Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Toxicol In Vitro ; 62: 104669, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31629065

RESUMO

Amodiaquine (ADQ), an antimalarial drug used in endemic areas, has been reported to be associated with liver toxicity; however, the mechanism underlying its hepatoxicity remains unclear. In this study, we examined the cytotoxicity of ADQ and its major metabolite N-desethylamodiaquine (NADQ) and the effect of cytochrome P450 (CYP)-mediated metabolism on ADQ-induced cytotoxicity. After a 48-h treatment, ADQ and NADQ caused cytotoxicity and induced apoptosis in HepG2 cells; NADQ was slightly more toxic than ADQ. ADQ treatment decreased the levels of anti-apoptotic Bcl-2 family proteins, which was accompanied by an increase in the levels of pro-apoptotic Bcl-2 family proteins, indicating that ADQ-induced apoptosis was mediated by the Bcl-2 family. NADQ treatment markedly increased the phosphorylation of JNK, extracellular signal-regulated kinase (ERK1/2), and p38, indicating that NADQ-induced apoptosis was mediated by MAPK signaling pathways. Metabolic studies using microsomes obtained from HepG2 cell lines overexpressing human CYPs demonstrated that CYP1A1, 2C8, and 3A4 were the major enzymes that metabolized ADQ to NADQ and that CYP1A2, 1B1, 2C19, and 3A5 also metabolized ADQ, but to a lesser extent. The cytotoxicity of ADQ was increased in CYP2C8 and 3A4 overexpressing HepG2 cells compared to HepG2/CYP vector cells, confirming that NADQ was more toxic than ADQ. Moreover, treatment of CYP2C8 and 3A4 overexpressing HepG2 cells with ADQ increased the phosphorylation of JNK, ERK1/2, and p38, but not the expression of Bcl-2 family proteins. Our findings indicate that ADQ and its major metabolite NADQ induce apoptosis, which is mediated by members of the Bcl-2 family and the activation of MAPK signaling pathways, respectively.


Assuntos
Amodiaquina/análogos & derivados , Apoptose/efeitos dos fármacos , Amodiaquina/toxicidade , Sobrevivência Celular/efeitos dos fármacos , Sistema Enzimático do Citocromo P-450/metabolismo , Células Hep G2 , Humanos , Isoenzimas/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo
2.
Antiviral Res ; 160: 175-182, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30395872

RESUMO

Ebola virus disease is a severe disease caused by highly pathogenic Ebolaviruses. Although it shows a high mortality rate in humans, currently there is no licensed therapeutic. During the recent epidemic in West Africa, it was demonstrated that administration of antimalarial medication containing amodiaquine significantly lowered mortality rate of patients infected with the virus. Here, in order to improve its antiviral activity, a series of amodiaquine derivatives were synthesized and tested for Ebola virus infection. We found that multiple compounds were more potent than amodiaquine. The structure-activity relationship analysis revealed that the two independent parts, which are the alkyl chains extending from the aminomethyl group and a halogen bonded to the quinoline ring, were keys for enhancing antiviral potency without increasing toxicity. When these modifications were combined, the antiviral efficacy could be further improved with the selectivity indexes being over 10-times higher than amodiaquine. Mechanistic evaluation demonstrated that the potent derivatives blocked host cell entry of Ebola virus, like the parental amodiaquine. Taken together, our work identified novel potent amodiaquine derivatives, which will aid in further development of effective antiviral therapeutics.


Assuntos
Amodiaquina/síntese química , Amodiaquina/farmacologia , Antivirais/síntese química , Antivirais/farmacologia , Ebolavirus/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Amodiaquina/toxicidade , Antimaláricos/síntese química , Antimaláricos/farmacologia , Antimaláricos/toxicidade , Antivirais/toxicidade , Relação Estrutura-Atividade
3.
Chem Res Toxicol ; 31(8): 739-744, 2018 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-29938495

RESUMO

If idiosyncratic drug-induced liver injury (IDILI) is immune mediated, then it is logical that immune modulators may be able to affect liver injury caused by a drug. We have previously shown that modulating the immune system by impairing programmed cell death protein (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) signaling, both receptors involved in immune tolerance, was capable of producing an animal model of amodiaquine (AQ) IDILI with characteristics very similar to IDILI in humans. Other immune modulators may also increase liver injury caused by drugs that cause IDILI in humans. In this study, myeloid derived suppressor cells (MDSCs), transforming growth factor beta (TGF-ß), and lymphocyte-activation gene 3 (LAG3) were targeted with antibodies, with and without PD-1 and CTLA-4 impairment. We found that anti-Gr1 antibodies used to deplete MDSCs led to a significant increase in AQ-induced liver injury in wild-type mice; however, the injury was actually less in PD-1-/- mice, with or without anti-CTLA-4, and it was less than we have previously observed in PD-1-/- mice combined with anti-CTLA-4 without anti-Gr1. Addition of anti-LAG3 or anti-TGF-ß antibodies produced a small increase ALT in AQ-treated wild-type mice. There was a significant increase in ALT in PD-1-/- mice co-treated with anti-LAG3 or anti-TGF-ß relative to AQ-treated wild-type mice. In the case of TGF-ß, this was further increased by the addition of anti-CTLA-4, but if anything, there appeared to be a paradoxical decrease when anti-CTLA-4 was combined with anti-LAG3. Overall, the results from this study were not always as expected, and they highlight the complexity of the immune response, in particular immune tolerance, which appears to be the dominant immune response to drugs that cause IDILI.


Assuntos
Adjuvantes Imunológicos/farmacologia , Amodiaquina/toxicidade , Antimaláricos/toxicidade , Doença Hepática Induzida por Substâncias e Drogas , Animais , Antígeno CTLA-4/metabolismo , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Receptor de Morte Celular Programada 1/metabolismo , Transdução de Sinais
4.
Birth Defects Res ; 109(14): 1075-1126, 2017 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-28646540

RESUMO

The World Health Organization currently recommends quinine+clindamycin for use against malaria in the first trimester. This may soon change to recommending artemisinin-based combination therapies (standard duration of dosing = 3 days). The non-artemisinin partner drugs include amodiaquine, lumefantrine, mefloquine, piperaquine, sulfadoxine+pyrimethamine, and pyronaridine. For quinine, clindamycin, and mefloquine and the combinations of sulfadoxine+pyrimethamine and artemether+lumefantrine, there are reports (including studies without internal comparison groups) that combined describe 304 to >1100 exposures of women in the first trimester for each drug with no conclusive evidence of adverse effects on pregnancy at therapeutic doses. This is despite the fact that all of these drugs or drug combinations caused embryo deaths and/or malformations in at least one animal species and all except lumefantrine had at least one exposure ratio <1. It now seems that these animal studies overestimated the risk of developmental toxicity in women with malaria. Three other non-artemisinins (amodiaquine, piperaquine, and pyronaridine) have few or no reported exposures in women in the first trimester and have exposure ratios ≤2 based on studies in pregnant rats and rabbits with dosing throughout organogenesis. However, none of these drugs caused embryo deaths or malformations in pregnant rats and rabbits with the exception of pyronaridine, which caused embryo deaths only at a dose that was excessively toxic to the mothers. Thus, for amodiaquine, piperaquine, and pyronaridine, the testing in animals did not reveal findings of concern and the exposure ratios were in the range of the other non-artemisinin antimalarials described above. Birth Defects Research 109:1075-1126, 2017. © 2017 The Authors. Birth Defects Research Published by Wiley Periodicals, Inc.


Assuntos
Antimaláricos/farmacologia , Antimaláricos/toxicidade , Amodiaquina/farmacologia , Amodiaquina/uso terapêutico , Amodiaquina/toxicidade , Animais , Antimaláricos/administração & dosagem , Antimaláricos/metabolismo , Artemisininas , Combinação de Medicamentos , Feminino , Humanos , Malária/tratamento farmacológico , Malária Falciparum/tratamento farmacológico , Naftiridinas/farmacologia , Naftiridinas/toxicidade , Gravidez , Primeiro Trimestre da Gravidez/efeitos dos fármacos , Pirimetamina/uso terapêutico , Pirimetamina/toxicidade , Quinina/administração & dosagem , Quinolinas/farmacologia , Quinolinas/toxicidade , Coelhos , Ratos , Sulfadoxina/uso terapêutico , Sulfadoxina/toxicidade , Teratogênicos/análise
5.
Toxicol Lett ; 275: 83-91, 2017 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-28478157

RESUMO

Amodiaquine (AQ), an antimalarial drug, widely prescribed in endemic areas of Africa and Asia, is used in combination with artesunate as recommended by the WHO. However, due to its idiosyncratic hepatotoxicity and agranulocytosis, the therapeutic use has been discontinued in most countries. Oxidative bioactivation to protein-reactive quinonimines (QIs) by hepatic cytochrome P450s and myeloperoxidase (MPO) have been suggested to be important mechanisms underlying AQ idiosyncratic toxicity. However, the inactivation of the reactive QIs by detoxifying enzymes such as human glutathione S-transferases (GSTs) and NAD(P)H:quinone oxidoreducatase 1 (NQO1) has not been characterized yet. In the present study, the activities of 15 recombinant human GSTs and NQO1 in the inactivation of reactive QIs of AQ and its pharmacological active metabolite, N-desethylamodiaquine (DEAQ) were investigated. The results showed that GSTP1-1, GSTA4-4, GSTM4-4, GSTM2-2 and GSTA2-2 (activity in decreasing order) were active isoforms in catalyzing GSH conjugation of reactive QIs of AQ and DEAQ. Additionally, NQO1 was shown to inactivate these QIs by reduction. Simulation of the variability of cytosolic GST-activity based on the hepatic GST contents from 22 liver donors, showed a large variation in cytosolic inactivation of QIs by GSH, especially at a reduced GSH-concentration. In conclusion, the present study demonstrates that a low hepatic expression of the active GSTs and NQO1 may increase the susceptibility of patients to AQ idiosyncratic hepatotoxicity.


Assuntos
Amodiaquina/análogos & derivados , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Glutationa Transferase/metabolismo , Microssomos Hepáticos/efeitos dos fármacos , NAD(P)H Desidrogenase (Quinona)/metabolismo , Amodiaquina/metabolismo , Amodiaquina/toxicidade , Biocatálise , Doença Hepática Induzida por Substâncias e Drogas/enzimologia , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Escherichia coli/genética , Glutationa Transferase/genética , Humanos , Técnicas In Vitro , Isoenzimas , Microssomos Hepáticos/enzimologia , NAD(P)H Desidrogenase (Quinona)/genética , Proteínas Recombinantes , Transfecção
6.
Chem Res Toxicol ; 30(6): 1327-1332, 2017 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-28525267

RESUMO

There is increasing evidence that most idiosyncratic drug-induced liver injury (IDILI) is immune mediated, and in most cases, reactive metabolites appear to be responsible for the induction of this immune response. Reactive metabolites can cause cell damage with the release of damage-associated molecular patterns (DAMPs), which is thought to be involved in immune activation. Presumably, the reason that the liver is a common target of idiosyncratic drug reactions is because it is the major site of drug metabolism and reactive metabolite formation. Inflammasomes can be activated by DAMPs, and this may be a common mechanism by which DAMPs initiate an immune response. In this study, we tested the ability of drugs to induce the release of DAMPs that activate inflammasomes. The drugs tested were amodiaquine and nevirapine; both are associated with significant incidences of severe IDILI. The hepatocytes were a human hepatocarcinoma functional liver cell-4 (FLC-4) cell line. For the detection of inflammasome activation, we used the human macrophage cell line, THP-1 cells. We found that the supernatant from the incubation of both drugs with FLC-4 cells for 7 days led to increased caspase-1 activity and production of IL-1ß by THP-1 cells. However, amodiaquine alone also directly activated THP-1 cells. This is presumably because the myeloperoxidase in THP-1 cells can bioactivate amodiaquine to a reactive metabolite. In contrast, nevirapine requires cytochromes P450 for reactive metabolite formation and therefore required incubation with hepatocytes. These results support the hypothesis that reactive metabolites of drugs can cause the release of DAMPs, which in turn can activate inflammasomes. Inflammasome activation may be an important step in the activation of the immune system by drugs, which in some patients can lead to IDILI. Our in vitro model is simple and convenient for evaluating inflammasome activation, and this may be a method to screen drugs for IDILI risk.


Assuntos
Amodiaquina/toxicidade , Hepatócitos/efeitos dos fármacos , Inflamassomos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Nevirapina/toxicidade , Amodiaquina/administração & dosagem , Amodiaquina/efeitos adversos , Células Cultivadas , Relação Dose-Resposta a Droga , Hepatócitos/metabolismo , Humanos , Inflamassomos/metabolismo , Macrófagos/metabolismo , Nevirapina/administração & dosagem , Nevirapina/efeitos adversos , Relação Estrutura-Atividade
7.
J Immunotoxicol ; 13(5): 694-712, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27416278

RESUMO

Amodiaquine (AQ) is associated with a relatively high incidence of idiosyncratic drug-induced liver injury (IDILI) and agranulocytosis. A previous study reported that a combination of high dose AQ and glutathione (GSH) depletion led to liver injury. However, the characteristics of this toxicity were very different from AQ-induced liver injury in humans. We developed a model of AQ-induced liver injury with characteristics similar to the injury in humans by treating mice with lower doses of AQ for several weeks. In this study we found that not only did GSH depletion not increase AQ covalent binding to hepatic proteins at this lower dose, but also it paradoxically prevented the liver injury. We extended the model to rats and found AQ treatment led to a mild delayed onset liver injury that resolved despite continued treatment with AQ. Immunohistochemistry indicated the presence of Kupffer cell activation, apoptosis and hepatocyte proliferation in the liver. There was also an increase in serum IL-2, IL-5, IL-9, IL-12, MCP-1 and TGFß, but a decrease in leptin. Coincident with the elevated serum ALT, the number of liver CD4(+) T-cells, IL-17 secreting cells and TH17/Treg cells increased at Week 3 and decreased during continued treatment. Increases in NK1.1+ cells and activated M2 macrophages were also observed during liver injury. These results suggest that the outcome of the liver injury was determined by the balance between effector and regulatory cells. Co-treatment with cyclosporin prevented AQ-induced liver injury, which supports an immune mechanism. Retinoic acid (RA), which has been reported to enhance natural killer (NK) cell activity, exacerbated AQ-induced liver injury. These results suggest that AQ-induced IDILI is immune mediated and the subsequent adaptation appears to represent immune tolerance.


Assuntos
Agranulocitose/imunologia , Doença Hepática Induzida por Substâncias e Drogas/imunologia , Células Matadoras Naturais/imunologia , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Agranulocitose/induzido quimicamente , Amodiaquina/toxicidade , Animais , Células Cultivadas , Ciclosporina/administração & dosagem , Citocinas/metabolismo , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Endogâmicos BN , Remissão Espontânea , Degenerações Espinocerebelares , Tretinoína/administração & dosagem
8.
Toxicol In Vitro ; 29(7): 1916-31, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26239606

RESUMO

Testing hepatotoxicity is a crucial step in the development and toxicological assessment of drugs and chemicals. Bio-activation can lead to the formation of metabolites which may present toxicity for the organism. Classical cytotoxic tests are not always appropriate and are often insufficient, particularly when non metabolically-competent cells are used as the model system, leading to false-positive or false-negative results. We tested over 24 h the effects of eight reference compounds on two different cell models: primary cultures of rat hepatocytes and FAO hepatoma cells that lack metabolic properties. We performed inter-assay validation between three classical cytotoxicity assays and real-time cell impedance data. We then complemented these experiments with high-content screening (HCS) to determine the cell function disorders responsible for the observed effects. Among the different assays used, the neutral red test seemed to be well suited to our two cell models, coupled with real-time cellular impedance which proved useful in the detection of bio-activation. Indeed, impedance monitoring showed a high sensitivity with interesting curve profiles yet seemed unsuitable for evaluation of viability on primary culture. Finally, HCS in the evaluation of hepatotoxicity is likely to become an essential tool for use in parallel to a classical cytotoxic assay in the assessment of drugs and environmental chemicals.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas , Hepatócitos/efeitos dos fármacos , Ensaios de Triagem em Larga Escala , Acetaminofen/toxicidade , Amodiaquina/toxicidade , Animais , Carbamazepina/toxicidade , Linhagem Celular Tumoral , Sobrevivência Celular , Células Cultivadas , Dietilestilbestrol/toxicidade , Eritromicina/toxicidade , Furosemida/toxicidade , Hepatócitos/metabolismo , Masculino , Vermelho Neutro/metabolismo , Ratos , Testes de Toxicidade , Tretinoína/toxicidade
9.
Antiviral Res ; 106: 125-34, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24680954

RESUMO

Dengue virus serotypes 1-4 (DENV1-4) are transmitted by mosquitoes which cause most frequent arboviral infections in the world resulting in ∼390 million cases with ∼25,000 deaths annually. There is no vaccine or antiviral drug currently available for human use. Compounds containing quinoline scaffold were shown to inhibit flavivirus NS2B-NS3 protease (NS2B-NS3pro) with good potencies. In this study, we screened quinoline derivatives, which are known antimalarial drugs for inhibition of DENV2 and West Nile virus (WNV) replication using the corresponding replicon expressing cell-based assays. Amodiaquine (AQ), one of the 4-aminoquinoline drugs, inhibited DENV2 infectivity measured by plaque assays, with EC50 and EC90 values of 1.08±0.09µM and 2.69±0.47 µM, respectively, and DENV2 RNA replication measured by Renilla luciferase reporter assay, with EC50 value of 7.41±1.09µM in the replicon expressing cells. Cytotoxic concentration (CC50) in BHK-21 cells was 52.09±4.25µM. The replication inhibition was confirmed by plaque assay of the extracellular virions as well as by qRT-PCR of the intracellular and extracellular viral RNA levels. AQ was stable for at least 96h and had minor inhibitory effect on entry, translation, and post-replication stages in the viral life cycle. DENV protease, 5'-methyltransferase, and RNA-dependent RNA polymerase do not seem to be targets of AQ. Both p-hydroxyanilino and diethylaminomethyl moieties are important for AQ to inhibit DENV2 replication and infectivity. Our results support AQ as a promising candidate for anti-flaviviral therapy.


Assuntos
Amodiaquina/farmacologia , Antimaláricos/farmacologia , Antivirais/farmacologia , Vírus da Dengue/efeitos dos fármacos , Vírus da Dengue/fisiologia , Replicação Viral/efeitos dos fármacos , Amodiaquina/toxicidade , Animais , Antimaláricos/toxicidade , Antivirais/toxicidade , Sobrevivência Celular/efeitos dos fármacos , Cricetinae , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Testes de Sensibilidade Microbiana , Ensaio de Placa Viral , Vírus do Nilo Ocidental/efeitos dos fármacos , Vírus do Nilo Ocidental/fisiologia
10.
Bioorg Med Chem ; 21(17): 4904-13, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23896611

RESUMO

A series of new deoxyamodiaquine-based compounds was synthesized via the modified TMSN3-Ugi multi-component reaction and evaluated in vitro for antiplasmodial activity. The most potent compounds, 6b, 6c and 6j, showed IC50 values in the range of 6-77nM against chloroquine-resistant K1- and W2-strains of Plasmodium falciparum. In vitro ADME characterization of frontrunner compounds 6b and 6c indicates that these two compounds are rapidly metabolized and have a high clearance rate in human and rat liver microsomes. This result correlated well with an in vivo pharmacokinetics study, which showed low bioavailability of 6c in rats. Tentative metabolite identification was determined by LC-MS and suggested metabolic lability of groups attached to the tertiary nitrogen. Preliminary studies on 6b and 6c suggested strong inhibitory activity against the major CYP450 enzymes. In silico docking studies were used to rationalize strong inhibition of CYP3A4 by 6c. Full characterization and biological evaluation of the metabolites is currently underway in our laboratories.


Assuntos
Aminoquinolinas/síntese química , Amodiaquina/análogos & derivados , Antimaláricos/síntese química , Tetrazóis/química , Tetrazóis/síntese química , Aminoquinolinas/farmacocinética , Aminoquinolinas/toxicidade , Amodiaquina/farmacocinética , Amodiaquina/toxicidade , Animais , Antimaláricos/farmacocinética , Antimaláricos/toxicidade , Sítios de Ligação , Células CHO , Sobrevivência Celular/efeitos dos fármacos , Cricetinae , Cricetulus , Citocromo P-450 CYP3A/metabolismo , Inibidores do Citocromo P-450 CYP3A , Resistência a Medicamentos/efeitos dos fármacos , Meia-Vida , Humanos , Microssomos Hepáticos/metabolismo , Simulação de Acoplamento Molecular , Plasmodium falciparum/efeitos dos fármacos , Estrutura Terciária de Proteína , Ratos , Relação Estrutura-Atividade , Tetrazóis/farmacocinética , Tetrazóis/toxicidade
11.
Pak J Biol Sci ; 15(7): 353-7, 2012 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-24163962

RESUMO

Malaria infection is a common cause of morbidity and mortality especially in the tropics and subtropics. This has led to the increased prophylactic use ofpyrethroid insecticides and/or Amodiaquine (Aq) to combat the parasitic protozoan infection. The aim of this study was to investigate the comparative haemodynamic effects of pyrethroid insecticide and amodiaquine in rats. Experimental rats were randomly allocated into seven groups of five rats in each. Groups 1, 2 and 3 were exposed to pyrethroid by inhalation for 1, 2 and 3 min, respectively, while groups 4, 5 and 6 were administered Aqper oral at 5, 10 and 15 mg kg(-1) b.wt., respectively. Control rats were neither exposed to pyrethroid nor administered Aq. Pyrethroid insecticide led to reduced systolic, diastolic and mean arterial pressures, but increased pulse pressure. Aq treatment did not cause any significant variation in haemodynamic variables. Heart rate was comparable in all groups. Results from the study provide extended safety/toxicity profile for pyrethroid use and Aq treatment. Aq showed no cardiotoxic potential, while pyrethroids have hypotensive effect. It is thus recommended that exposure to pyrethroids should be minimized.


Assuntos
Amodiaquina/farmacologia , Antimaláricos/farmacologia , Hemodinâmica/efeitos dos fármacos , Inseticidas/farmacologia , Piretrinas/farmacologia , Amodiaquina/toxicidade , Animais , Antimaláricos/toxicidade , Pressão Arterial/efeitos dos fármacos , Feminino , Frequência Cardíaca/efeitos dos fármacos , Hipotensão/induzido quimicamente , Hipotensão/fisiopatologia , Inseticidas/toxicidade , Masculino , Piretrinas/toxicidade , Ratos , Ratos Sprague-Dawley , Medição de Risco
12.
Med Mal Infect ; 39(1): 57-60, 2009 Jan.
Artigo em Francês | MEDLINE | ID: mdl-19013042

RESUMO

Two previously healthy young women presented with a lethal hepatitis a few days after the onset of an artesunate-amodiaquine combination at the recommended doses for a bout of fever. Nothing proved the fever was due to malaria, the toxic cause of hepatitis, or to the drugs used. Imputability was based on chronology (fever, drug combination, sudden onset of severe fatigue, hepatitis lethal in a few days) and on the absence of any other evident cause for hepatitis. Severe hepatitis under prolonged amodiaquine treatment has been reported since 1985, the risk is estimated at 1/15500 treatments and the symptoms usually appear within 10 to 160 days. The current international recommendations are to promptly treat uncomplicated malaria access, with an artemisinin-based combination therapy, especially with artesunate-amodiaquine. The risk of iterative amodiaquine use could be the same as prolonged treatments, given that the suspected toxicity mechanisms are metabolite accumulation or an immunoallergic phenomenon. All adverse effects must be reported.


Assuntos
Amodiaquina/toxicidade , Antimaláricos/toxicidade , Artemisininas/toxicidade , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Malária/tratamento farmacológico , Adulto , Artesunato , Evolução Fatal , Feminino , Humanos
13.
Toxicology ; 256(1-2): 101-9, 2009 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-19059302

RESUMO

Amodiaquine is an antimalarial drug that causes life-threatening agranulocytosis and hepatotoxicity in about 1 in 2000 patients, which is usually associated with an inflammatory response. It was found that the LC(50) (2h) of amodiaquine towards isolated rat hepatocytes was 1mM. The cytotoxic mechanism involved protein carbonylation as well as P450 activation to a reactive metabolite. The cytotoxicity, however, was not reactive oxygen species (ROS)-mediated, as ROS scavengers did not prevent cytotoxicity or protein carbonylation, and it was not accompanied by glutathione (GSH) oxidation or intracellular H(2)O(2) formation. On the other hand, the cytotoxicity could be attributed to a quinoneimine metabolite formation which formed GSH conjugates and GSH-depleted hepatocytes were much more susceptible to amodiaquine. Furthermore, when a non-toxic H(2)O(2) generating system and peroxidase was used to mimic the products formed by inflammatory immune cells, only 15microM amodiaquine was required to cause 50% cell death. In the absence of amodiaquine, hepatocyte viability and glutathione levels were not affected by the H(2)O(2) generating system with or without peroxidase. The toxicity mechanism of amodiaquine in this hepatocyte H(2)O(2)/peroxidase model involved oxidative stress, as cytotoxicity was accompanied by GSH oxidation, decreased mitochondrial membrane potential and protein carbonyl formation which were inhibited by ROS scavengers, 4-hydroxy-2,2,6,6-tetramethylpiperidene-1-oxyl (TEMPOL) or mannitol suggesting a role for a semiquinoneimine radical and ROS in the amodiaquine-H(2)O(2)-mediated cytotoxic mechanism.


Assuntos
Amodiaquina/toxicidade , Antimaláricos/toxicidade , Hepatócitos/efeitos dos fármacos , Inflamação/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Animais , Antioxidantes/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Óxidos N-Cíclicos/farmacologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Glutationa/metabolismo , Hepatócitos/enzimologia , Peróxido de Hidrogênio/toxicidade , Inflamação/enzimologia , Inflamação/patologia , Masculino , Manitol/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Membranas Mitocondriais/efeitos dos fármacos , Oxirredução , Carbonilação Proteica/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Marcadores de Spin
14.
Photochem Photobiol ; 83(6): 1415-27, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18028216

RESUMO

This article describes the results of a coupled photophysical and photobiological study aimed at understanding the phototoxicity mechanism of the antimalarial drugs amodiaquine (AQ), primaquine (PQ) and chloroquine (CQ). Photophysical experiments were carried out in aqueous solutions by steady-state and time-resolved spectrometric techniques to obtain information on the different decay pathways of the excited states of the drugs and on the transient species formed upon laser irradiation. The results showed that all three drugs possess very low fluorescence quantum yields (10(-2)-10(-4)). Laser flash photolysis experiments proved the occurrence of photoionization processes leading to the formation of a radical cation in all three systems. In the case of AQ the lowest triplet state was also detected. Together with the photophysical properties the photobiological properties of the antimalarial drugs were investigated under UV irradiation, on various biological targets through a series of in vitro assays. Phototoxicity on mouse 3T3 fibroblast and human keratinocyte cell lines NCTC-2544 was detected for PQ and CQ but not for AQ. In particular, PQ- and CQ-induced apoptosis was revealed by the externalization of phosphatidylserine. Furthermore, upon UV irradiation, the drugs caused significant variations of the mitochondrial potential (Deltapsi(mt)) measured by flow cytometry. The photodamages produced by the drugs were also evaluated on proteins, lipids and DNA. The combined approaches were useful in understanding the mechanism of phototoxicity induced by these antimalarial drugs.


Assuntos
Amodiaquina/química , Cloroquina/química , Primaquina/química , Amodiaquina/toxicidade , Animais , Antimaláricos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Cloroquina/toxicidade , DNA/genética , Dano ao DNA , Humanos , Peroxidação de Lipídeos/efeitos dos fármacos , Peroxidação de Lipídeos/efeitos da radiação , Lisossomos/efeitos dos fármacos , Lisossomos/efeitos da radiação , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/efeitos da radiação , Estrutura Molecular , Fotobiologia , Fotoquímica , Primaquina/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Salmão , Espectrofotometria
15.
Chem Biol Interact ; 121(1): 77-97, 1999 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-10418972

RESUMO

Ethical, economic and technical reasons hinder regular supply of freshly isolated hepatocytes from higher mammals such as monkey for preclinical evaluation of drugs. Hence, we aimed at developing optimal and reproducible protocols to cryopreserve and thaw parenchymal liver cells from this major toxicological species. Before the routine use of these protocols, we validated them through a multi-laboratory study. Dissociation of the whole animal liver resulted in obtaining 1-5 billion parenchymal cells with a viability of about 86%. An appropriate fraction (around 20%) of the freshly isolated cells was immediately set in primary culture and various hepato-specific tests were performed to examine their metabolic, biochemical and toxicological functions as well as their ultrastructural characteristics. The major part of the hepatocytes was frozen and their functionality checked using the same parameters after thawing. The characterization of fresh and thawed monkey hepatocytes demonstrated the maintenance of various hepato-specific functions. Indeed, cryopreserved hepatocytes were able to survive and to function in culture as well as their fresh counterparts. The ability for synthesis (proteins, ATP, GSH) and conjugation and secretion of biliary acids was preserved after deep freeze storage. A better stability of drug metabolizing activities than in rodent hepatocytes was observed in monkey. After thawing, Phase I and Phase II activities (cytochrome P450, ethoxycoumarin-O-deethylase, aldrin epoxidase, epoxide hydrolase, glutathione transferase, glutathione reductase and glutathione peroxidase) were well preserved. The metabolic patterns of several drugs were qualitatively and quantitatively similar before and after cryopreservation. Lastly, cytotoxicity tests suggested that the freezing/thawing steps did not change cell sensitivity to toxic compounds.


Assuntos
Criopreservação , Avaliação Pré-Clínica de Medicamentos/métodos , Fígado/fisiologia , Preservação de Órgãos , Trifosfato de Adenosina/metabolismo , Amodiaquina/toxicidade , Animais , Sobrevivência Celular , Células Cultivadas , Enzimas/metabolismo , Eritromicina/toxicidade , Estudos de Avaliação como Assunto , Furosemida/toxicidade , Glutationa/metabolismo , Fígado/citologia , Fígado/efeitos dos fármacos , Macaca fascicularis , Masculino , Testes de Toxicidade
16.
Chem Res Toxicol ; 11(12): 1586-95, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9860505

RESUMO

Life-threatening agranulocytosis and hepatotoxicity during prophylactic administration of amodiaquine have led to its withdrawal. Agranulocytosis is thought to involve bioactivation to a protein-reactive quinoneimine metabolite. The toxicity of amodiaquine and the lack of cheap drugs have prompted a search for alternative antimalarial agents. The aim of this study was to determine the metabolism and neutrophil toxicity of amodiaquine, pyronaridine, and other related antimalarial agents. Horseradish peroxidase and hydrogen peroxide were used to activate drugs to their respective quinoneimine metabolites. Metabolites were trapped as stable glutathione conjugates, prior to analysis by LC/MS. Amodiaquine was metabolized to a polar metabolite (m/z 661), identified as a glutathione adduct. Tebuquine was converted to two polar metabolites. The principal metabolite (m/z 686) was derived from glutathione conjugation and side chain elimination, while the minor metabolite gave a protonated molecule (m/z 496). Only parent ions were identified when chloroquine, cycloquine, or pyronaridine was incubated with the activating system and glutathione. Calculation of the heat of formation of the drugs, however, demonstrated that amodiaquine, tebuquine, cycloquine, and pyronaridine readily undergo oxidation to their quinoneimine. None of the antimalarial compounds depleted the level of intracellular glutathione (1-300 microM) when incubated with neutrophils alone. Additionally, with the exception of tebuquine, no cytotoxicity below 100 microM was observed. In the presence of the full activating system, however, all compounds except chloroquine resulted in depletion of the level of glutathione and were cytotoxic. Pretreating the cells with glutathione and other antioxidants inhibited metabolism-dependent cytotoxicity. In summary, our data show that amodiaquine and related antimalarials containing a p-aminophenol moiety undergo bioactivation in vitro to chemically reactive and cytotoxic intermediates. In particular, pyronaridine, which is currently being investigated in humans, was metabolized to a compound which was toxic to neutrophils. Thus, the possibility that it will cause agranulocytosis in clinical practice cannot be excluded, and will require careful monitoring.


Assuntos
Amodiaquina/toxicidade , Antimaláricos/toxicidade , Naftiridinas/toxicidade , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Adulto , Amodiaquina/farmacocinética , Antimaláricos/farmacocinética , Biotransformação , Sobrevivência Celular/efeitos dos fármacos , Glutationa/metabolismo , Peroxidase do Rábano Silvestre/metabolismo , Humanos , Peróxido de Hidrogênio/metabolismo , Técnicas In Vitro , Naftiridinas/farmacocinética , Oxirredução
17.
Mutagenesis ; 13(6): 619-24, 1998 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-9862194

RESUMO

Comparative mutagenic and genotoxic effects of three antimalarial drugs, chloroquine, primaquine and amodiaquine, were assessed in the Ames mutagenicity assay (in strains TA97a, TA100, TA102 and TA104) and in vivo sister chromatid exchange (SCE) and chromosome aberration (CA) assays in bone marrow cells of mice. These are the most commonly used antimalarial drugs available at present throughout the world. The results of the bacterial mutagenicity assays showed a very weak mutagenic effect of all three drugs in Salmonella strains TA97a and TA100 both with and without S9 mix and in TA104 only with S9 mix. The results of the in vivo SCE and CA assays indicate that these three drugs are genotoxic in bone marrow cells of mice.


Assuntos
Amodiaquina/toxicidade , Antimaláricos/toxicidade , Cloroquina/toxicidade , Primaquina/toxicidade , Animais , Medula Óssea/efeitos dos fármacos , Aberrações Cromossômicas , Masculino , Camundongos , Testes de Mutagenicidade/métodos , Mutagênicos/toxicidade , Salmonella typhimurium/efeitos dos fármacos , Salmonella typhimurium/genética , Troca de Cromátide Irmã/efeitos dos fármacos
18.
J Pharmacol Exp Ther ; 280(2): 884-93, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9023303

RESUMO

The development and clinical use of 4-aminoquinoline antimalarial agents such as amodiaquine have been limited by toxicity to neutrophils. We have investigated the chemical basis of amodiaquine-induced toxicity and compared the findings with those for established antimalarial drugs proposed for human use. Amodiaquine, like chloroquine, mefloquine and halofantrine, was lysosomotropic and accumulated in human neutrophils. Amodiaquine did not lead to impairment of either cellular function or cell viability at therapeutic levels. In contrast to other antimalarial agents, amodiaquine (because it contains a 4-aminophenol function) depleted glutathione in activated neutrophils, by formation of an electrophilic quinoneimine metabolite. Bioactivation was accompanied by the expression of a drug-related antigen on the cell surface, which was recognized by drug-specific antibodies, suggesting that a type II hypersensitivity reaction is responsible for the observed toxicity. Similar bioactivation and accumulation were observed for the structurally related amopyroquine. The effects of chemical modifications at the 3'- and 5'-positions, which are known to enhance antimalarial activity, were also investigated. The introduction of a lipophilic 5'-chlorophenyl group and 3'-t-butyl group blocked bioactivation but enhanced cellular accumulation, with resultant impairment of function and neutrophil viability, whereas introduction of a second cationic dialkylamino group (bis-mannich compounds) blocked bioactivation and reduced cellular accumulation, without producing noticeable effects on cellular function and viability. These data provide a chemical rationale for the idiosyncratic agranulocytosis observed with amodiaquine, and they suggest that similar toxicity might be anticipated for amopyroquine but is less likely with bis-mannich antimalarial agents such as pyronaridine.


Assuntos
Amodiaquina/análogos & derivados , Amodiaquina/sangue , Antimaláricos/sangue , Leucócitos Mononucleares/metabolismo , Neutrófilos/metabolismo , Amodiaquina/farmacocinética , Amodiaquina/toxicidade , Antimaláricos/farmacocinética , Antimaláricos/toxicidade , Sobrevivência Celular/efeitos dos fármacos , Desenho de Fármacos , Glutationa/sangue , Humanos , Técnicas In Vitro , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/patologia , Estrutura Molecular , Neutrófilos/efeitos dos fármacos , Neutrófilos/patologia , Relação Estrutura-Atividade , Acetato de Tetradecanoilforbol/farmacologia , Trítio
19.
Biochem Pharmacol ; 43(7): 1421-30, 1992 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-1567466

RESUMO

A glutathione conjugate of amodiaquine has been isolated and characterized from rat bile after administration of [14C]amodiaquine (50 mumol/kg, 5.0 muCi/rat) to anaesthetized male Wistar rats. Thioether conjugates of amodiaquine in rat bile accounted for a total of 12% of the dose, 5 hr after administration of the drug. In addition, 1% of the dose remained in the liver covalently bound to tissue proteins after 5 hr. These findings provide direct evidence that a chemically reactive metabolite, amodiaquine quinoneimine, has been formed from the drug in vivo. A second major metabolite, desethylamodiaquine, accounting for 14% of the given dose, was present in the liver after 5 hr. Enzyme inhibition studies with ketoconazole-pretreated rats showed that both amodiaquine quinoneimine and desethylamodiaquine formation can be catalysed by cytochrome P450. The demonstration that amodiaquine readily and extensively forms a metabolite in vivo, with strong reactivity towards protein and non-protein thiol groups, may help to explain the idiosyncratic toxicity observed in man.


Assuntos
Amodiaquina/metabolismo , Bile/metabolismo , Fígado/metabolismo , Amodiaquina/toxicidade , Animais , Biotransformação , Radioisótopos de Carbono , Cisteína/metabolismo , Glutationa/metabolismo , Iminas/metabolismo , Cetoconazol/farmacologia , Masculino , Espectrometria de Massas , Ratos , Ratos Endogâmicos
20.
Br J Clin Pharmacol ; 29(4): 479-85, 1990 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-2328196

RESUMO

The cytotoxicity of amodiaquine (AQ), amodiaquine quinoneimine (AQQI) and desethylamodiaquine (AQm) has been assessed in comparison with that of chloroquine (CQ) using mononuclear leucocytes (MNL) and granulocyte/monocyte colony forming units (GM-CFU) from haematologically normal subjects. Toxicity toward MNL was assessed after 2 h and 16 h incubations with each compound. After 2 h, AQ, AQm and AQQI but not CQ (within the concentration range 1-100 mumols l-1) produced a significant decrease in cell viability. After 16 h, all four compounds significantly increased cell death. After both 2 h and 16 h incubations CQ was the least toxic and AQQI the most toxic of the four compounds towards MNL. Toxicity to GM-CFU was assessed by the inhibition of colony formation in vitro. After 10-14 days incubation, there was significant concentration-dependent inhibition of colony formation by AQ, AQm, AQQI and CQ (within the range 0.1-10.0 mumols l-1). There were no significant differences between the ability of the four compounds to inhibit colony formation but toxicity towards GM-CFU was observed at drug concentrations at least 10-fold lower than those that were toxic to MNL. These data show that the four compounds are equally toxic in vitro toward GM-CFU, although some differences in their toxicity toward MNL were seen. The possible mechanisms of AQ's toxicity are discussed.


Assuntos
Amodiaquina/toxicidade , Granulócitos/efeitos dos fármacos , Leucócitos Mononucleares/efeitos dos fármacos , Amodiaquina/análogos & derivados , Medula Óssea/efeitos dos fármacos , Células da Medula Óssea , Sobrevivência Celular/efeitos dos fármacos , Cloroquina/toxicidade , Ensaio de Unidades Formadoras de Colônias , Humanos , Técnicas In Vitro
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...