Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 130
Filtrar
1.
Hum Exp Toxicol ; 23(4): 173-85, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15171568

RESUMO

Haematopoietic suppression is one of the dose-limiting side effects of chronic zidovudine (AZT) therapy. We tested the hypothesis that AZT would reduce mitochondrial DNA (mtDNA) content in haematopoietic progenitors causing impaired haematopoiesis and mitochondrial dysfunction. We studied the effects of AZT 0-50 microM in vitro, on normal human CD34+ haematopoietic progenitor cells cultured ex vivo for up to 12 days. The mean AZT IC50 for granulocyte (phenotype CD15+/CD14-) and erythroid (phenotype glycophorin+/CD45-) cell proliferation was 2.5 microM (SD+/-0.7) and 0.023 microM (SD+/-0.005), respectively. In myeloid-rich cell cultures, the mean lactate content of the media, compared to untreated controls, increased by 86% (SD+/-23) at 10 microM AZT and in erythroid-rich cultures it increased by 134% (SD+/-24) in the presence of 0.5 microM AZT. In myeloid-rich cultures the AZT IC50 for the reduction in the mitochondrial/nuclear DNA content ratio was 5.6 microM, whereas in erythroid rich cultures this AZT IC50 was < 0.0005 microM. AZT produced concentration-dependent inhibition of CD34+ progenitor proliferation into both myeloid and erythroid lineages; erythropoiesis was more sensitive than myelopoiesis. Concurrently, AZT reduced steady state mtDNA content, while increasing lactate production. These findings support the hypothesis that mtDNA is one of the intracellular targets involved in the pathogenesis of AZT-associated bone marrow progenitor cell toxicity.


Assuntos
Fármacos Anti-HIV/toxicidade , Antígenos CD34/metabolismo , DNA Mitocondrial/antagonistas & inibidores , Células-Tronco Hematopoéticas/efeitos dos fármacos , Ácido Láctico/biossíntese , Zidovudina/toxicidade , Fármacos Anti-HIV/administração & dosagem , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , DNA Mitocondrial/biossíntese , Relação Dose-Resposta a Droga , Eritrócitos/efeitos dos fármacos , Eritrócitos/metabolismo , Granulócitos/efeitos dos fármacos , Granulócitos/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Humanos , Immunoblotting , Zidovudina/administração & dosagem
2.
Antimicrob Agents Chemother ; 45(10): 2733-9, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11557462

RESUMO

Tenofovir DF is an antiviral nucleotide with activity against human immunodeficiency virus type 1 (HIV-1). The pharmacokinetics, safety, and activity of oral tenofovir DF in HIV-1-infected adults were evaluated in a randomized, double-blind, placebo-controlled, escalating-dose study of four doses (75, 150, 300, and 600 mg given once daily). Subjects received a single dose of tenofovir DF or a placebo, followed by a 7-day washout period. Thereafter, subjects received their assigned study drug once daily for 28 days. Pharmacokinetic parameters were dose proportional and demonstrated no change with repeated dosing. Reductions in plasma HIV-1 RNA were dose related at tenofovir DF doses of 75 to 300 mg, but there was no increase in virus suppression between the 300- and 600-mg dose cohorts, despite dose-proportional increases in drug exposure. Grade III or IV adverse events were limited to laboratory abnormalities, including elevated creatine phosphokinase and liver function tests, which resolved with or without drug discontinuation and without sequelae. No patients developed detectable sequence changes in the reverse transcriptase gene.


Assuntos
Adenina/análogos & derivados , Adenina/farmacocinética , Fármacos Anti-HIV/farmacocinética , Infecções por HIV/metabolismo , Organofosfonatos , Compostos Organofosforados/farmacocinética , Adenina/efeitos adversos , Adulto , Fármacos Anti-HIV/efeitos adversos , Método Duplo-Cego , Tolerância a Medicamentos , Feminino , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Compostos Organofosforados/efeitos adversos , RNA Viral/sangue , RNA Viral/efeitos dos fármacos , Tenofovir
3.
Antimicrob Agents Chemother ; 43(2): 271-7, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9925517

RESUMO

The pharmacokinetics and bioavailability of 1-[((S)-2-hydroxy-2-oxo-1,4,2-dioxaphosphorinan-5-yl)methyl]cytosi ne (cyclic HPMPC) were examined at four doses in 22 patients with human immunodeficiency virus infection. Two groups of six patients received a single dose of cyclic HPMPC at 1.5 or 3.0 mg/kg of body weight by each of the oral and intravenous routes in a random order with a 2-week washout period between administrations. Additional patients received single intravenous doses of cyclic HPMPC at 5.0 mg/kg (n = 6) or 7.5 mg/kg (n = 4). Serial serum and urine samples were collected at intervals over 24 h after dosing. The concentrations of cyclic HPMPC and cidofovir in serum and urine samples were determined by validated reverse-phase ion-pairing high-performance liquid chromatography methods with derivatization and fluorescence detection. After intravenous administration of cyclic HPMPC, concentrations of cyclic HPMPC declined in a biexponential manner, with a mean +/- standard deviation half-life of 1.09 +/- 0.12 h (n = 22). The pharmacokinetics of cyclic HPMPC were independent of dose over the dose range of 1.5 to 7.5 mg/kg. The total clearance of cyclic HPMPC from serum and the volume of distribution of intravenous cyclic HPMPC were 198 +/- 39.6 ml/h/kg and 338 +/- 65.1 ml/kg, respectively (n = 22). The renal clearance of cyclic HPMPC (132 +/- 27.3 ml/h/kg; n = 22) exceeded the creatinine clearance (86.2 +/- 16.3 ml/h/kg), indicating active tubular secretion. The cyclic HPMPC excreted in urine in 24 h accounted for 71.3% +/- 16.0% of the administered dose. Cidofovir was formed from cyclic HPMPC in vivo with a time to the maximum concentration in serum of 1.64 +/- 0.23 h (n = 22). Cidofovir levels declined in an apparent monoexponential manner, with a mean terminal half-life of 3.98 +/- 1.26 h (n = 22). The cidofovir excreted in urine in 24 h accounted for 9.40% +/- 2.33% of the administered cyclic HPMPC dose. Exposure to cidofovir after intravenous administration of cyclic HPMPC was dose proportional and was 14.9% of that from an equivalent dose of cidofovir. The present study suggests that intravenous cyclic HPMPC also has a lower potential for nephrotoxicity in humans compared to that of intravenous cidofovir. The oral bioavailabilities of cyclic HPMPC were 1.76% +/- 1.48% and 3.10% +/- 1.16% with the administration of doses of 1.5 and 3.0 mg/kg, respectively (n = 6 per dose). The maximum concentrations of cyclic HPMPC in serum were 0.036 +/- 0.021 and 0.082 +/- 0.038 microgram/ml after the oral administration of doses of 1.5 and 3.0 mg/kg, respectively. Cidofovir reached quantifiable levels in the serum of only one patient for each of the 1.5- and 3.0-mg/kg oral cyclic HPMPC doses.


Assuntos
Fármacos Anti-HIV/farmacocinética , Citosina/análogos & derivados , Infecções por HIV/metabolismo , Organofosfonatos , Compostos Organofosforados/farmacocinética , Administração Oral , Adulto , Fármacos Anti-HIV/sangue , Fármacos Anti-HIV/metabolismo , Disponibilidade Biológica , Cidofovir , Citosina/sangue , Citosina/metabolismo , Citosina/farmacocinética , Feminino , Humanos , Injeções Intravenosas , Masculino , Pessoa de Meia-Idade , Compostos Organofosforados/sangue , Compostos Organofosforados/metabolismo , Pró-Fármacos/farmacocinética
4.
Antiviral Res ; 38(3): 209-12, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9754889

RESUMO

For systemic use, the anti-cytomegalovirus (CMV) agent foscarnet must be given intravenously because oral administration results in unmeasurable or barely measurable plasma levels. At low pH, foscarnet decomposes via an acid-catalyzed decarboxylation; therefore, poor oral bioavailability might be due to decomposition of foscarnet in gastric acid. We evaluated whether increasing gastric pH with ranitidine would enhance the absorption of oral foscarnet in six asymptomatic HIV-infected individuals. Each volunteer received two oral 4000-mg (60 mg/kg) doses of foscarnet, preceded intravenously by a 20-min infusion of either ranitidine 50 mg in D5W or D5W alone in a randomized, double-blind, cross-over study. Intragastric pH monitoring revealed that subjects had evidence of gastric acid production (pH < 2.0) prior to administration of ranitidine and increased gastric pH (pH > 6.0) following ranitidine administration. Most foscarnet plasma levels were below the assay limit of detection (33 microM) with only 4/30 levels detectable after D5W and 8/30 after ranitidine. Urinary recovery of foscarnet increased after ranitidine pretreatment. A mean recovery of 9.9% of the drug was realized in the urine in 24 h following ranitidine pretreatment compared to 6.2% of the dose after D5W pretreatment (P < 0.03). We estimate that 9.9% recovery in the urine in 24 h is equivalent to absorption of 17.1% of the oral dose. In spite of the enhanced bioavailability associated with ranitidine pretreatment, the degree of absorption is still insufficient to achieve effective plasma concentrations for the treatment of CMV or acyclovir-resistant herpes viruses. We conclude that gastric acidity is a determinant of foscarnet absorption, albeit not a major one. Oral foscarnet is unlikely to be clinically useful even if administered in the setting of increased gastric pH.


Assuntos
Antivirais/administração & dosagem , Foscarnet/administração & dosagem , Ácido Gástrico/química , Infecções por HIV/metabolismo , Administração Oral , Antivirais/sangue , Antivirais/farmacocinética , Antivirais/urina , Disponibilidade Biológica , Cromatografia Líquida de Alta Pressão , Estudos Cross-Over , Método Duplo-Cego , Foscarnet/sangue , Foscarnet/farmacocinética , Foscarnet/urina , Soropositividade para HIV , Antagonistas dos Receptores H2 da Histamina/farmacologia , Humanos , Concentração de Íons de Hidrogênio , Ranitidina/farmacologia
5.
Antimicrob Agents Chemother ; 42(9): 2380-4, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9736567

RESUMO

9-[2-(R)-(Phosphonomethoxy)propyl]adenine (PMPA) is a nucleotide analogue with potent antiretroviral activity in vitro and in simian models. A randomized, double-blind, placebo-controlled, dose-escalation clinical trial of intravenous PMPA monotherapy was conducted in 20 human immunodeficiency virus (HIV)-infected adults with CD4 cell counts of >/=200 cells/mm3 and plasma HIV RNA levels of >/=10,000 copies/ml. Two dose levels were evaluated (1 and 3 mg/kg of body weight/day). Ten subjects were enrolled at each dose level (eight randomized to receive PMPA and two randomized to receive placebo). On day 1, a single dose of PMPA or placebo was administered by intravenous infusion. Beginning on study day 8, PMPA or placebo was administered once daily for an additional 7 consecutive days. All subjects tolerated dosing without significant adverse events. Mean peak serum PMPA concentrations were 2.7 +/- 0.9 and 9.1 +/- 2.1 microgram/ml in the 1- and 3-mg/kg cohorts, respectively. Serum concentrations declined in a biexponential fashion, with a terminal half-life of 4 to 8 h. At 3 mg/kg/day, a single infusion of PMPA resulted in a 0.4 log10 median decline in plasma HIV RNA by study day 8. Following 7 consecutive days of study drug administration thereafter, the median changes in plasma HIV RNA from baseline were -1.1, -0.6, and 0.1 log10 in the 3-mg/kg/day, 1-mg/kg/day, and placebo dose groups, respectively. Following the final dose in the 3-mg/kg/day cohort, the reduction in HIV RNA was sustained for 7 days before returning toward baseline. Further studies evaluating an oral prodrug of PMPA are under way.


Assuntos
Adenina/análogos & derivados , Antivirais/uso terapêutico , Infecções por HIV/tratamento farmacológico , Organofosfonatos , Compostos Organofosforados/uso terapêutico , Adenina/efeitos adversos , Adenina/farmacocinética , Adenina/uso terapêutico , Adolescente , Adulto , Método Duplo-Cego , Feminino , Infecções por HIV/imunologia , Infecções por HIV/virologia , Humanos , Masculino , Pessoa de Meia-Idade , Compostos Organofosforados/efeitos adversos , Compostos Organofosforados/farmacocinética , RNA Viral/sangue , Linfócitos T/imunologia , Tenofovir
6.
AIDS ; 12(9): F79-83, 1998 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-9662190

RESUMO

BACKGROUND: Simple and affordable intervention strategies are needed to reduce the rate of HIV transmission from mother to infant in developing countries. Simian immunodeficiency virus (SIV) infection of newborn rhesus macaques is considered to be a useful model of human pediatric HIV infection. OBJECTIVE: To investigate whether short-term 9-[2-(phosphonomethoxy)propyl]adenine (PMPA) administration can protect newborn rhesus macaques against perinatal SIV infection. DESIGN AND METHODS: Eight newborn macaques were inoculated orally with highly virulent SIVmac within the first 3 days of life. Four of these animals were untreated controls. The other four animals were given one dose of PMPA (30 mg/kg subcutaneously) 4 h before oral SIV inoculation, and were then given a second and final dose of PMPA 24 h later. RESULTS: All four untreated control animals were persistently SIV-positive within 2 weeks after virus inoculation. In contrast, no virus could be detected in the four animals that received two doses of PMPA; these animals were seronegative and healthy at 10 months. CONCLUSIONS: Two doses of PMPA prevented SIV infection of newborn macaques. Our data suggest that short-term administration of PMPA to HIV-infected pregnant women at the onset of labor and to their newborns after delivery may reduce the rate of intrapartum HIV transmission.


Assuntos
Adenina/análogos & derivados , Fármacos Anti-HIV/administração & dosagem , Organofosfonatos , Compostos Organofosforados/administração & dosagem , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Vírus da Imunodeficiência Símia , Adenina/administração & dosagem , Animais , Animais Recém-Nascidos , Anticorpos Antivirais/sangue , Relação Dose-Resposta a Droga , Macaca mulatta , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/genética , Vírus da Imunodeficiência Símia/imunologia , Tenofovir , Viremia
7.
J Infect Dis ; 176(2): 406-13, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9237705

RESUMO

A randomized, double-blind, placebo-controlled, dose-escalation study of adefovir dipivoxil, an oral prodrug of adefovir, was conducted in 36 human immunodeficiency virus (HIV)-infected subjects to evaluate its anti-HIV activity, safety, and pharmacokinetics. Subjects received placebo or one of three dosages of adefovir dipivoxil daily for 14 days. Median decreases in serum p24 antigen of 31% (P = .02), 25% (P = .31), and 30% (P = .01) occurred in each drug-treated group, respectively, compared with an increase of 17% in the placebo group. Median decreases in serum HIV RNA of 0.4-0.6 log10 copies/mL occurred in the drug-treated groups (P = .03), compared with no change in the placebo group. Gastrointestinal complaints and reversible liver transaminase elevations were the most frequently noted adverse events. Decreases in serum free carnitine occurred in each drug-treated group during treatment. After 14 days of dosing, adefovir dipivoxil demonstrated anti-HIV activity and was best tolerated at the lowest dosage studied, 125 mg daily.


Assuntos
Adenina/análogos & derivados , Fármacos Anti-HIV/uso terapêutico , Infecções por HIV/tratamento farmacológico , Organofosfonatos , Adenina/efeitos adversos , Adenina/farmacocinética , Adenina/uso terapêutico , Adulto , Fármacos Anti-HIV/efeitos adversos , Fármacos Anti-HIV/farmacocinética , Carnitina/sangue , Sistema Digestório/efeitos dos fármacos , Método Duplo-Cego , Feminino , Proteína do Núcleo p24 do HIV/sangue , Humanos , Masculino , Pessoa de Meia-Idade , RNA Viral/sangue
9.
Am J Med ; 101(1): 68-76, 1996 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8686718

RESUMO

PURPOSE: To compare the efficacy and safety of inpatient oral antibiotic treatment (oral) versus standard parenteral antibiotic treatment (intravenous) for right-sided staphylococcal endocarditis in injection drug users. PATIENTS AND METHODS: In a prospective, randomized, non-blinded trial, febrile injection drug users were assigned to begin oral or intravenous (IV) treatment on admission, before blood culture results were available. Oral therapy consisted of ciprofloxacin and rifampin. Parenteral therapy was oxacillin or vancomycin, plus gentamicin for the first 5 days. Antibiotic dosing was adjusted for renal dysfunction. Administration of other antibacterial drugs was not permitted during the treatment or follow-up periods. Bacteremic subjects having right-sided staphylococcal endocarditis received 28 days of inpatient therapy with the assigned antibiotics. Test-of-cure blood cultures were obtained during inpatient observation 6 and 7 days after the completion of antibiotic therapy, and again at outpatient follow-up 1 month later. Criteria for treatment failure and for drug toxicity were prospectively defined. RESULTS: Of 573 injection drug users who were hospitalized because of a febrile illness and suspected right-sided staphylococcal endocarditis, 93 subjects (16.2%) had two or more sets of blood cultures positive for staphylococci; 85 of these bacteremic subjects (14.8%) satisfied diagnostic criteria for at least possible right-sided staphylococcal endocarditis (no other source of bacteremia was apparent) and entered the trial. Forty-four (oral, 19; IV, 25) of these 85 subjects completed inpatient treatment and evaluation including test-of-cure blood cultures. There were four treatment failures (oral, 1 [5.2%]; IV, 3 [12.0%]; not significant, Fisher's exact test). Drug toxicity was significantly more common in the parenterally treated group (oral, 3%; IV, 62%; P < 0.0001), consisting largely of oxacillin-associated increases in liver enzymes. CONCLUSIONS: For selected patients with right-sided staphylococcal endocarditis, oral ciprofloxacin plus rifampin is effective and is associated with less drug toxicity than is intravenous therapy.


Assuntos
Anti-Infecciosos/administração & dosagem , Endocardite Bacteriana/tratamento farmacológico , Infecções Estafilocócicas/tratamento farmacológico , Abuso de Substâncias por Via Intravenosa/complicações , Administração Oral , Adulto , Antibacterianos/administração & dosagem , Antibióticos Antituberculose/administração & dosagem , Ciprofloxacina/administração & dosagem , Endocardite Bacteriana/etiologia , Endocardite Bacteriana/mortalidade , Feminino , Gentamicinas/administração & dosagem , Humanos , Infusões Intravenosas , Tempo de Internação , Masculino , Oxacilina/administração & dosagem , Penicilinas/administração & dosagem , Estudos Prospectivos , Rifampina/administração & dosagem , Infecções Estafilocócicas/etiologia , Infecções Estafilocócicas/mortalidade , Resultado do Tratamento , Vancomicina/administração & dosagem
10.
Antiviral Res ; 29(2-3): 153-61, 1996 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8739595

RESUMO

We conducted a single-center, double-blind, placebo-controlled phase I study in HIV-positive subjects to ascertain the safety, tolerance, bioavailability, pharmacokinetics, and maximum tolerated dose of HPMPC (cidofovir). Five subjects were randomized to receive drug and two to receive placebo at each of three dosage tier (1, 3, and 10 mg/kg) with a 2-week washout period doses. Subjects at 1 and 3 mg/kg received single doses of HPMPC by subcutaneous (s.c.) intravenous (i.v.), and oral (p.o.) routes, while subjects at 10 mg/kg received only i.v. and p.o. doses. For subjects already taking zidovudine, zidovudine AUC values are determined before and then with HPMPC administration for each route. The AUC values of HPMPC were dose-proportional. Subcutaneous bioavailability was essentially equivalent to that of the intravenous route, but the development of transient local fibrosis ad the volumes needed for subcutaneous dosing precluded higher subcutaneous dosing than 3 mg/kg. Oral bioavailability was poor, estimated to be less than 5%. Drug elimination was predominantly renal. Nephrotoxicity in one subject was the only serious adverse event observed. This subject had a significant lag period prior to oral absorption and also had the highest AUC values for both HPMPC and zidovudine. We found no consistent effect on zidovudine AUC concomitant HPMPC.


Assuntos
Antivirais/farmacocinética , Citosina/análogos & derivados , Infecções por HIV/tratamento farmacológico , Organofosfonatos , Compostos Organofosforados/farmacocinética , Adolescente , Adulto , Antivirais/efeitos adversos , Disponibilidade Biológica , Cidofovir , Citosina/efeitos adversos , Citosina/farmacocinética , Método Duplo-Cego , Vias de Administração de Medicamentos , Feminino , Infecções por HIV/metabolismo , Infecções por HIV/urina , Humanos , Masculino , Pessoa de Meia-Idade , Compostos Organofosforados/efeitos adversos , Zidovudina/farmacocinética
11.
Antimicrob Agents Chemother ; 40(2): 433-6, 1996 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8834893

RESUMO

We extend the observation that inhibitors of pyrimidine biosynthesis are active against human cytomegalovirus by demonstrating that methotrexate (MTX) has preferential activity against cytomegalovirus replication. The 50% and 90% inhibitory concentrations of MTX for inhibition of cytomegaloviral DNA replication at 3 days postinfection in MRC-5 cells were 0.05 and 0.2 microM, respectively. No cell toxicity was observed in uninfected confluent cells at the highest concentration tested (1 microM). Under similar conditions (3 days of treatment with 0.2 microM MTX), intracellular dTTP pools were diminished in cytomegalovirus-infected cells (87% decrease relative to untreated infected cells, P < 0.001) but were not reduced in uninfected cells. A potential explanation for the preferential antiviral effect of MTX was that human cytomegalovirus-infected cells preferentially accumulated MTX. Increased intracellular accumulation and increased polyglutamation of MTX were observed in cytomegalovirus-infected cells compared with uninfected cells. Increased uptake of [3H]MTX by cytomegalovirus-infected cells was first observed at 48 h postinfection, with threefold-higher accumulation within infected cells. By 96 h, accumulation had increased to approximately fourfold in comparison with uninfected cells. The uptake of [3H]MTX was saturable and was blocked by addition of unlabelled MTX. Intracellular MTX in infected cells was almost entirely in the polyglutamated form, as demonstrated by thin-layer chromatography, whereas intracellular MTX was almost exclusively in the parent form in uninfected cells.


Assuntos
Antivirais/farmacologia , Citomegalovirus/efeitos dos fármacos , Antagonistas do Ácido Fólico/farmacologia , Metotrexato/farmacologia , Replicação Viral/efeitos dos fármacos , Linhagem Celular , Cromatografia em Camada Fina , Citomegalovirus/metabolismo , Relação Dose-Resposta a Droga , Fibroblastos , Humanos , Pulmão/metabolismo , Metotrexato/metabolismo , Nucleotídeos de Timina/metabolismo
13.
Antiviral Res ; 28(2): 121-31, 1995 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8585766

RESUMO

The magnitude and duration of the antiviral and clinical effect of alpha-interferon was measured in healthy volunteers. A single 3 million unit intramuscular dose of interferon was given either alone (controls) or after 72 h of concomitant medications. These medications included either aspirin (650 mg every 4 h), acetaminophen (650 mg every 4 h), or prednisone (40 mg per day). Peripheral blood mononuclear cells were assayed for resistance to vesicular stomatitis virus infection and induction of 2'-5'-oligoadenylate synthetase activity as evidence of interferon's antiviral effect. Co-administration of acetaminophen increased both antiviral parameters by more than 70% (P < 0.05) and reduced symptoms after interferon dosing, compared to controls. Aspirin and prednisone did not demonstrate any significant differences from controls in antiviral effect. As a group, acetaminophen, aspirin, and prednisone reduced the clinical symptoms by 47% compared to controls (P = 0.03) after interferon dosing, although individual drug comparisons failed to reach statistical significance. Independent of treatment group, the changes in antiviral markers after interferon dosing correlated closely with each other (r = 0.72, P < 0.001), but neither correlated with symptoms or fever (r < 0.30, P > 0.05). Acetaminophen enhances the antiviral effects of a single intramuscular dose of alpha-interferon, considering the parameters measured in these healthy volunteers.


Assuntos
Acetaminofen/farmacologia , Antivirais/farmacologia , Aspirina/farmacologia , Interferon-alfa/farmacologia , Prednisona/farmacologia , Infecções por Rhabdoviridae/tratamento farmacológico , Vírus da Estomatite Vesicular Indiana/efeitos dos fármacos , 2',5'-Oligoadenilato Sintetase/biossíntese , Adulto , Sinergismo Farmacológico , Humanos , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/enzimologia , Fatores de Tempo
14.
Antimicrob Agents Chemother ; 39(6): 1247-52, 1995 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7574510

RESUMO

The pharmacokinetics of cidofovir (HPMPC; (S)-1-[3-hydroxy-2-(phosphonylmethoxy)propyl]cytosine) were examined at five dose levels in three phase I/II studies in a total of 42 human immunodeficiency virus-infected patients (with or without asymptomatic cytomegalovirus infection). Levels of cidofovir in serum following intravenous infusion were dose proportional over the dose range of 1.0 to 10.0 mg/kg of body weight and declined biexponentially with an overall mean +/- standard deviation terminal half-life of 2.6 +/- 1.2 h (n = 25). Approximately 90% of the intravenous dose was recovered unchanged in the urine in 24 h. The overall mean +/- standard deviation total clearance of the drug from serum (148 +/- 25 ml/h/kg; n = 25) approximated renal clearance (129 +/- 42 ml/h/kg; n = 25), which was significantly higher (P < 0.001) than the baseline creatinine clearance in the same patients (83 +/- 21 ml/h/kg; n = 12). These data indicate that active tubular secretion played a significant role in the clearance of cidofovir. The steady-state volume of distribution of cidofovir was approximately 500 ml/kg, suggesting that the drug was distributed in total body water. Repeated dosing with cidofovir at 3.0 and 10.0 mg/kg/week did not alter the pharmacokinetics of the drug. Concomitant administration of intravenous cidofovir and oral probenecid to hydrated patients had no significant effect on the pharmacokinetics of cidofovir at a 3.0-mg/kg dose. At higher cidofovir doses, probenecid appeared to block tubular secretion of cidofovir and reduce its renal clearance to a level approaching glomerular filtration.


Assuntos
Antivirais/farmacocinética , Citosina/análogos & derivados , Infecções por HIV/metabolismo , Hospedeiro Imunocomprometido , Organofosfonatos , Compostos Organofosforados/farmacocinética , Infecções Oportunistas Relacionadas com a AIDS/complicações , Infecções Oportunistas Relacionadas com a AIDS/tratamento farmacológico , Infecções Oportunistas Relacionadas com a AIDS/metabolismo , Adulto , Antivirais/administração & dosagem , Antivirais/análise , Cidofovir , Infecções por Citomegalovirus/complicações , Infecções por Citomegalovirus/tratamento farmacológico , Infecções por Citomegalovirus/metabolismo , Citosina/administração & dosagem , Citosina/análise , Citosina/farmacocinética , Relação Dose-Resposta a Droga , Quimioterapia Combinada , Feminino , Infecções por HIV/complicações , Infecções por HIV/tratamento farmacológico , Humanos , Rim/metabolismo , Testes de Função Renal , Masculino , Taxa de Depuração Metabólica , Pessoa de Meia-Idade , Compostos Organofosforados/administração & dosagem , Compostos Organofosforados/análise , Probenecid/administração & dosagem , Probenecid/farmacologia
15.
Drugs ; 49 Suppl 2: 159-63, 1995.
Artigo em Inglês | MEDLINE | ID: mdl-8549287

RESUMO

The main types of adverse effects associated with quinolones are uncommon and reversible and vary in frequency among different agents. Phototoxicity appears more frequent with lomefloxacin than with some other quinolones. Three mechanisms have been proposed to explain the neurotoxic effects, including rare proconvulsant activity, associated with quinolone therapy. Arthropathy remains a dilemma for paediatricians deciding whether to use quinolones in growing children. Importantly, the experience with temafloxacin, which has now been withdrawn from the market, emphasises the need for thorough postmarketing surveillance. Nonetheless, it should be remembered that the fluoroquinolones as a group are effective and very well tolerated antimicrobial drugs.


Assuntos
Anti-Infecciosos/efeitos adversos , Fluoroquinolonas , Dermatite Fototóxica/patologia , Humanos , Artropatias/induzido quimicamente , Doenças do Sistema Nervoso/induzido quimicamente , Quinolonas/efeitos adversos
16.
Antimicrob Agents Chemother ; 38(10): 2404-8, 1994 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7840578

RESUMO

HPMPC [(S)-1-(3-hydroxy-2-phosphonylmethoxypropyl)cytosine] is a potent inhibitor of human cytomegalovirus (HCMV) replication as determined by conventional tissue culture methods in which the drug concentration remains constant over time. Previous studies have shown HPMPC to have a long intracellular half-life. Despite its relatively short extracellular half-life, HPMPC might provide significant anti-HCMV activity long after the elimination of the drug by first-order kinetics. We addressed this hypothesis by measuring the activity of HPMPC in a novel cell culture perfusion system. This system allows us to compare the activity of HPMPC when given as a continuous infusion with its activity when given as a single-bolus dose followed by elimination that simulates the drug's in vivo pharmacokinetics. We show that continuous infusions maintaining maximum concentrations (Cmaxs) of 0.05, 0.10, 0.31, and 1.0 micrograms/ml and achieving areas under the drug concentration-time curves (AUCs) of 8.4, 17, 50, and 162 micrograms.h/ml, respectively, result in 27, 56, 63, and 88% inhibition of viral DNA accumulation, respectively, compared with an untreated control. Single-bolus doses achieving Cmaxs of 0.10, 1.25, 3.0, and 7.7 micrograms/ml with an elimination half-life of 20 h achieved AUCs of 2.4, 32, 78, and 138 micrograms.h/ml and resulted in 0, 48, 69, and 87% inhibition of HCMV DNA accumulation. Single-bolus doses achieving Cmaxs of 3.9 and 12 micrograms/ml with an elimination half-life of 6.5 h achieved AUCs of 34 and 105 micrograms.h/ml, respectively, resulting in 15 and 76% inhibition of viral DNA accumulation. Comparison of Cmax-versus-effect curves for these three regimens suggests that maximum concentration is not the only important pharmacokinetic determinant of HPMPC's antiviral activity. Similar comparisons of AUC-versus-effect curves for continuous and bolus dosing suggest that the AUC is an important determinant of antiviral activity for AUCs greater than 100 micrograms . h/ml. We conclude that single-bolus doses of HPMPC potently inhibit HCMV DNA accumulation but that this activity is more heavily influenced by the AUC than the Cmax at the upper end of the AUC range tested. At lower AUCs, some other parameter may be the primary determinant of antiviral activity. Our cell culture perfusion system provides a novel, efficient, and convenient method for addressing questions relating the effects of constantly changing drug concentrations to antiviral effects.


Assuntos
Antivirais/farmacologia , Citomegalovirus/efeitos dos fármacos , Citosina/análogos & derivados , Organofosfonatos , Compostos Organofosforados/farmacologia , Células Cultivadas , Cidofovir , Citosina/administração & dosagem , Citosina/farmacocinética , Citosina/farmacologia , Meia-Vida , Humanos , Compostos Organofosforados/administração & dosagem , Compostos Organofosforados/farmacocinética
17.
Antimicrob Agents Chemother ; 38(5): 1036-40, 1994 May.
Artigo em Inglês | MEDLINE | ID: mdl-8067734

RESUMO

Zidovudine delays the progression of infection and prolongs the survival of human immunodeficiency virus (HIV)-infected patients, but these benefits are limited by dose-related toxicity and the cost of the drug. Dipyridamole, in micromolar concentrations, acts synergistically with zidovudine, reducing the anti-HIV 95% inhibitory concentration of zidovudine 5- to 10-fold in vitro. We sought to establish a well-tolerated dose of dipyridamole for use in combination with zidovudine and to detect clinically significant pharmacokinetic interactions. Both objectives are essential for planning studies of the efficacy of the zidovudine-dipyridamole combination. Eleven asymptomatic HIV-infected subjects (median CD4+ cell count, 311 cells per mm3), 10 of whom had been on zidovudine at 500 mg/day for at least 6 months, were admitted to the study. Zidovudine pharmacokinetics were measured on day 1. Dipyridamole was then begun at 600 mg/day (subjects 1 to 3) or 450 mg/day (subjects 4 to 11), and zidovudine and dipyridamole pharmacokinetics were measured on day 5. All subjects given 600 mg of dipyridamole per day developed headache or nausea, or both. Six of eight subjects given dipyridamole at 450 mg/day developed headache or mild nausea that resolved after a median of 2 days. The area under the zidovudine concentration-time curve was not significantly different on day 1 in comparison with that on day 5 (P = 0.11). Symptoms were significantly correlated with the maximum zidovudine concentrations, which were achieved when dipyridamole was dosed concomitantly (p = 0.03). Total (free and protein-bound) dipyridamole trough concentrations were near those demonstrating synergy with zidovudine against HIV in vitro. Dipyridamole was highly protein bound, with a median free/total dipyridamole ratio of 0.7%; the percent free/total dipyridamole ratio was inversely correlated with alpha 1 acid glycoprotein concentrations (r2 =0.66). Results of the study indicate that adjustment of the zidovudine dose was not required to achieve equivalent zidovudine concentrations when zidovudine was administered in combination with dipyridamole at the doses studied. In the short study described here, the zidovudine-dipyridamole combinations was well tolerated in asymptomatic HIV-infected subjects after the occurrence of mild transient symptoms.


Assuntos
Dipiridamol/farmacologia , Soropositividade para HIV/metabolismo , Zidovudina/efeitos adversos , Zidovudina/farmacocinética , Adulto , Dipiridamol/efeitos adversos , Dipiridamol/sangue , Combinação de Medicamentos , Interações Medicamentosas , Soropositividade para HIV/complicações , Humanos , Radioisótopos do Iodo , Masculino , Orosomucoide/metabolismo
18.
Clin Infect Dis ; 18(4): 533-43, 1994 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8038306

RESUMO

Although monitoring serum vancomycin concentrations in clinical practice is commonplace, the data supporting this practice are meager. The rationale for monitoring these concentrations is to improve the effectiveness and/or reduce the toxicity of the drug. However, there are no data to suggest that monitoring serum vancomycin concentrations improves the effectiveness of therapy. In addition, despite many case reports of vancomycin-associated nephrotoxicity and ototoxicity, it is unclear whether this agent truly causes such conditions. Moreover, there is no evidence that adherence to specific ranges of vancomycin concentrations will preclude these events. Finally, vancomycin pharmacokinetics are sufficiently predictable that adequate serum drug concentrations can be obtained with dosing methods that take into account the patient's age, weight, and renal function. Safe and effective vancomycin dosage regimens can be constructed with these empirical dosing methods, whereas monitoring vancomycin levels increases the cost of therapy without improving the safety or efficacy of treatment.


Assuntos
Vancomicina/sangue , Infecções Bacterianas/sangue , Infecções Bacterianas/tratamento farmacológico , Ensaios Clínicos como Assunto , Perda Auditiva/induzido quimicamente , Humanos , Rim/efeitos dos fármacos , Vancomicina/administração & dosagem , Vancomicina/efeitos adversos
19.
Antimicrob Agents Chemother ; 37(3): 602-4, 1993 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8384821

RESUMO

We studied the effects of 2-acetylpyridine 5-[(dimethylamino)thiocarbonyl]-thiocarbonohydrazone (1110U81 or A1110U), a potent inhibitor of the ribonucleotide reductases encoded by herpes simplex virus types 1 and 2 and by varicella-zoster virus, against human cytomegalovirus (HCMV) replication in infected MRC-5 cells. We show that 1110U81 is a potent inhibitor of HCMV DNA replication (50% inhibitory concentration [IC50], 3.6 microM; IC90, 5.6 microM) and also potentiates the effects of ganciclovir (GCV) against HCMV. The IC90 of GCV is reduced from 65 microM when GCV alone is given to 2.8 microM when GCV is combined with 1110U81 at a molar ratio of 1:1.


Assuntos
Antivirais/farmacologia , Citomegalovirus/efeitos dos fármacos , Ganciclovir/farmacologia , Hidrazonas/farmacologia , Piridinas/farmacologia , Replicação Viral/efeitos dos fármacos , Células Cultivadas , Citomegalovirus/fisiologia , Replicação do DNA/efeitos dos fármacos , DNA Viral/biossíntese , Sinergismo Farmacológico , Humanos , Hibridização de Ácido Nucleico
20.
Antimicrob Agents Chemother ; 37(3): 429-35, 1993 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7681656

RESUMO

Poly(I):poly(C12U) (mismatched double-stranded RNA; atvogen), an interferon inducer, is active against human immunodeficiency virus in vitro. To determine the extent and duration of the biologic effects of poly(I):poly(C12U), we administered a single dose of the drug to healthy volunteers in a randomized, double-blind, placebo-controlled 2-week crossover study. We analyzed blood for alpha and gamma interferons, neopterin, 2',5'-oligoadenylate synthetase, lymphocyte surface markers, lymphocyte proliferation after exposure to soluble antigens and mitogens, and natural killer cell activity. Minimal biologic effects were observed after administration of a single 200-mg dose to four volunteers; therefore, the dose was increased to 600 mg in 10 subjects. Only neopterin levels and symptoms were greater after administration of 600 mg of poly(I):poly(C12U) than after administration of placebo (Wilcoxon signed rank sum test, P = 0.06). A definite response in 2',5'-oligoadenylate synthetase activity, however, was seen in a few subjects. Neither alpha nor gamma interferon was detectable in serum after poly(I):poly(C12U) dosing. The neopterin changes after administration of poly(I):poly(C12U) were similar at both poly(I):poly(C12U) dose levels, with an early decrease at 6 h, a peak at 1 day, and a gradual decrease toward the baseline over the following 3 days. A mild flu-like syndrome occurred in one-half of the subjects following administration of poly(I):poly(C12U) and in only one subject following administration of placebo. This syndrome resolved within 16 h after poly(I):poly(C12U) dosing. We conclude that poly(I):poly(C12U) does not induce measurable levels of interferon and causes only minimal biologic or toxic effects among those parameters measured after administration of a single dose in the 200- to 600-mg dose range in health volunteers.


Assuntos
Antivirais/farmacologia , Poli I-C/farmacologia , Poli U/farmacologia , RNA de Cadeia Dupla/farmacologia , 2',5'-Oligoadenilato Sintetase/efeitos dos fármacos , Adulto , Antivirais/administração & dosagem , Biomarcadores , Biopterinas/análogos & derivados , Biopterinas/análise , Método Duplo-Cego , Humanos , Interferons/análise , Células Matadoras Naturais/efeitos dos fármacos , Ativação Linfocitária/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Neopterina , Neutrófilos/efeitos dos fármacos , Poli I-C/administração & dosagem , Poli U/administração & dosagem , RNA de Cadeia Dupla/administração & dosagem , Subpopulações de Linfócitos T/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA