Skip to main content
Log in

Delavirdine

A Review of its Use in HIV Infection

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

Summary

Abstract

Delavirdine, a bisheteroarylpiperazine derivative, is a non-nucleoside reverse transcriptase inhibitor (NNRTI) that allosterically binds to HIV-1 reverse transcriptase, inhibiting both the RNA-and DNA-directed DNApolymerase functions of the enzyme.

Delavirdine in combination with nucleoside reverse transcriptase inhibitors (NRTIs) produced sustained reductions in plasma viral loads and improvements in immunological responses in large randomised, double-blind, placebo-controlled studies of 48 to 54 weeks’ duration. In patients with advanced HIV infection, triple therapy with delavirdine, zidovudine and lamivudine, didanosine or zalcitabine for 1 year significantly prolonged the time to virological failure compared with dual therapy (delavirdine plus zidovudine or 2 NRTIs; p < 0.0001). After 50 weeks’ treatment, plasma HIV RNA levels were below the limit of detection (LOD; <50 copies/ml) for 40% of patients receiving triple therapy but for only 6% of those receiving dual NRTI therapy.

Preliminary results suggest that delavirdine also has beneficial effects on surrogate markers as a component of protease inhibitor-containing triple or quadruple regimens. At 16 to 48 weeks, the minimum mean reduction in plasma viral load from baseline was 2.5 log 10 copies/ml and mean CD4+ counts increased by 100 to 313 cells/µ1. The proportion of patients with plasma HIV RNA levels below the LOD (usually 200 to 500 copies/ml) ranged from 48 to 100% after ≥16 weeks. Delavirdine was also effective as a component of saquinavir soft gel capsule-containing salvage regimens.

Since delavirdine shares a common metabolic pathway (cytochrome P450 3A pathway) with other NNRTIs, HIV protease inhibitors and several drugs used to treat opportunistic infections in patients infected with HIV, the drug is associated with a number of pharmacokinetic interactions. Some of these drug interactions are clinically significant, necessitating dosage adjustments or avoidance of co-administration. Delavirdine is not recommended for use with lovastatin, sim-vastatin, rifabutin, rifampicin, sildenafil, ergot derivatives, quinidine, midazolam, carbamazepine, phenobarbital or phenytoin. Importantly, the drug favourably increases the plasma concentration of several protease inhibitors.

Delavirdine is generally well tolerated. Skin rash is the most frequently reported adverse effect, occurring in 18 to 50% of patients receiving delavirdine-containing combination therapy in clinical trials. Although a high proportion of patients developed a rash, it was typically mild to moderate in intensity, did not result in discontinuation or adjustment of treatment in most patients and resolved quickly. The occurrence of Stevens-Johnson syndrome was rare (1 case in 1000 patients). A retrospective analysis of pooled clinical trial data indicated that there was no significant difference in the incidence of liver toxicity, liver failure or noninfectious hepatitis between delavirdine-containing and non-delavirdine-containing antiretroviral treatment groups. In addition, the incidence of lipodystrophy, metabolic lipid disorders, hyperglycaemia and hypertriglyceridaemia was not significantly different between these 2 treatment groups.

Conclusions: In combination with NRTIs, delavirdine produces sustained improvements in surrogate markers of HIV disease and prolongs the time to virological failure in adult patients with HIV infection. Preliminary data of delavirdine as a component of protease inhibitor-containing triple or quadruple highly active antiretroviral therapy regimens indicate that patients achieve marked improvements in virological and immunological markers. The drug is generally well tolerated, with a transient skin rash, typically of mild to moderate intensity, being the most common adverse effect. Delavirdine is an effective component of recommended antiretroviral treatment strategies for adult patients with HIV infection and, in combination with 2 NRTIs as a first-line therapy, the drug has the advantage of sparing protease inhibitors for subsequent use. Since delavirdine favourably increases plasma concentrations of several protease inhibitors, the drug may also be beneficial as a component of salvage therapy in combination with protease inhibitors.

Pharmacodynamic Profile

Delavirdine, a bisheteroarylpiperazine derivative, is a non-nucleoside reverse transcriptase inhibitor (NNRTI) that prevents HIV-1 replication by noncompetitively inhibiting reverse transcriptase. Allosteric binding of the drug results in a stable conformational change in the polymerase site that inhibits both the RNA-and DNA-directed DNA polymerase functions of the enzyme.

The drug is highly selective for HIV-1 reverse transcriptase, with minimal effects against HIV-2 reverse transcriptase or human DNA polymerase α or δ. The 50% inhibitory concentrations (IC50) of delavirdine ranged from 0.008 to 0.9 µmol/L against laboratory strains of HIV-1 in various cell culture studies. Delavirdine also exhibited good inhibitory activity against patient-derived HIV strains in cultured peripheral blood mononuclear cells, including isolates that were highly resistant to zidovudine or didanosine.

When combined with other antiretroviral agents, delavirdine has additive or synergistic activity against HIV-1 strains. There were no reports of cytotoxic or antagonistic effects of delavirdine in combination with other antiretroviral agents at 75 to 95% inhibition levels.

Resistance with delavirdine monotherapy develops rapidly. However, administration with other antiretroviral agents markedly slows the rate of acquisition of mutations. The most frequent resistance mutations associated with delavirdine occur at codons 103 [lysine to asparagine (K103N)] and 181 [tyrosine to cysteine (Y181C)], which confer resistance to nearly all NNRTIs. A single amino acid change in HIV-1 reverse transcriptase is sufficient to confer high level resistance to delavirdine, as is the case for other NNRTIs. Importantly, the potential for cross-resistance between delavirdine-resistant mutants and protease inhibitors or nucleoside reverse transcriptase inhibitors (NRTIs) is probably low.

Pharmacokinetic Profile

Delavirdine is rapidly absorbed following oral administration. Maximum plasmaconcentrations after single or multiple delavirdine 400mg doses were 7.22 and 26.6 µmol/L, respectively, and were reached in 1.17 and 2.23 hours. The reported oral bioavailability of the drug is 85%.

Administration of multiple 400mg doses of delavirdine with food does not significantly affect the rate or extent of absorption. However, concomitant administration with antacids or in patients with gastrohypoacidity reduced the rate and extent of absorption of delavirdine at steady state in HIV-infected patients.

The apparent volume of distribution of oral delavirdine was 65L. The drug is highly bound to plasma proteins (≈98%) in humans, primarily to albumin. The high level of plasma protein binding limits the amount of delavirdine available for CNS penetration, with cerebrospinal fluid concentrations averaging 0.39% of the corresponding plasma concentrations.

With regard to elimination, delavirdine demonstrates nonlinear pharmacoki-netics. The plasma elimination half-life values with single and multiple 400mg doses of delavirdine were 2.39 and 4.12 hours, with corresponding oral clearance values of 60.3 and 7.79 L/h. In healthy volunteers, similar amounts of radio-labelled drug were excreted in both faeces (≈44%) and urine (=51%).

Delavirdine undergoes extensive hepatic metabolism, with <5% of the drug appearing unchanged in the urine in healthy volunteers. The cytochrome P450 (CYP) 3A pathway is the major metabolic route, although ≈20% is metabolised by CYP2D6. The drug is extensively metabolised, mainly to desalkyl-delavirdine and unidentified pyridine hydroxy metabolites, all of which are biologically in-active. Delavirdine reversibly inhibits the activity of CYP3A and also inhibits CYP2C9.

Since delavirdine shares a common metabolic pathway (CYP3A pathway) with other NNRTIs, HIV protease inhibitors and several drugs used to treat opportunistic infections in patients infected with HIV (e.g. fluconazole, rifampicin and rifabutin), the drug is associated with a number of pharmacokinetic interactions. Some of these drug interactions are clinically significant, necessitating dosage adjustments or avoidance of coadministration. Importantly, delavirdine favourably increases the plasma concentration of several protease inhibitors.

Clinical Efficacy

In combination with nucleoside reverse transcriptase inhibitors (NRTIs): Triple therapy with delavirdine and 2 NRTIs effectively suppressed HIV viral loads (mean change from baseline 0.73 to 2.1 log10 HIV RNA copies/ml) and improved immunological responses (mean increase from baseline 65 to 111 CD4+ cells/µl) in antiretroviral-naive and -experienced patients in large randomised, double-blind, multicentre studies of 48 to 54 weeks’ duration. In patients with advanced HIV infection, delavirdine in combination with zidovudine plus lamivudine, didanosine or zalcitabine for 1 year significantly prolonged the time to virological failure compared with dual therapy (delavirdine plus zidovudine or 2 NRTIs; p < 0.001). After 50 weeks’ treatment, plasma HIV RNA levels were below the limit of detection (LOD; 50 copies/ml) for 40% of patients receiving triple therapy but for only 6% of those receiving dual NRTI therapy.

Antiretroviral therapy-naive patients with advanced HIV infection receiving triple therapy with delavirdine plus zidovudine and a second NRTI achieved more marked improvements in CD4+ cell counts than recipients of dual therapy (mean increase at 54 weeks was 122 vs 63 cells/µl; p = 0.006). 23.4% of recipients of delavirdine-containing therapy had plasma viral loads below the LOD (50 copies/ml) compared with 5.5% of recipients of dual NRTI therapy at 54 weeks (p < 0.05). In patients with prior antiretroviral experience, 13.8% achieved plasma viral loads below the LOD (50 copies/ml) with delavirdine-containing triple therapy compared with 1.6% of recipients of dual NRTI therapy.

In combination with protease inhibitors and NRTIs: Delavirdine also demonstrated beneficial effects on surrogate markers as a component of protease inhibitor-containing triple or quadruple regimens, with marked and sustained reductions in plasma viral loads (minimum mean change from baseline in HIV RNA of 2.5 logio copies/ml) and improvements in immunological responses (mean increase from baseline in CD4+ count of 100 to 313 cells/µl) at 16 to 48 weeks. Respective changes from baseline in non-delavirdine-containing therapy groups were a minimum mean decrease in HIV RNA of 1.9 logio copies/ml and an increase of 45 to 313 CD4+ cells/µl. The proportion of patients with plasma HIV RNA levels below the LOD (usually 200 to 500 copies/ml) ranged from 48 to 100% after ≥16 weeks.

There were no significant differences in beneficial effects on surrogate markers with delavirdine-containing quadruple therapy compared with triple therapy. At 24 weeks, all patients receiving delavirdine, indinavir, zidovudine and lamivudine had plasma viral loads below the LOD (400 copies/ml) compared with 67 and 95% of those receiving triple therapy with delavirdine, indinavir and zidovudine or lamivudine and 73% of those receiving indinavir, lamivudine and zidovudine. All of these groups had increases in CD4+ cell counts (≥168 cells/µl) at 48 weeks, with marked reductions in plasma viral loads (2.8 log10 copies/ml). Moreover, delavirdine plus zidovudine and indinavir significantly (p < 0.05) prolonged the time to virological failure compared with non-delavirdine-containing triple therapy.

In patients with limited antiretroviral therapy experience, delavirdine 600mg twice daily in combination with nelfinavir, stavudine and/or didanosine showed a sustained reduction in mean plasma viral load of ≥1.9 logio copies/ml (LOD 400 copies/ml) from weeks 24 to 48, with a mean change from baseline in CD4+ cell counts during this period of ≥150 cells/µl. 85% of patients achieved reductions in plasma viral load below the LOD (400 copies/ml), with 50% of patients having viral loads below 50 copies/ml.

Preliminary 16-week results indicate that recipients of delavirdine twice daily (1200 mg/day) in combination with saquinavir soft gel capsules (SGC), lamivudine and/or zidovudine achieved similar virological and immunological responses to those receiving delavirdine 3 times daily (1200 mg/day) in combination with these drugs. Delavirdine was also effective as a component of saquinavir SGC salvage regimens in patients with a median of 14 months’ indinavir experience and extensive NRTI experience. Of the 30% of patients achieving a virological response by 16 weeks (plasma viral load <500 copies/ml), 59% had sustained reductions in viral load at 48 weeks.

Tolerability

Adverse effect data from clinical trials of oral delavirdine (at a dosage of 1200 mg/day) in adult patients with HIV infection indicate the drug is generally well tolerated when given in combination with NRTIs and/or protease inhibitors. In phase I/II clinical studies, the most frequent adverse effects occurring with delavirdine, zidovudine and/or didanosine treatment were skin rash (38.3% of patients vs 4.8% with zidovudine and didanosine), headache (24.2 vs 14.6%), nausea (14.8 vs 4.9%), fatigue (14.1 vs 9.8%) and pruritus (10.2 vs 7.3%). In phase II and III clinical trials, skin rash occurred in 18 to 50% of patients receiving delavirdine-containing antiretroviral therapy and resulted in discontinuation of the drug in 4.3% of patients.

Skin rash was generally an erythematous, macropapular rash of mild to moderate intensity, was mildly pruritic, often confluent and usually developed 7 to 15 days after commencing delavirdine treatment. The rash typically peaked over 2 to 3 days and then resolved. Continuation of therapy in the presence of the rash (>85% of patients continued) did not affect the rate of resolution of the rash. Severe or life-threatening rash (e.g. erythema multiform, Stevens-Johnson syndrome) has only been reported rarely (1 case in 1000 patients) and usually resolved on discontinuation of delavirdine therapy. Rash occurred more frequently in patients with low CD4+ cell counts (<300 cells/µl), particularly those with CD4+ cell counts below 100 cells/µl, although there was no change in the severity of the rash.

A retrospective analysis of pooled clinical trial data indicated that there was no significant difference in the incidence of liver toxicity, liver failure or non-infectious hepatitis between the delavirdine and control groups receiving NRTIs and/or protease inhibitors. The incidence of grade 3 to 4 elevations of bilirubin or of the liver enzymes AST and ALT was not significantly different between the delavirdine combination (n = 5330) and non-delavirdine combination (n = 1690) therapy groups. The respective incidence of liver failure in these 2 groups was 0.11 and 0.12%, with no incidence of liver failure attributable to delavirdine therapy. Noninfectious hepatitis occurred in 0.24 and 0.29% of recipients, respectively, in the overall study population.

In addition, retrospective analysis of these pooled data indicated that the incidence of lipodystrophy, metabolic lipid disorders, hyperglycaemia and hyper-triglyceridaemia was not significantly different between delavirdine-containing and non-delavirdine-containing therapy groups.

Dosage and Administration

The recommended dosage for delavirdine in adults aged ≥16 years is 400mg (2 × 200mg tablets) 3 times daily. The drug should be used as part of a combination regimen with at least 2 other antiretroviral agents. Delavirdine may be taken without regard to food. Patients with achlorhydria should take delavirdine with an acidic beverage and the drug should be taken at least 1 hour apart from antacids. When given in combination with didanosine, delavirdine should be taken 1 hour before didanosine.

As <5% of the drug is excreted unchanged in the urine, renal impairment is not anticipated to affect plasma delavirdine concentrations; however, it is not known yet whether dosage adjustment is required in patients with HIV infection and renal impairment. Delavirdine should be used with caution in patients with hepatic impairment.

There are a number of clinically relevant drug interactions with delavirdine, necessitating dosage modifications of the drug and/or interacting agents. Coadministration of delavirdine with lovastatin, simvastatin, rifabutin, rifampicin, sildenafil, ergot derivatives, quinidine, midazolam, carbamazepine, phenobarbital or phenytoin is not recommended. Although no specific dosage adjustments have been recommended for most protease inhibitors, dosage adjustment of indinavir has been suggested. In combination with nelfinavir, the requirement for dosage adjustment of either agent is still to be determined; however, when these drugs are used in combination, absolute neutrophil counts should be monitored.

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.

Fig. 1
Fig. 2
Table I
Fig. 3
Table II
Table III
Table IV
Fig. 4
Table V
Fig. 5
Fig. 6

Similar content being viewed by others

References

  1. Fauci AS. The AIDS epidemic: considerations for the 21st century. N Engl J Med 1999 Sep 30; 341: 1046–50

    Article  PubMed  CAS  Google Scholar 

  2. Temesgen Z. Overview of HIV infection. Ann Allergy Asthma Immunol 1999 Jul; 83: 1–5

    Article  PubMed  CAS  Google Scholar 

  3. Carpenter CCJ, Cooper DA, Fischl MA, et al. Antiretroviral therapy in adults: updated recommendations of the International AIDS Society —USA Panel. JAMA 2000 Jan 19; 283: 381–90

    Article  PubMed  CAS  Google Scholar 

  4. Panel on Clinical Practices for Treatment of HIV Infection. Guidelines for the use of Antiretroviral agents in HIV-infected adults and adolescents [online]. Available from: URL: http://www.hivatis.org [Accessed 2000 Nov 21]

  5. Pozniak A, Gazzard BG, Churchill D, et al. British HIV Association (BHIVA) guidelines for the treatment of adults with HIV-infected adults with antiretroviral therapy [online]. Available from: URL: http://www.aidsmap.com/bhiva/bhivagd1299.htm [Accessed 2000 Nov 22]

  6. Moyle G. Triple nucleoside analogue antiretroviral therapy: expanding the treatment approaches for management of HIV infection. Expert Opin Invest Drug 1999 Aug; 8: 1147–55

    Article  CAS  Google Scholar 

  7. Working Group on Antiretroviral Therapy and Medical Management of HTV-Infected Children. Guidelines for the use of antiretroviral agents in pediatric HIV infection [online]. Available from: http://www.hivatis.org [Accessed 2000 Jul 12]

  8. Bonfanti P, Capetti A, Rizzardini G. HIV disease treatment in the era of HAART. Biomed Pharmacother 1999 Mar; 53: 93–105

    Article  PubMed  CAS  Google Scholar 

  9. De-Clercq E. The role of non-nucleoside reverse transcriptase inhibitors (NNRTIs) in the therapy of HIV-1 infection. Antiviral Res 1998 Jun; 38: 153–79

    Article  PubMed  CAS  Google Scholar 

  10. De Clercq E. Non-nucleoside reverse transcriptase inhibitors (NNRTIs). Expert Opin Invest Drug 1994; 3(3): 253–71

    Article  Google Scholar 

  11. Esnouf R, Ren J, Ross C, et al. Mechanism of inhibition of HIV-1 reverse transcriptase by non-nucleoside inhibitors. Nature Struct Biol 1995; 2(4): 303–8

    Article  PubMed  CAS  Google Scholar 

  12. Spence RA, Kati WM, Anderson KS, et al. Mechanism of inhibition of HIV-1 reverse transcriptase by nonnucleoside inhibitors. Science 1995; 267: 988–93

    Article  PubMed  CAS  Google Scholar 

  13. Tantillo C, Ding J, Jacobo-Molina A, et al. Locations of AntiAIDS drug binding sites and resistance mutations in the three-dimensional structure of HIV-1 reverse transcriptase. Implications for mechanisms of drug inhibition and resistance. J Mol Biol 1994; 243(3): 369–87

    CAS  Google Scholar 

  14. Kohlstaedt LA, Wang J, Friedman JM, et al. Crystal structure at 3.5A resolution of HIV-1 reverse transcriptase complexed with an inhibitor. Science 1992; 256: 1783–90

    Article  PubMed  CAS  Google Scholar 

  15. Althaus IW, Chou JJ, Gonzales AJ, et al. Kinetic studies with the non-nucleoside human immunodeficiency virus type-1 reverse transcriptase inhibitor U-90152E. Biochem Pharmacol 1994 Jun 1; 47: 2017–28

    Article  PubMed  CAS  Google Scholar 

  16. Cohen KA, Hopkins J, Ingraham RH, et al. Characterisation of the binding site for nevirapine (BI-RG-587), a nonnucleoside inhibitor of human immunodeficiency virus type-1 reverse-transcriptase. J Biol Chem 1991; 266(22): 14670–4

    PubMed  CAS  Google Scholar 

  17. Wu JC, Warren TC, Adams J, et al. A novel dipyridodiazepinone inhibitor of HIV-1 reverse transcriptase acts through a non-substrate binding site. Biochemistry 1991; 30(8): 2022–6

    Article  PubMed  CAS  Google Scholar 

  18. Esnouf RM, Ren J, Hopkins AL, et al. Unique features in the structure of the complex between HIV-1 reverse transcriptase and the bis(heteroaryl)piperazine (BHAP) U-90152 explain resistance mutations for this nonnucleoside inhibitor. Proc Natl Acad Sci U S A 1997; 94: 3984–9

    Article  PubMed  CAS  Google Scholar 

  19. Dueweke TJ, Kézdy F, Waszak GA, et al. The binding of a novel bisheteroarylpiperazine mediates inhibition of human immunodeficiency virus type 1 reverse transcriptase. J Biol Chem 1992; 267(1): 27–30

    PubMed  CAS  Google Scholar 

  20. Ding J, Das K, Moereels H, et al. Structure of HIV-1 RT/TIBO R86183 complex reveals similarity in the binding of diverse nonnucleoside inhibitors. StructBiol 1995; 2(5): 407–15

    CAS  Google Scholar 

  21. Gu Z, Quan Y, Li Z, et al. Effects of non-nucleoside inhibitors of human immunodeficiency virus type 1 in cell-free recombinant reverse transcriptase assays. J Biol Chem 1995 Dec 29; 270: 31046–51

    Article  PubMed  CAS  Google Scholar 

  22. Dueweke TJ, Poppe SM, Romero DL, et al. U-90152, a potent inhibitor of human immunodeficiency virus type 1 replication. Antimicrob Agents Chemother 1993 May; 37: 1127–31

    Article  PubMed  CAS  Google Scholar 

  23. Pagano PJ, Chong K-T. In vitro inhibition of human immunodeficiency virus type 1 by a combination of delavirdine (U-90152) with protease inhibitor U-75875 or interferon-a. J Infect Dis 1995 Jan; 171: 61–7

    Article  PubMed  CAS  Google Scholar 

  24. Witvrouw M, Pannecouque C, Van Laethem K, et al. Activity of non-nucleoside reverse transcriptase inhibitors against fflV-2 and SIV. AIDS 1999 Aug 20; 13: 1477–83

    Article  PubMed  CAS  Google Scholar 

  25. Peterson PK, Gekker G, Hu S et al. Anti-human immunodeficiency virus type 1 activities of U-90152 and U-75875 in human brain cell cultures. Antimicrob Agents Chemother 1994 Oct; 38: 2465–8

    Article  PubMed  CAS  Google Scholar 

  26. Chong K-T, Pagano PJ. Inhibition of human immunodeficiency virus type 1 infection in vitro by combination of delavirdine, zidovudine and didanosine. Antiviral Res 1997 Mar; 34: 51–63

    Article  PubMed  CAS  Google Scholar 

  27. Chong K-T, Pagano PJ, Hinshaw RR. Bisheteroarylpiperazine reverse transcriptase inhibitor in combination with 3-azido-3-deoxythymidine or 2,3-dideoxycytidine synergistically inhibits human immunodeficiency virus type 1 replication in vitro. Antimicrob Agents Chemother 1994 Feb; 38: 288–93

    Article  PubMed  CAS  Google Scholar 

  28. Pagano PJ, Chong KT. Synergistic inhibition of human immunodeficiency virus type 1 replication in vitro by two-and three-drug combinations of delavirdine, lamivudine and zidovudine. Antiviral Chem Chemother 1997; 8(4): 333–41

    CAS  Google Scholar 

  29. Mirtsuyasu RT. Interferon alpha in the treatment of AIDS-related Kaposi’s sarcoma. Br J Haematol 1991; 79 Suppl. 1: 69–73

    Article  Google Scholar 

  30. Lane HC. The role of α-interferon in patients with human immunodeficiency virus infection. Semin Oncol 1991; 18 Suppl. 7: 46–52

    PubMed  CAS  Google Scholar 

  31. Coffin JM. HIV population dynamics in vivo: implications for genetic variation, pathogenesis, and therapy. Science 1995; 267: 483–9

    Article  PubMed  CAS  Google Scholar 

  32. D’Aquila RT. HIV-1 chemotherapy and drug resistance. Clin Diagn Virol 1995; 3(4): 299–316

    Article  PubMed  Google Scholar 

  33. Hirsch MS, Brun-Vézinet F, D’Aquila RT, et al. Antiretroviral drug resistance testing in adult HIV-1 infection. recommendations of an International AIDS Society —USA Panel. JAMA 2000; 283: 2417–26

    CAS  Google Scholar 

  34. Domingo E, Martinez-Salas E, Sobrino F, et al. The quasispecies (extremely heterogeneous) nature of viral RNA genome populations: biological relevance: a review. Gene 1985; 40: 1–8

    Article  PubMed  CAS  Google Scholar 

  35. Mayers DL. Prevalence and incidence of resistance to zidovudine and other antiretroviral drugs. Am J Med 1997 May 19; 102 Suppl. 5B: 70–5

    Article  PubMed  CAS  Google Scholar 

  36. Mellors JW, Larder BA, Schinazi RF. Mutations in HIV-1 reverse transcriptase and protease associated with drug resistance. Int Antiviral News 1995; 3(1): 8–13

    Google Scholar 

  37. Bacheler LT. Resistance to non-nucleoside inhibitors of HIV-1 reverse transcriptase. Drug Resist Update 1999; 2(1): 56–67

    Article  CAS  Google Scholar 

  38. Demeter LM, Shafer RW, Meehan PM, et al. Delavirdine susceptibilities and associated reverse transcriptase mutations in human immunodeficiency virus type 1 isolates from patients in a phase I/II trial of delavirdine monotherapy (ACTG 260). Antimicrob Agents Chemother 2000 Mar; 44: 794–7

    Article  PubMed  CAS  Google Scholar 

  39. Dueweke TJ, Pushkarskaya T, Poppe SM, et al. A mutation in reverse transcriptase of bis(heteroaryl)piperazine-resistant human immunodeficiency virus type 1 that confers increased sensitivity to other nonnucleoside inhibitors. Proc Natl Acad Sci U S A 1993 May 15; 90: 4713–7

    Article  PubMed  CAS  Google Scholar 

  40. Gerondelis P, Archer RH, Palaniappan C, et al. The P236L delavirdine-resistant human immunodeficiency virus type 1 mutant is replication defective and demonstrates alterations in both RNA 5-end-and DNA 3-end-directed RNase H activities. J Virol 1999 Jul; 73: 5803–13

    PubMed  CAS  Google Scholar 

  41. Fan NS, Evans DB, Rank KB, et al. Mechanism of resistance to U-90152S and sensitization to L-697,661 by a proline to leucine change at residue 236 of human immunodeficiency virus type 1 (HIV-1) reverse transcriptase. FEBS Lett 1995 Feb 13; 359: 233–8

    Article  PubMed  CAS  Google Scholar 

  42. Griffit B, Morse G, Demeter L, et al. Relationships between delavirdine (DLV) plasma levels, HIV RNA responses and DLV resistance during combination therapy with zidovudine, and didanosine [abstract]. 12th World AIDS Conference. 1998 Jun 28; Geneva, 52

  43. 1999 Physicians GenRx. Delavirdine mesylate. In: Mosby’s Complete Drug Reference. 9th ed. 1999: II615-8

  44. Morse GD, Fischl MA, Shelton MJ, et al. Single-dose pharmacokinetics of delavirdine mesylate and didanosine in patients with human immunodeficiency virus infection. Antimicrob Agents Chemother 1997 Jan; 41: 169–74

    PubMed  CAS  Google Scholar 

  45. Cheng C-L, Smith DE, Carver PL, et al. Steady-state pharma-cokinetics of delavirdine in HIV-positive patients: effect on erythromycin breath test. Clin Pharmacol Ther 1997 May; 61: 531–43

    Article  PubMed  CAS  Google Scholar 

  46. Cox SR, Phillips L, Grasela TH. Development of a nonlinear population pharmacokinetic model for delavirdine mesylate in HIV-1 patients [abstract]. 11th World AIDS Conference. 1996 Jul 7; 1: 321

    Google Scholar 

  47. Shelton M, Adams J, Hewitt R, et al. Reduced delavirdine exposure in HIV+ subjects with gastric hypoacidity [abstract]. 35th International Conference on Antimicrobial Agents and Chemothertherapy; 1995 Sep 17–20; San Francisco (CA), 6

  48. Para M, Morese G, Fischl M, et al. Plasma protein binding of delavirdine in HIV-infected patients in ACTG 260 [abstract]. 11th World AIDS Conference. 1996 Jul 7; 78–9

  49. Morse GD, Fischl MA, Cox SR, et al. Effect of food on the steady-state pharmacokinetics of delavirdine mesylate in HIV+ patients [abstract]. 35th International Conference on Antimicrobial Agents and Chemotherapy; 1995 Sep 17–20; San Francisco (CA), 210

  50. Cox SR, Phillips L. Simulations with a population pharmacokinetic model for delavirdine predict the same systemic steady-state exposure for 600mg BID and 400mg TID regimens [abstract]. Clin Infect Dis 1998 Oct; 27: 1008

    Google Scholar 

  51. Been-Tiktak AMM, Boucher CAB, Brun-Vezinet F, et al. Efficacy and safety of combination therapy with delavirdine and zidovudine: a European/Australian phase II trial. Int J Antimicrob Agents 1999 Jan; 11: 13–21

    Article  PubMed  CAS  Google Scholar 

  52. Khalilieh SG, Forrest A, Shelton M, et al. Population pharmacokinetics of delavirdine and its major metabolite, N-desalkyl delavirdine [abstract]. Pharmacotherapy 1997 Sep–Oct; 17: 1113

    Google Scholar 

  53. Cox SR, Della-Coletta AA, Turner SW, et al. Single-dose pharmacokinetic studies with delavirdine mesylate: dose proportionality and effects of food and antacid [abstract]. 34th International Conference on Antimicrobial Agents and Chemotherapy; 1994 Oct 4–7 Orlando (FL), 82

  54. Staton BA, Johnson MG, Friis JM, et al. Simple, rapid and sensitive high-performance liquid Chromatographic determination of delavirdine and its N-desisopropyl metabolite in human plasma. J Chromatogr B Biomed Appl 1995 Jun 9; 668: 99–106

    Article  PubMed  CAS  Google Scholar 

  55. Veldkamp AI, van Heeswijk RP, Hoetelmans RM, et al. Rapid quantification of delavirdine, a novel non-nucleoside reverse transcriptase inhibitor, in human plasma using isocratic reversed-phase high-performance liquid chromatography with fluorescence detection. J Chromatogr B Biomed Sci Appl 1999 Apr 30; 727: 151–7

    Article  PubMed  CAS  Google Scholar 

  56. Welage LS, Carver PL, Revankar S, et al. Alterations in gastric acidity in patients infected with human immunodeficiency virus. Clin Infect Dis 1996; 21: 1431–8

    Article  Google Scholar 

  57. Chaput AJ, D’Ambrosio R, Morse GD. In vitro protein-binding characteristics of delavirdine and its N-dealkylated metabolite. Antiviral Res 1996 Oct; 32: 81–9

    Article  PubMed  CAS  Google Scholar 

  58. Davey Jr RT, Chaitt DG, Reed GF, et al. Randomized, controlled phase I/II trial of combination therapy with delavirdine (U-90152S) and conventional nucleosides in human immunodeficiency virus type 1-infected patients. Antimicrob Agents Chemother 1996 Jul; 40: 1657–64

    PubMed  CAS  Google Scholar 

  59. Voorman RL, Maio SM, Hauer MJ, et al. Metabolism of delavirdine, a human immunodeficiency virus type-1 reverse transcriptase inhibitor, by microsomal cytochrome P450 in humans, rats, and other species: probable involvement of CYP2D6 and CYP3A. Drug Metab Dispos 1998 Jul; 26: 631–9

    PubMed  CAS  Google Scholar 

  60. Voorman RL, Maio SM, Payne NA, et al. Microsomal metabolism of delavirdine: evidence for mechanism-based inactivation of human cytochrome P450 3A. J Pharmacol Exp Ther 1998 Oct; 287: 381–8

    PubMed  CAS  Google Scholar 

  61. Chang M, Sood VK, Kloosterman DA, et al. Identification of the metabolites of the HIV-1 reverse transcriptase inhibitor delavirdine in monkeys. Drug Metab Dispos 1997 Jul; 25: 814–27

    PubMed  CAS  Google Scholar 

  62. Chang M, Sood VK, Wilson GJ, et al. Metabolism of the HIV-1 reverse transcriptase inhibitor delavirdine in mice. Drug Metab Dispos 1997 Jul; 25: 828–39

    PubMed  CAS  Google Scholar 

  63. Chang M, Sood VK, Wilson GJ, et al. Metabolism of the human immunodeficiency virus type 1 reverse transcriptase inhibitor delavirdine in rats. Drug Metab Dispos 1997 Feb; 25: 228–42

    PubMed  CAS  Google Scholar 

  64. American Society of Hospital Pharmacists. Delavirdine mesylate. In: McEvoy GK, editor. American Hospital Formulary Services Drug Information. Bethesda (MD): Datapharma Publications Limited, 2000: 597–600.

    Google Scholar 

  65. Wintergerst U, Engelhorn C, Kurowski M, et al. Pharmacokinetic interaction of amprenavir in combination with efavirenz or delavirdine in HIV-infected. AIDS 2000; 14(12): 1866–8

    Article  PubMed  CAS  Google Scholar 

  66. Tran JQ, Petersen C, Garrett M, et al. Delavirdine significantly increases plasma concentrations of amprenavir in healthy volunteers [abstract]. Proceedings of the 5th International Conference on Drug Therapy in HIV Infection; 2000 Oct 22–26; Glasgow

  67. Ferry JJ, Herman BD, Carel BJ, et al. Pharmacokinetic drug-drug interaction study of delavirdine and indinavir in healthy volunteers. J Acquir Immune Defic Syndrom Hum Retrovirol 1998 Jul 1; 18: 252–9

    Article  CAS  Google Scholar 

  68. Tran JQ, Petersen C, Garrett MK, et al. The pharmacokinetics (PK) and tolerability of indinavir (IDV) and delavirdine (DLV) administered in the absence and presence of food in healthy volunteers [abstract/poster]. Proceedings of the 40th Interscience Conference on Antimicrobial Agents and Chemotherapy. 2000 Sep 17–20; Toronto

  69. National AIDS Treatment Advocacy Project. NATAP reports 1998 Jan; 1 (3–4):19–21 [online]. Available from: URL: http://www.natap.org/reports/report3.html [Accessed 2000 Nov 21]

  70. Cox SR, Schneck DW, Herman BD, et al. Delavirdine (DLV) and nelfinavir (NFV): a pharmacokinetic (PK) drug-drug interaction study in healthy adult volunteers [abstract no. 345]. 5th Conference on Retroviral and Opportunistic Infections. 1998 Feb 1–5; Chicago

  71. Shelton MJ, Hewitt RG, Adams JM, et al. Delavirdine mesylate pharmacokinetics during combination therapy with ritonavir [abstract]. 37th International Conference on Antimicrobial Agents and Chemotherapy; 1997 Sep 28; Toronto, 13

  72. Morse GD, Shelton MJ, Hewitt RG, et al. Ritonavir (RIT) pharmacokinetics (PK) during combination therapy with delavirdine (DLV) [abstract no. 343]. 5th Conference of Retroviral and Opportunistic Infections; 1998 Feb 1–5; Chicago

  73. Cox S, Conway B, Freimuth W, et al. Pilot study of BID and TID combination of saquinavir-SGC (S),delavirdine (D), zidovudine (ZDV) and lamivudine (3TC) as initial therapy: pharmacokinetic (PK) interaction between S-SGC and D [abstract no. 82]. 7th Conference on Retroviruses and Opportunistic Infections. 2000 Jan 30–Feb 2; San Francisco

  74. Cox SR, Ferry JJ, Batts DH, et al. Delavirdine (D)and marketed protease inhibitors (PIs): pharmacokinetic (PK) interaction studies in healthy volunteers [abstract no. 372]. 4th Conference on Retroviruses and Opportunistic Infections; 1997 Jan 22–26; Washington, DC

  75. Cox SR, Cohn SE, Greisberger C, et al. Evaluation of the steady-state pharmacokinetic interaction between didanosine and delavirdine mesylate in HIV+ patients [abstract]. 35th International Conference on Antimicrobial Agents and Chemotherapy; 1995 Sep 17–20; San Francisco (CA), 210

  76. Morse GD, Cox SR, DeRemer MF, et al. Zidovudine pharmacokinetics during an escalating, multiple-dose study of delavirdine mesylate [abstract]. 34th International Conference on Antimicrobial Agents and Chemotherapy; 1994 Oct 4–7 Orlando (FL), 132

  77. Borin MT, Cox SR, Herman BD, et al. Effect of fluconazole on the steady-state pharmacokinetics of delavirdine in human immunodeficiency virus-positive patients. Antimicrob Agents Chemother 1997 Sep; 41: 1892–7

    PubMed  CAS  Google Scholar 

  78. Borin MT, Chambers JH, Carel BJ, et al. Pharmacokinetic study of the interaction between rifabutin and delavirdine mesylate in HIV-1 infected patients. Antiviral Res 1997 Jun; 35: 53–63

    Article  PubMed  CAS  Google Scholar 

  79. Borin MT, Chambers JH, Carel BJ, et al. Pharmacokinetic study of the interaction between rifampin and delavirdine mesylate. Clin Pharmacol Ther 1997 May; 61: 544–53

    Article  PubMed  CAS  Google Scholar 

  80. Malaty LI, Kuper JJ. Drug interactions of HIV protease inhibitors. Drug Saf 1999; 20(2): 147–69

    Article  PubMed  CAS  Google Scholar 

  81. Raboud JM, Montaner JSG, Conway B, et al. Suppression of plasma viral load below 20 copies/ml is required to achieve a long-term response to therapy. AIDS 1998; 12: 1619–24

    Article  PubMed  CAS  Google Scholar 

  82. Powderly WG, Saag MS, Chapman S, et al. Predictors of optimal virological response to potent antiretroviral therapy. AIDS 1999; 13: 1873–80

    Article  PubMed  CAS  Google Scholar 

  83. Gatell J, Kuritzkes D, Green S, et al. Twice daily dosing of delavirdine in combination with nelfinavir, didanosine, and stavudine results in significant decreases in viral burden [abstract]. 39th Interscience Conference on Antimicrobial Agents and Chemotherapy. 1999 Sep 26; San Francisco; 520

  84. Willoughby R, Watson D, Welliver R, et al. Phase I evaluation of delavirdine in HIV-1-infected pediatric patients [abstract]. 39th Interscience Conference on Antimicrobial Agents and Chemotherapy. 1999 Sep 26; San Francisco; 525

  85. Para MF, Meehan P, Holden-Wiltse J, et al. ACTG 260: a randomized, phase I–II, dose-ranging trial of the anti-human immunodeficiency virus activity of delavirdine monotherapy. Antimicrob Agents Chemother 1999 Jun; 43: 1373–8

    PubMed  CAS  Google Scholar 

  86. Friedland GH, Pollard R, Griffith B, et al. Efficacy and safety of delavirdine mesylate with zidovudine and didanosine compared with two-drug combinations of these agents in persons with HIV disease with CD4 counts of 100 to 500 cells/mm3 (ACTG 261). J Acquir Immune Defic Syndrom Hum Retrovirol 1999 Aug 1; 21: 281–92

    Article  CAS  Google Scholar 

  87. Moyle G, de Cian CW, Hawkins D, et al. Final 54-week analysis of a placebo-controlled trial (13C) of delavirdine plus two nucleoside analogs (NA) versus two NA in drug-naive and -experienced indivduals [abstract]. 39th ICAAC 1999 Sep 26: 520

    Google Scholar 

  88. Para M, Conway B, Green S, et al. Safety and efficacy of delavirdine in combination with zidovudine and lamivudine: final 52-week analysis [abstract/poster]. 39th Interscience Conference on Antimicrobial Agents and Chemotherapy. 1999 Sep 26; San Francisco; 520

  89. Hawkins D, Wood R, Horban A, et al. Final 54-week analysis of a study comparing the safety and antiretroviral activity of dual nucleoside analogues plus delavirdine (DLV) in an advanced treatment-naive HIV-infected patients [abstract no. 550]. 7th European Conference on Clinical Aspects and Treatment of HIV-Infection; 1999 Oct 23; Lisbon, 119

  90. Hawkins D, Wood R, Horban A, et al. Final 54-week analysis of a study comparing the safety and the antiretroviral activity of dual nucleosides analogues plus placebo versus dual nucleoside analogues plus delavirdine in an advanced, nucleo-side analogue-experienced HIV-infected population. 7th European Conference on Clinical Aspects and Treatment of fflV-Infection; 1999 Oct 23; Lisbon, 119–120

  91. Conway B, Cisneros LN, Williams K, et al. Randomized, open-label comparison of saquinavir soft gel capsules (SQV), lamivudine (3TC) and delavirdine (DLV) twice or thrice daily vs SQV/3TC/zidovudine (ZDV) vs SQV/3TC/ZDV/DLV in the initial treatment of HIV infection [abstract]. Can J Infect Dis 1999 Sep–Oct; 10 Suppl. D: 26D

    Google Scholar 

  92. Eron J, McKinley G, LeClercq P, et al. Potential antiviral activity using delavirdien and reduced-dose indinavir combination therapies [abstract]. Proceedings of the 7th Conference on Retroviruses and Opportunistic Infections; 2000 Jan 30–Feb 2; San Franscico

  93. Preliminary study results show use of Rescriptor with indinavir at lower doses decreases viral load. [Press release]

  94. Kuritzkes DR, Bassett RL, Johnson VA, et al. Continued lamivudine versus delavirdine in combination with indinavir and zidovudine or stavudine in lamivudine-experienced patients: results of Adults AIDS Clinical Trials protocol Group 370. AIDS 2000; 14(11): 1553–61

    Article  PubMed  CAS  Google Scholar 

  95. Para M, Beal J, Rathbun C, et al. Potent activity with lower doses of indinavir, using delavirdine in combination with zidovudine: 48-week analysis [abstract]. 39th Interscience Conference on Antimicrobial Agents and Chemotherapy; 1999 Sep 26; San Francisco; 522

  96. Walmsley S, Wathen L, Paxton L, et al. Durable viral suppression with rescriptor (delavirdine) and indinavir combination therapies [abstract]. Can J Infect Dis 1999 Mar–Apr; 10 Suppl. B: 25B

    Google Scholar 

  97. Blanco JL, Mallolas J, Sarasa M, et al. A pilot study of a twice daily (BID) combination of indinavir/delavirdine plus nucleoside analogues for salvage therapy in HTV-1 infected patients [abstract]. Proceedings of the 40th Interscience Conference on Antimicrobial Agents and Chemotherapy; 2000 Sep 17–20; Toronto

  98. Gulick RM, Hu XJ, Fiscus SA, et al. Salvage therapy with saquinavir SGC (SQV) in combination with ritonavir (RTV) or nelfinavir (NFV) plus delavirdine (DLV), adefovir dipivoxil (ADV), or both —ACTG 359 [abstract no. 235]. 7th Conference on Retroviruses and Opportunistic Infections; 2000 Jan 30–Feb 2; San Francisco (CA)

  99. Freimuth WW. Delavirdine mesylate, a potent non-nucleoside HIV-1 reverse transcriptase inhibitor. Adv Exp Med Biol 1996; 394: 279–89

    PubMed  CAS  Google Scholar 

  100. Para M, Slater L, Daly P, et al. Delavirdine in combination therapy has a favorable liver safety profile in HIV-1 patients [abstract]. 39th Interscience Conference on Antimicrobial Agents and Chemotherapy. 1999 Sep 26; San Francisco; 17

  101. Para M, Conway B, Kuritzkes D, et al. Treatment with delavirdine in combination with NRTIs and/or PIs is not associated with lipodistrophy or metabolic lipid/glucose disturbances [abstract no. 66]. Antivir Ther 1999; 4: 70

    Google Scholar 

  102. Mocroft A, Vella S, Benfield TL, et al. Changing patterns of mortality across Europe in patients infected with HIV-1. Lancet 1998; 352: 1725–30

    Article  PubMed  CAS  Google Scholar 

  103. Palella Jr FJ, Delaney KM, Moorman AC, et al. Declining morbidity and mortality among patients with advanced human immunodeficiencey virus infection. N Engl JMed 1998; 338: 853–60

    Article  Google Scholar 

  104. Vittinghoff E, Scheer S, O’Malley P, et al. Combination antiretroviral therapy and recent declines in AIDS incidence and mortality. J Infect Dis 1999; 179: 717–20

    Article  PubMed  CAS  Google Scholar 

  105. Boerma JT, Nunn AJ, Whitworth JAG. Mortality impact of the AIDS epidemic: evidence from community studies in less developed countries. AIDS 1998; 12 Suppl. 1: S3–14

    PubMed  Google Scholar 

  106. Wong JK, Hezareh M, Gunthard HF, et al. Recovery of replication-competent HIV despite prolonged suppression of plasma viremia. Science 1997; 278: 1291–5

    Article  PubMed  CAS  Google Scholar 

  107. Finzi D, Blankson J, Siliciano JD, et al. Latent infection of CD4+ T cells provides a mechanism for lifelong persistence of HIV-1, even in patients on effective combination therapy. Nature Med 1999; 5(5): 512–7

    Article  PubMed  CAS  Google Scholar 

  108. Finzi D, Hermankova M, Pierson T, et al. Identification of a reservoir for HIV-1 in patients on highly active antiretroviral therapy. Science 1997; 278: 1295–300

    Article  PubMed  CAS  Google Scholar 

  109. Harrigan PR, Whaley M, Montaner JSG. Rate of HIV-1 RNA rebound upon stopping antiretroviral therapy. AIDS 1999; 13: F59–62

    Article  PubMed  CAS  Google Scholar 

  110. Holtzer CD, Roland M. The use of combination antiretroviral therapy in HIV-infected patients. Ann Pharmacother 1999 Feb; 33: 198–209

    Article  PubMed  CAS  Google Scholar 

  111. Reedijk M, Wigersma L, Mohrs J. When do HIV-infected persons start with antiretroviral therapy? A retrospective analysis of patients’ monitoring and treatment status in general practice, as compared with the 1991 Dutch HIV treatment guidelines. Fam Pract 1998 Dec; 15: 525–8

    Article  PubMed  CAS  Google Scholar 

  112. Gazzard B, Moyle G, BHIVA Guidelines Writing Committee. 1998 revision to the British HIV Association guidelines for antiretroviral treatment of HIV seropositive individuals. Lancet 1998 Jul 25; 352: 314–6

    Article  PubMed  CAS  Google Scholar 

  113. Clumeck N. Choosing the best initial therapy for HIV-1 infection. N Engl J Med 1999; 341(25): 1925–6

    Article  PubMed  CAS  Google Scholar 

  114. Friedland GH, Williams A. Attaining higher goals in HIV treatment: the central importance of adherence. AIDS 1999; 13 Suppl. 1: S61–72

    PubMed  Google Scholar 

  115. Moyle GJ, Gazzard BG. A risk-benefit assessment of HIV protease inhibitors. Drug Saf 1999 Apr; 20: 299–321

    Article  PubMed  CAS  Google Scholar 

  116. Rana KZ, Dudley MN. Human immunodeficiency virus protease inhibitors. Pharmacotherapy 1999 Jan; 19: 35–59

    Article  PubMed  CAS  Google Scholar 

  117. Barsdley-Elliot A, Perry CM. Nevirapine: a review of its use in the prevention and treatment of paediatric infection. Paediatr Drugs 2000; 2(4): 373–407

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Lesley J. Scott.

Additional information

Various sections of the manuscript reviewed by: M. Barry, National Centre for Pharmacoeconomics, St. James’s Hospital, Dublin, Ireland; B.G. Gazzard , Chelsea and Westminster Hospital, London, England; P. Hay, St. George’s Healthcare NHS Trust, St.George’s Hospital, London, England; L. Naessens, Rega Institute for Medical Research, Katholieke University Leuven, Leuven, Belgium; M. Para, Division of Infectious Diseases, Ohio State University Hospitals, Columbus, Ohio, USA; A-M. Taburet, Bicêtre Hospital, Paris, France

Data Selection

Sources: Medical literature published in any language since 1980 on Delavirdine, identified using Medline and EMBASE supplemented by AdisBase (a proprietary database of Adis International, Auckland, New Zealand). Additional references were identified from the reference lists of published articles. Bibliographical information, including contributory unpublished data, was also requested from the company developing the drug.

Search strategy: Medline search terms were ‘Delavirdine’ or ‘Delavirdine mesylate’ or ‘U-90152’ or ‘U-90152S’. EMBASE search terms were ‘Delavirdine’ or ‘Delavirdine mesylate’ or ‘U-90152’ or ‘U-90152S’. AdisBase search terms were ‘Delavirdine’ or ‘Delavirdine-mesylate’ or ‘U-90152’ or ‘U-90152S’. Searches were last updated 31 Oct 2000.

Selection: Studies in patients with HIV infection who received delavirdine. Inclusion of studies was based mainly on the methods section of the trials. When available, large, well controlled trials with appropriate statistical methodology were preferred. Relevant pharmacodynamic and pharmacokinetic data are also included.

Index terms: HIV infection, non-nucleotide reverse transcriptase inhibitor, pharmacodynamics, pharmacokinetics, therapeutic use.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Scott, L.J., Perry, C.M. Delavirdine. Drugs 60, 1411–1444 (2000). https://doi.org/10.2165/00003495-200060060-00013

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/00003495-200060060-00013

Keywords

Navigation