection, molecular biology, and treatment of retroviral infection. Journal of the American Academy of Dermatology 22: 1175–1195, 1990

    Article  PubMed  CAS  Google Scholar 

  • Whitcup SM, Butler KM, Caruso R, de Smet MD, Rubin B, et al. Retinal toxicity in human immunodeficiency virus-infected children treated with 2′, 3′-dideoxyinosine. American Journal of Ophthalmology 113: 1–7, 1992

    PubMed  CAS  Google Scholar 

  • Willocks L, Brettle R, Keen J, Valentine C, Pinching AJ. Formulations of didanosine (ddI) and salt overload. Correspondence. Lancet 339: 190, 1992

    Article  PubMed  CAS  Google Scholar 

  • Yarchoan R, Mitsuya H, Myers CE, Broder S. Clinical pharmacology of 3′-azido-2′, 3′-dideoxythymidine (zidovudine) and related dideoxynucleosides. New England Journal of Medicine 321: 726–738, 1989

    Article  PubMed  CAS  Google Scholar 

  • Yarchoan R, Mitsuya H, Pluda JM, Marczyk KS, Thomas RV, et al. The National Cancer Institute phase I study of 2′, 3′-di-deoxyinosine administration in adults with AIDS or AIDS-related complex: analysis of activity and toxicity profiles. Reviews of Infectious Diseases 12(Suppl. 5): S522–S533, 1990a

    Article  PubMed  Google Scholar 

  • Yarchoan R, Pluda JM, Thomas RV, Mitsuya H, Brouwers P, et al. Long-term toxicity/activity profile of 2′, 3′-dideoxyinosine in AIDS or AIDS-related complex. Lancet 336: 526–529, 1990b

    Article  PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Additional information

Various sections of the manuscript reviewed by: E. De Clercq, Rega Institute for Medical Research, Katholieke Universiteit Leuven, Leuven, Belgium; F.-D. Goebel, Medizinische Poliklinik der Universitat München, München, Federal Republic of Germany; C. Katlama, Groupe Hospitalier, Pitie-Salpetriere, Paris, France; J.S. Lambert, Infectious Disease Unit, University of Rochester Medical Center, Rochester, New York, USA; A. Matsuda, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan; B. Mueller, Pediatrics Branch, National Institutes of Health, National Cancer Institute, Bethesda, Maryland, USA; H. Mitsuya, Experimental Retrovirology Section, Medicine Branch, The Clinical Oncology Program, National Cancer Institute, Bethesda, Maryland, USA; H.C. Neu, Division of Infectious Diseases, College of Physicians and Surgeons of Columbia University, New York, New York, USA; A.J. Pinching, Department of Immunology, St Mary’s Hospital Medical School, London, England; PA. Pizzo, Pediatrics Branch, National Institutes of Health, National Cancer Institute, Bethesda, Maryland, USA; B. Rolinski, Medizinische Poliklinik der Universitat München, München, Federal Republic of Germany; E. Sandström, Department of Dermatology, Venhalsan, Södersjukhuset, Stockholm, Sweden; J.A. Vale, West Midlands Poisons Unit, Dudley Road Hospital, Birmingham, England; R. Yarchoan, Retroviral Diseases Section, National Cancer Institute, Bethesda, Maryland, USA.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Faulds, D., Brogden, R.N. Didanosine. Drugs 44, 94–116 (1992). https://doi.org/10.2165/00003495-199244010-00008

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/00003495-199244010-00008

Navigation