Comparative preclinical drug metabolism and pharmacokinetic evaluation of novel 4-aminoquinoline anti-malarials.

The disposition of three 4-aminoquinoline leads, namely isoquine (ISO), des-ethyl isoquine (DEI) and N-tert-butyl isoquine (NTBI), were studied in a range of in vivo and in vitro assays to assist in selecting an appropriate candidate for further development. Analogous to amodiaquine (ADQ), ISO undergoes oxidative N-dealkylation to form DEI in vivo. Blood clearance of DEI was as much as 10-fold lower than that of ISO in animals and after oral administration, metabolite exposure exceeded that of parent by as much as 14-fold. Replacement of the N-ethyl with an N-tert-butyl substituent substantially reduced N-dealkylation as blood clearance of NTBI was approximately 2 to 3-fold lower than DEI in mouse, rat, dog and monkey. Mean NTBI oral bioavailability was generally higher than the other leads (>/=68%). Blood cell association was substantial for NTBI, particularly in dog and monkey, where blood to plasma concentration ratios >4 were observed. Human plasma protein binding was similar for NTBI, DEI, and des-ethyl amodiaquine (DEA). Allometric scaling predicted human blood clearance (CL) for NTBI to be low ( approximately 12% liver blood flow). All the 4-aminoquinolines inhibited recombinant human cytochrome P450 2D6 with similar potency; DEI also inhibited 1A2. On balance, NTBI appeared the most promising lead to progress towards full development.

[1]  T. Fazili,et al.  Severe falciparum malaria: a case report. , 2010, The Journal of the Oklahoma State Medical Association.

[2]  C. Davis,et al.  Preclinical drug metabolism and pharmacokinetic evaluation of GW844520, a novel anti-malarial mitochondrial electron transport inhibitor. , 2006, Journal of pharmaceutical sciences.

[3]  K. Kirk,et al.  Defining the role of PfCRT in Plasmodium falciparum chloroquine resistance , 2005, Molecular microbiology.

[4]  X. Xu,et al.  Quantitative determination of a novel dual PPAR α/γ agonist using on-line turbulent flow extraction with liquid chromatography-tandem mass spectrometry , 2005 .

[5]  S. Satpathy,et al.  Severe falciparum malaria , 2004, Indian journal of pediatrics.

[6]  Ruth H. Hughes,et al.  Isoquine and related amodiaquine analogues: a new generation of improved 4-aminoquinoline antimalarials. , 2003, Journal of medicinal chemistry.

[7]  Ronald T Borchardt,et al.  A comparison of commonly used polyethoxylated pharmaceutical excipients on their ability to inhibit P-glycoprotein activity in vitro. , 2002, Journal of pharmaceutical sciences.

[8]  I. Mahmood,et al.  Interspecies scaling of biliary excreted drugs. , 2002, Journal of pharmaceutical sciences.

[9]  Tommy B Andersson,et al.  Amodiaquine clearance and its metabolism to N-desethylamodiaquine is mediated by CYP2C8: a new high affinity and turnover enzyme-specific probe substrate. , 2002, The Journal of pharmacology and experimental therapeutics.

[10]  J. Polli,et al.  Rational use of in vitro P-glycoprotein assays in drug discovery. , 2001, The Journal of pharmacology and experimental therapeutics.

[11]  Wei Tang,et al.  Heterotropic cooperativity of cytochrome P450 3A4 and potential drug-drug interactions. , 2001, Current drug metabolism.

[12]  H. Ginsburg,et al.  Cellular Uptake of Chloroquine Is Dependent on Binding to Ferriprotoporphyrin IX and Is Independent of NHE Activity in Plasmodium falciparum , 1999, The Journal of cell biology.

[13]  B. W. Penman,et al.  Microtiter plate assays for inhibition of human, drug-metabolizing cytochromes P450. , 1997, Analytical biochemistry.

[14]  Tim Morris,et al.  Physiological Parameters in Laboratory Animals and Humans , 1993, Pharmaceutical Research.

[15]  A. Breckenridge,et al.  Drug-protein conjugates--XVIII. Detection of antibodies towards the antimalarial amodiaquine and its quinone imine metabolite in man and the rat. , 1989, Biochemical pharmacology.

[16]  L. Coulombel,et al.  Amodiaquine‐induced immune agranulocytosis , 1989, British journal of haematology.

[17]  K. Neftel,et al.  Amodiaquine induced agranulocytosis and liver damage. , 1986, British medical journal.

[18]  G. Edwards,et al.  FREQUENCY OF SEVERE NEUTROPENIA ASSOCIATED WITH AMODIAQUINE PROPHYLAXIS AGAINST MALARIA , 1986, The Lancet.

[19]  C. D. Fitch,et al.  High-affinity accumulation of chloroquine by mouse erythrocytes infected with Plasmodium berghei. , 1974, The Journal of clinical investigation.

[20]  P. Beaune,et al.  IN VITRO METABOLISM OF CHLOROQUINE : IDENTIFICATION OF CYP 2 C 8 , CYP 3 A 4 , AND CYP 2 D 6 AS THE MAIN ISOFORMS CATALYZING N-DESETHYLCHLOROQUINE FORMATION , 2003 .

[21]  P. J. Vries,et al.  Pharmacokinetic Interactions of Antimalarial Agents , 2001, Clinical pharmacokinetics.

[22]  M. Newell Vertical transmission of HIV-1 infection. , 2000, Transactions of the Royal Society of Tropical Medicine and Hygiene.

[23]  M. Bayliss,et al.  Utility of hepatocytes to model species differences in the metabolism of loxtidine and to predict pharmacokinetic parameters in rat, dog and man. , 1999, Xenobiotica; the fate of foreign compounds in biological systems.

[24]  D. Sullivan,et al.  Hemoglobin metabolism in the malaria parasite Plasmodium falciparum. , 1997, Annual review of microbiology.

[25]  P. Nguyen-Dinh,et al.  Sensitive analysis of blood for amodiaquine and three metabolites by high-performance liquid chromatography with electrochemical detection. , 1986, Journal of chromatography.