Skip to main content
Log in

Atovaquone

A Review of its Pharmacological Properties and Therapeutic Efficacy in Opportunistic Infections

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

Abstract

Synopsis

Atovaquone has been investigated as an alternative agent for oral use in the treatment of both mild to moderate Pneumocystis carinii pneumonia (PCP) and toxoplasmosis, opportunistic infections commonly experienced by patients with AIDS. In patients with mild to moderate PCP, a dosage of 750mg 3 times daily (administered in tablet form) has similar overall therapeutic efficacy (defined as clinical response without a treatment-limiting adverse event) to the conventional therapies oral cotrimoxazole (trimethoprim-sulfamethoxazole) and intravenous pentamidine, respectively. Response rates to atovaquone are lower than those achieved with cotrimoxazole, but atovaquone has superior tolerability. Atovaquone recipients experienced significantly fewer treatment-limiting adverse effects than patients treated with cotrimoxazole (7 vs 20%) or pentamidine (4 vs 36%).

Mortality rates were higher among atovaquone-treated patients than in cotrimoxazole recipients (7 vs 0.6%) 4 weeks after completion of therapy in a large comparative trial, although most deaths were caused by bacterial infections. However, a similar rate of mortality was reported for atovaquone- and pentamidine-treated patients (16 vs 17% 8 weeks after discontinuation of therapy) in another study.

In predominantly small numbers of patients with toxoplasmosis, of whom most were unresponsive to conventional agents, atovaquone 750mg 4 times daily (administered as tablets) produced a complete or partial radiological response rate of 37 to 87.5%. 52% of patients achieved a complete or partial clinical response after 6 weeks of treatment in the largest trial (n = 87), although the incidence of toxoplasmosis-related death was 24% 18 weeks after therapy was initiated.

Thus, atovaquone will be a useful option for the treatment of patients with mild to moderate PCP who are intolerant or unresponsive to cotrimoxazole, especially if the increased plasma drug concentrations observed with the suspension further improve response rates. Atovaquone should also be considered a promising agent for the treatment of toxoplasmosis.

Pharmacodynamic Properties

Atovaquone is a hydroxynaphthoquinone which acts as an inhibitor of the mitochondrial respiratory chain. Atovaquone has intrinsic activity against Pneumocystis carinii; a median drug concentration of 1.4 μmol/L (0.513 mg/L) inhibited 50% of organisms (IC50) in vitro. The IC50 value of atovaquone was lower [about 0.05 μmol/L (0.018 mg/L)] when measured by oxygen consumption of isolated P. carinii from rat lungs. Atovaquone 100 mg/kg/day demonstrated activity as a prophylactic agent against, and as a treatment of, P. carinii pneumonia (PCP) in the corticosteroid-treated rat model. It had greater prophylactic activity than intravenous pentamidine or clindamycin plus primaquine, similar activity to dapsone plus trimethoprim, erythromycin plus sulfafurazole or pyrimethamine plus sulfadoxine, but lower activity than cotrimoxazole (trimethoprim-sulfamethoxazole), azithromycin plus sulfamethoxazole or clarithromycin plus sulfamethoxazole in this model. It was less effective than cotrimoxazole or pyrimethamine plus dapsone or in corticosteroid-treated rats with concomitant P. carinii and Toxoplasma gondii infection. The therapeutic and prophylactic activity of atovaquone is dose-proportional. Importantly, it appears that atovaquone may be microbicidal against P. carinii, in contrast with cotrimoxazole, which is microbistatic. No antagonistic effects were noted in mice given atovaquone in combination with other agents used in the treatment of opportunistic infections.

In a few patients with PCP, 42 days of therapy with atovaquone removed all signs of P. carinii in bronchoalveolar lavage fluid. The recommended regimen of 21 days’ therapy reduced this ratio by more than 50%, which is associated with a decreased risk of relapse within 6 months. Measurement of arterial blood gas (a measure of pneumonia resolution) showed that treatment with atovaquone 750mg 3 times daily improves oxygenation in patients with AIDS and PCP.

Atovaquone has activity against T gondii in in vitro models, with significant differences in susceptibility of different strains. Intracellular replication of 6 of 7 strains of T. gondii tachyzoites were inhibited by a drug concentration of 4.8 × 10−9 to 4.8 × 10−6 mol/L (0.00176 to 1.76 mg/L); the IC50 value of atovaquone against the virulent RH strain was 6.4 × 10−8 mol/L (0.0235 mg/L). Higher concentrations of atovaquone [2.8 × 10−4 mol/L (100 mg/L)] are required to kill bradyzoites within cysts, although it was the only agent, when compared with clindamycin, fluorouracil, pyrimethamine and sulfadiazine, to demonstrate cysticidal activity. Atovaquone was less effective when the time to initiation of treatment was increased or lower drug doses were administered. In mice with chronic T. gondii infection, treatment with atovaquone 100 or 200 mg/kg/day for 2 to 12 weeks resulted in a steady decline in the number of brain cysts compared with untreated controls.

Pharmacokinetic Properties

Atovaquone has slow and irregular absorption when administered orally in tablet form. Maximum plasma concentrations (Cmax) and area under the concentration-time curve at steady-state increased linearly over the dosage range 100 to 750 mg/day in volunteers with asymptomatic HIV infection. Mean Cmax at steady-state was lower in patients with AIDS and PCP than in volunteers with asymptomatic HIV infection; administration of atovaquone 750mg 3 times daily in tablet form resulted in a mean Cmax of 13.9 mg/L in patients with AIDS and PCP. Atovaquone has a long terminal plasma half-life (77 hours, when administered at a dosage of 750mg 3 then 2 times daily). Increased drug absorption (at least 3-fold) is observed when atovaquone is administered with or soon after meals, particularly a high fat meal, compared with the fasting state. Administration of atovaquone as an aqueous suspension, compared with as a tablet, also improves drug absorption approximately 2-fold. After undergoing enterohepatic cycling, atovaquone is predominantly excreted in the faeces in healthy volunteers. Less than 0.6% of the drug is renally excreted. Atovaquone is more than 99.9% protein bound and does not cross the blood-brain barrier to any appreciable extent.

Steady-state plasma atovaquone concentrations (Css) have been correlated with therapeutic response and survival and, in patients with toxoplasmosis, delay to disease progression. It appears that the incidence of some adverse events, particularly rash, is also correlated with Css in atovaquone-treated patients. The pharmacokinetic profile of atovaquone has not been evaluated in the elderly or in patients with renal or hepatic impairment, and only limited study has been performed in children.

Therapeutic Efficacy

In all clinical trials of atovaquone identified, the drug was administered orally, in tablet form.

Patients enrolled in trials assessing the efficacy of atovaquone against PCP generally had mild to moderate disease and all had AIDS. In noncomparative trials, treatment with atovaquone 750mg 2 to 4 times daily produced a clinical response in 77 to 83% of patients. In comparative trials, atovaquone 750mg 3 times daily demonstrated similar overall therapeutic efficacy (defined as clinical response without development of a treatment-limiting adverse event) to the conventional therapies, oral cotrimoxazole and intravenous pentamidine. Fewer atovaquone than cotrimoxazole recipients responded, although the incidence of treatment-limiting adverse effects was significantly lower in atovaquone-treated patients than in cotrimoxazole or pentamidine recipients. Mortality rates after completion of therapy were higher in atovaquone, than in cotrimoxazole, recipients (7 vs 0.6% at 4 weeks follow-up; most deaths were not PCP-related), but were similar in atovaquone- and pentamidine-treated patients (16 vs 17% 8 weeks after treatment was discontinued) in another trial.

Patients enrolled in a few trials evaluating atovaquone in the treatment of cerebral toxoplasmosis also had HIV infection and, in most studies, were unresponsive to, or unable to tolerate, previous standard therapy for T. gondii infection. Atovaquone 750mg 4 times daily showed radiological efficacy in all identified trials (complete or partial response in 37 to 87.5% of patients), although most trials enrolled small numbers of patients (n = 5 to 24). In the largest trial, 52% of 87 patients achieved a complete or partial clinical response and 12% had stable disease after 6 weeks of therapy. In this study, 40% of patients had died at 18 weeks’ follow-up, 24% because of toxoplasmosis. No patients in the remaining trials were reported to have died from toxoplasmosis. Atovaquone has also shown promise as an agent for maintenance therapy in patients with cerebral toxoplasmosis and as a treatment for ocular toxoplasmosis.

Tolerability

Atovaquone tablets, most commonly administered at a dosage of 750mg 3 times daily, have a superior tolerability profile in patients with AIDS and PCP, compared with cotrimoxazole or pentamidine. The incidence of treatment-limiting adverse effects was lower with atovaquone than with cotrimoxazole (7 vs 20%) or pentamidine (4 vs 36%). Rash, nausea, diarrhoea, headache, vomiting, fever and insomnia occurred in 10 to 23% of patients receiving atovaquone; rash and liver dysfunction were treatment-limiting in 4 and 3% of patients, respectively. Laboratory abnormalities were observed in up to 10% of patients with PCP receiving atovaquone in comparative trials, although abnormal hepatocellular function or elevated amylase levels tended to occur more frequently in cotrimoxazole recipients.

In trials of atovaquone conducted in patients with toxoplasmosis, few adverse events were reported. Documented events included headache, nausea and diarrhoea. Rash or liver toxicity were treatment-limiting in a small number of patients. Toxic epidermal necrolysis also caused discontinuation of atovaquone therapy in 1 patient and erythema multiforme in another. In an unspecified number of patients, neutrophil and platelet counts and transaminase levels fluctuated in abnormal ranges, but changes were not severe or clearly attributable to atovaquone.

The tolerability profile of atovaquone suspension appears to be similar to that of the tablet formulation.

Dosage and Administration

The recommended oral dosage of atovaquone, when administered in tablet form for treatment of mild to moderate PCP, is 750mg 3 times daily for 21 days. The suspension should be taken twice daily at a dose of 750mg. Atovaquone must be administered with meals as failure to do so may reduce therapeutic efficacy; alternative therapy should be considered in patients unable to take the drug with meals and in those with malabsorption conditions and/or severe diarrhoea. Rifampicin (rifampin) and atovaquone should not be administered concomitantly, as subtherapeutic atovaquone concentrations may result. Atovaquone tablets 750mg 4 times daily for 42 days, followed by maintenance therapy, in most instances with the same dosage or with a 3 times daily regimen, have been used for treating patients with toxoplasmosis in clinical trials.

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

  1. Murray JF, Mills J. Pulmonary infectious complications of human immunodeficiency virus infection. Part II. Am Rev Respir Dis 1990; 141: 1582–98

    PubMed  CAS  Google Scholar 

  2. Rogers PL, Lane HC, Henderson DK, et al. Admission of AIDS patients to a medical intensive care unit: causes and outcome. Crit Care Med 1989; 17_(2): 113–7

    Article  Google Scholar 

  3. Wächter RM, Luce JM, Turner J, et al. Intensive care of patients with the acquired immunodeficiency syndrome. Outcome and changing patterns of utilization. Am Rev Respir Dis 1986; 134: 891–6

    PubMed  Google Scholar 

  4. Masur H, Meier P, McCutchan JA, et al. Consensus statement on the use of corticosteroids as adjunctive therapy for Pneumocystis pneumonia in the acquired immunodeficiency syndrome. N Engl J Med 1990; 323: 1500–4

    Article  Google Scholar 

  5. Zackrison LH, Tsou E. Pneumocystis carinni: a deadly opportunist. Am Fam Physician 1991; 44(2): 528–41

    PubMed  CAS  Google Scholar 

  6. Alberts B, Bray D, Lewis J, et al. Energy conversion: mitochondria and chloroplasts. In: Molecular biology of the cell. 2nd ed. New York: Garlan Publishing, 1989: 341–404

    Google Scholar 

  7. Anwar D, Hogan S, Thompson S, et al. Atovaquone (566C80) is effective treatment in AIDS patients with microsporidiosis [abstract 309]. In: Program and Abstracts of the 2nd National Conference on Human Retroviruses and Related Infections; 1995 Jan 29–Feb 2; Washington DC, 1995.

    Google Scholar 

  8. Comley JC, Mullin RJ, Wolfe LA, et al. A radiometric method for objectively screening large numbers of compounds against Pneumocystis carinii in vitro. J Protozool 1991 Nov–Dec; 38: 144S–6S

    PubMed  CAS  Google Scholar 

  9. Gutteridge WE. 566C80, an antimalarial hydroxynaphthoquinone with broad spectrum: experimental activity against opportunistic parasitic infections of AIDS patients. J Protozool 1991 Nov-Dec; 38: S141–3

    Google Scholar 

  10. Hughes WT, Gray VL, Gutteridge WE, et al. Efficacy of a hydroxynaphthoquinone, 566C80, in experimental Pneumocystis carinii pneumonitis. Antimicrob Agents Chemother 1990 Feb; 34: 225–8

    Article  PubMed  CAS  Google Scholar 

  11. Hughes WT. Limited effect of trimethoprim-sulfamethoxazole prophylaxis on Pneumocystis carinii. Antimicrob Agents Chemother 1979; 16(3): 333–5

    Article  PubMed  CAS  Google Scholar 

  12. Comley JCW, Sterling AM. Effect of atovaquone and atovaquone drug combinations on prophylaxis of Pneumocystis carinii pneumonia in SCID mice. Antimicrob Agents Chemother 1995; 39(4): 806–11

    Article  PubMed  CAS  Google Scholar 

  13. Hughes WT, Killmar JT, Oz HS. Relative potency of 10 drugs with anti-Pnuemocystis carinii activity in an animal model. J Infect Dis 1994; 170: 906–11

    Article  PubMed  CAS  Google Scholar 

  14. Brun-Pascaud M, Chau F, Simonpoli A-M, et al. Experimental evaluation of combined prophylaxis against murine pneumocystosis and toxoplasmosis. J Infect Dis 1994; 170: 653–8

    Article  PubMed  CAS  Google Scholar 

  15. Dohn MN, Frame PT, Baughman RP, et al. Open-label efficacy and safety trial of 42 days of 566C80 for Pneumocystis carinii pneumonia in AIDS patients. J Protozool 1991 Nov–Dec; 38: 220S–1S

    PubMed  CAS  Google Scholar 

  16. Colangelo G, Baughman RP, Dohn MN, et al. Follow-up bronchoalveolar lavage in AIDS patients with Pneumocystis carinii pneumonia. Pneumocystic carinii burden predicts early relapse. Am Rev Respir Dis 1991; 143: 1067–71

    PubMed  CAS  Google Scholar 

  17. Falloon J, Kovacs J, Hughes W, et al. A preliminary evaluation of 566C80 for the treatment of pneumocystis pneumonia in patients with the acquired immunodeficiency syndrome. N Engl J Med 1991 Nov 28; 325: 1534–8

    Article  PubMed  CAS  Google Scholar 

  18. Shelhamer JH, Ognibene FP, Macher AM, et al. Persistence of Pneumocystis carinii in lung tissue of acquired immunodeficiency syndrome patients treated for pneumocystis pneumonia. Am Rev Respir Dis 1984; 130: 1161–5

    PubMed  CAS  Google Scholar 

  19. Araujo FG, Huskinson J, Remington JS. Remarkable in vitro and in vivo activities of the hydroxynaphthoquinone 566C80 against tachyzoites and tissue cysts of Toxoplasma gondii. Antimicrob Agents Chemother 1991 Feb; 35: 293–9

    Article  PubMed  CAS  Google Scholar 

  20. Romand S, Pudney M, Derouin F. In vitro and in vivo activities of the hydroxynaphthoquinone atovaquone alone or combined with pyrimethamine, sulfadiazine, clarithromycin, or minocycline against Toxoplasma gondii. Antimicrob Agents Chemother 1993 Nov; 37: 2371–8

    Article  PubMed  CAS  Google Scholar 

  21. Araujo FG, Huskinson-Mark J, Gutteridge WE, et al. In vitro and in vivo activities of the hydroxynaphthoquinone 566C80 against the cyst form of Toxoplasma gondii. Antimicrob Agents Chemother 1992 Feb; 36: 326–30

    Article  PubMed  CAS  Google Scholar 

  22. Huskinson-Mark J, Araujo FG, Remington JS. Evaluation of the effect of drugs on the cyst form of Toxoplasma gondii. J Infect Dis 1991 Jul; 164: 170–7

    Article  PubMed  CAS  Google Scholar 

  23. Ferguson DJP, Huskinson-Mark J, Araujo FG, et al. An ultrastructural study of the effect of treatment with atovaquone in brains of mice chronically infected with the ME49 strain of Toxoplasma gondii. Int J Exp Pathol 1994 Apr; 75: 111–6

    PubMed  CAS  Google Scholar 

  24. Araujo FG, Lin T, Remington JS. The activity of atovaquone (566C80) in murine toxoplasmosis is markedly augmented when used in combination with pyrimethamine or sulfadiazine. J Infect Dis 1993 Feb; 167: 494–7

    Article  PubMed  CAS  Google Scholar 

  25. Fry M, Pudney M. Site of action of the antimalarial hydroxynaphthoquinone, 2-[trans-4- (4′-chlorophenyl) cyclohexyl]-3-hydroxy-1,4-naphthoquinone (566C80). Biochem Pharmacol 1992 Apr 1; 43: 1545–53

    Article  PubMed  CAS  Google Scholar 

  26. Seymour KK, Lyons SD, Phillips L, et al. Cytotoxic effects of inhibitors of de novo pyrimidine biosynthesis upon Plasmodiumfalciparum. Biochemistry 1994 May 3; 33: 5268–74

    Article  PubMed  CAS  Google Scholar 

  27. Pfefferkorn ER, Borotz SE, Nothnagel RF. Mutants of Toxoplasma gondii resistant to atovaquone (566C80) or decoquinate. J Parasitol 1993 Aug; 79: 559–64

    Article  PubMed  CAS  Google Scholar 

  28. Ittarat I, Asawamahasakda W, Meshnick SR. The effects of anti-malarials on the Plasmodium falciparum dihydroorotate de-hydrogenase. Exp Parasitol 1994 Aug; 79: 50–6

    Article  PubMed  CAS  Google Scholar 

  29. Ittarat I, Asawamahasakda W, Bartlett MS, et al. Effects of atovaquone and other inhibitors on Pneumocystis carinii dihydroorotate dehydrogenase. Antimicrob Agents Chemother 1995 Feb; 39(2): 325–8

    Article  PubMed  CAS  Google Scholar 

  30. Rolan PE, Mercer AJ, Weatherley BC, et al. Examination of some factors responsible for a food-induced increase in absorption of atovaquone. Br J Clin Pharmacol 1994 Jan; 37: 13–20

    Article  PubMed  CAS  Google Scholar 

  31. Hughes WT, Kennedy W, Shenep JL, et al. Safety and pharmacokinetics of 566C80, a hydroxynaphthoquinone with anti-Pneumocystis carinii activity: a phase I study in human immunodeficiency virus (HIV)-infected men. J Infect Dis 1991 Apr; 163: 843–8

    Article  PubMed  CAS  Google Scholar 

  32. Lavelle J, Trapnell C, Byrne R, et al. The absolute bioavailability of atovaquone tablets and suspension in HIV-seropositive volunteers [abstract PIII-43]. Clin Pharmacol Ther 1994; 55(2): 192

    Google Scholar 

  33. Wellcome. Mepron (atovaquone) suspension. England: Wellcome, 1995. Scientific brochure. (Data on file).

  34. Hughes W, Leoung G, Kramer F, et al. Comparison of atovaquone (566C80) with trimethoprim-sulfamethoxazole to treat Pneumocystis carinii pneumonia in patients with AIDS. N Engl J Med 1993 May 27; 328: 1521–7

    Article  PubMed  CAS  Google Scholar 

  35. Hudson AT, Dickins M, Ginger CD, et al. 566C80: a potent broad spectrum anti-infective agent with activity against malaria and opportunistic infections in AIDS patients. Drugs Exp Clin Res 1991; 17(9): 427–35

    PubMed  CAS  Google Scholar 

  36. Falloon J, Boenning C, Pagano G, et al. The pharmacokinetics of atovaquone suspension in patients with HIV infection [abstract 242]. In: Program and Abstracts of the 1st National Conference on Human Retroviruses and Related Infections; 1993 Dec 12–16; Washington DC, 1993.

    Google Scholar 

  37. A unique nonsulfonamide therapy for the treatment of PCP. An effective oral option for mild to moderate Pneumocystis carinii pneumonia in individuals who are intolerant to TMP-SMX. USA: Burroughs Wellcome Co, 1995. (Data on file).

  38. Dohn MN, Weinberg WG, Torres RA, et al. Oral atovaquone compared with intravenous pentamidine for Pneumocystis carinii pneumonia in patients with AIDS. Ann Intern Med 1994 Aug 1; 121: 174–80

    PubMed  CAS  Google Scholar 

  39. Torres RA, Weinberg W, Stansell J, et al. Atovaquone for salvage treatment and supression of toxoplasmic encephalitis in patients with acquired immunodeficiency syndrome (AIDS). England: Wellcome, 1995. (Data on file).

  40. Hughes WT, LaFon SW, Scott JD, et al. Adverse events associated with trimethoprim-sulfamethoxazole and atovaquone during the treatment of AIDS-related Pneumocystis carinii pneumonia. J Infect Dis 1995; 171: 1295–301

    Article  PubMed  CAS  Google Scholar 

  41. Sadler BM, Blum MR. Relationship between steady-state plasma concentrations of atovaquone (Css) and the use of various concomitant medications in AIDS patients with Pneumocystis carinii pneumonia [abstract PO-B31-2213]. IXth International Conference on AIDS/IVth STD World Congress; 1993 Jun 6–11; Berlin, Germany. 504.

    Google Scholar 

  42. Sadler BM, Caldwell P, Scott JD, et al. Drug interactions between rifampin and atovaquone (MEPRON) in HIV+ asymptomatic volunteers [abstract]. England: Wellcome, 1995. (Data on file).

  43. Lee BL, Täuber MG, Sadler B, et al. Atovaquone inhibits the glucuronidation and increases the serum concentrations of zidovudine [abstract]. In: 34th Interscience Conference on Antimicrobial Agents and Chemotherapy.: 1994: 81

  44. Hopkin JM, Wakefield AE. Epidemiology of Pneumocystis carinii infection: application of molecular genetic methods. Saudi Med J 1994; 15(1): 14–6

    Google Scholar 

  45. Hoover DR, Saah AJ, Bacellar H, et al. Clinical manifestations of AIDS in the era of pneumocystis prophylaxis. N Engl J Med 1993 Dec; 329: 1922–6

    Article  PubMed  CAS  Google Scholar 

  46. White A, Rogers M, Andrews E, et al. Results of a treatment IND for Mepron (atovaquone) theapy in patients with acute PCP [abstract WS-B14-1]. IXth International Conference on AIDS/IVth STD World Congress; 1993 Jun 6–11; Berlin, Germany.

    Google Scholar 

  47. Epstein LJ, Mohsenifar Z, Daar ES, et al. Clinical experience with atovaquone: a new drug for treating Pneumocystis carinii pneumonia. Am J Med Sci 1994 Jul; 308: 5–8

    Article  PubMed  CAS  Google Scholar 

  48. Simpson DM, Tagliati M. Neurologic manifestations of HIV infection. Ann Intern Med 1994; 121: 769–85

    PubMed  CAS  Google Scholar 

  49. Oksenhendler E, Charreau I, Tournerie C, et al. Toxoplasma gondii infection in advanced HIV infection. AIDS 1994; 8: 483–7

    Article  PubMed  CAS  Google Scholar 

  50. Luft BJ, Remington JS. Toxoplasmic encephalitis in AIDS. Clin Infect Dis 1992; 15: 211–22

    Article  PubMed  CAS  Google Scholar 

  51. Katlama C. New perspectives on the treatment and prophylaxis of Toxoplasma gondii infection. Curr Opin Infect Dis 1992; 5: 833–9

    Article  Google Scholar 

  52. Clumeck N, Katlama C, Ferrero T, et al. Atovaquone (1,4, —hydroxynaphtoquinone, 566C80) in the treatment of acute cerebral toxoplasmosis (CT) in AIDS patients (P) [abstract 1217]. 32nd Interscience Conference on Antimicrobial Agents and Chemotherapy: 1992 Oct 11–14; Anaheim, Caifornia, 313.

    Google Scholar 

  53. Grundman M, Torres RA, Thorn M, et al. Neuroradiologic response to 566C80 salvage therapy for CNS toxoplasmosis [poster B3185]. VII International Conference on AIDS. III STD World Congress, Amsterdam, The Netherlands, 19-24 July, 1992.

    Google Scholar 

  54. Kovacs JA, NIAID-Clinical CIAIDSP. Efficacy of atovaquone in treatment of toxoplasmosis in patients with AIDS. Lancet 1992 Sep 12; 340: 637–8

    Article  PubMed  CAS  Google Scholar 

  55. Lafeuillade A, Pellegrino P, Poggi C, et al. Efficacité de l’ atovaquone dans les toxoplasmoses résistantes du SIDA [letter]. Presse Med 1993 Oct 30; 22: 1708

    PubMed  CAS  Google Scholar 

  56. Katlama C, Mouthon B, Gourdon D, et al. Atovaquone (AT) as long term maintenance therapy (MT) for toxoplasmic encephalitis (TE) in AIDS [abstract]. England: Wellcome, 1995. (Data on file).

  57. Guelar A, Miró JM, Mallolas J, et al. Alternative therapeutic options for AIDS patients with cerebral toxoplasmosis: use of clarithromycin and atovaquone [in Spanish]. Enfermed Infec Micro Clin 1994 Mar; 12: 137–40

    CAS  Google Scholar 

  58. Kovacs JA, Polis MA, Baird B, et al. Evaluation of azithromycin or the combination of 566C80 and pyrimethamine in the treatment of toxoplasmosis [abstract PoB 3199]. VIII International Conference on AIDS/III STD World Congress; 1992 Jul 19–24; Amsterdam, The Netherlands, B120.

  59. Sheppard JD, Blick G, Caldwell PT, et al. Treatment of ocular toxoplasmosis with atovaquone, a cysticidal hydroxynaphthoquinone [abstract 785]. In: Program and Abstracts of the 1993 Conference of the Association for Research in Vision and Ophthalmology.

  60. Lopez JS, de Smet MD, Masur H, et al. Orally administered 566C80 for treatment of ocular toxoplasmosis in a patient with the acquired immunodeficiency syndrome [letter]. Am J Ophthalmol 1992 Mar 15; 113: 331–3

    PubMed  CAS  Google Scholar 

  61. Smith GH. Treatment of infections in the patient with acquired immunodeficiency syndrome. Arch Intern Med 1994 May 9; 154: 949–73

    Article  PubMed  CAS  Google Scholar 

  62. Gordin FM, Simon GL, Wofsy CB, et al. Adverse reactions to trimethoprim-sulfamethoxazole in patients with the acquired immunodeficiency syndrome. Ann Intern Med 1984; 100: 495–9

    PubMed  CAS  Google Scholar 

  63. Hughes WT. Prevention and treatment of Pneumocystis carinii pneumonia. Annu Rev Med 1991; 42: 287–95

    Article  PubMed  CAS  Google Scholar 

  64. Smith NA, Nelson MR, Gazzard BG, et al. The use of atovaquone in HIV positive patients [abstract]. AIDS 1994 Nov; 8 Suppl. 4: S37

    Article  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Additional information

Various sections of the manuscript reviewed by: F. Derouin, Laboratoire de Parasitologie-Mycologie, Hôpital Saint-Louis, Paris, France; M.N. Dohn, Holmes Division, University of Cincinnati Medical Center, Cincinnati, Ohio, USA; B.G. Gazzard, Chelsea & Westminster Hospital, London, England; W. Hughes, Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA; S.R. Meshnick, Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, Michigan, USA; R. Murphy, AIDS Clinical Treatment Unit, Section of Infectious Diseases, Northwestern University Medical School, Chicago, Illinois, USA; J.K. Rockstroh, Allgemeine Innere Medizin, Medizinische Universitätsklinik, Bonn, Germany; T. Sauerbruch, Allgemeine Innere Medizin, Medizinische Universitätsklinik, Bonn, Germany; M.M.E. Schneider, Department of Internal Medicine, Immunology & Infectious Diseases, University Hospital, Utrecht, The Netherlands; N. Smith, Department of Genito-Urinary Medicine, St Thomas’ Hospital Trust, London, England.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Spencer, C.M., Goa, K.L. Atovaquone. Drugs 50, 176–196 (1995). https://doi.org/10.2165/00003495-199550010-00011

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/00003495-199550010-00011

Keywords

Navigation