Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Travel Med Infect Dis ; 45: 102211, 2022.
Article in English | MEDLINE | ID: mdl-34801714

ABSTRACT

BACKGROUND: Tafenoquine is a long-acting 8-aminoquinoline approved for antimalarial prophylaxis for ≤6 months. Additional data is needed to establish the drug's longer-term safety profile, including potential ophthalmic or neuropsychiatric effects. METHOD: This was a randomized, double-blind, placebo-controlled trial in 600 healthy adults. Eligible subjects were randomized 1:1 to receive tafenoquine 200 mg weekly (antimalarial prophylactic regimen) or placebo for 52 weeks. Scheduled safety visits occurred at Weeks 4, 12, 24, 52 (dosing completed), and 64 (final follow-up). Safety assessments included ophthalmic changes, general and neuropsychiatric adverse events (AEs), and laboratory value changes. RESULTS: The percentage of subjects with a protocol-defined Serious Ophthalmic Safety Event was lower in the Tafenoquine Group (18.2%) versus the Placebo Group (19%, p = 0.308). There was no significant difference between the percentages of subjects with at least one AE in the Tafenoquine Group (91.0%) versus Placebo (89.9%, p = 0.65). Common AEs seen at a significantly higher incidence for tafenoquine included reversible cornea verticillata (54.5%) and nausea (13.0%), leading to 0.0% and 0.7% discontinuations. Psychiatric AEs occurred at similar percentages in both study groups. Reversible changes in hemoglobin, methemoglobin, creatinine, and blood urea nitrogen (BUN) were noted. CONCLUSIONS: This study supports the safety of extended 52-week tafenoquine prophylaxis. CLINICAL TRIAL REGISTRATION NUMBER/CLINICALTRIALS. GOV IDENTIFIER: NCT03320174.


Subject(s)
Antimalarials , Adult , Aminoquinolines , Antimalarials/adverse effects , Chemoprevention , Double-Blind Method , Humans , Incidence , Treatment Outcome
2.
J Med Chem ; 63(11): 6179-6202, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32390431

ABSTRACT

The global impact of malaria remains staggering despite extensive efforts to eradicate the disease. With increasing drug resistance and the absence of a clinically available vaccine, there is an urgent need for novel, affordable, and safe drugs for prevention and treatment of malaria. Previously, we described a novel antimalarial acridone chemotype that is potent against both blood-stage and liver-stage malaria parasites. Here, we describe an optimization process that has produced a second-generation acridone series with significant improvements in efficacy, metabolic stability, pharmacokinetics, and safety profiles. These findings highlight the therapeutic potential of dual-stage targeting acridones as novel drug candidates for further preclinical development.


Subject(s)
Acridones/chemistry , Antimalarials/chemistry , Acridones/pharmacokinetics , Acridones/pharmacology , Acridones/therapeutic use , Administration, Oral , Animals , Antimalarials/pharmacokinetics , Antimalarials/pharmacology , Antimalarials/therapeutic use , Cell Survival/drug effects , Disease Models, Animal , Female , Half-Life , Hep G2 Cells , Humans , Life Cycle Stages/drug effects , Malaria/drug therapy , Malaria/pathology , Male , Mice , Mice, Inbred C57BL , Plasmodium falciparum/drug effects , Plasmodium falciparum/isolation & purification , Structure-Activity Relationship
3.
J Med Chem ; 62(7): 3475-3502, 2019 04 11.
Article in English | MEDLINE | ID: mdl-30852885

ABSTRACT

Malaria remains one of the deadliest diseases in the world today. Novel chemoprophylactic and chemotherapeutic antimalarials are needed to support the renewed eradication agenda. We have discovered a novel antimalarial acridone chemotype with dual-stage activity against both liver-stage and blood-stage malaria. Several lead compounds generated from structural optimization of a large library of novel acridones exhibit efficacy in the following systems: (1) picomolar inhibition of in vitro Plasmodium falciparum blood-stage growth against multidrug-resistant parasites; (2) curative efficacy after oral administration in an erythrocytic Plasmodium yoelii murine malaria model; (3) prevention of in vitro Plasmodium berghei sporozoite-induced development in human hepatocytes; and (4) protection of in vivo P. berghei sporozoite-induced infection in mice. This study offers the first account of liver-stage antimalarial activity in an acridone chemotype. Details of the design, chemistry, structure-activity relationships, safety, metabolic/pharmacokinetic studies, and mechanistic investigation are presented herein.


Subject(s)
Acridones/chemistry , Acridones/pharmacology , Antimalarials/chemistry , Antimalarials/pharmacology , Drug Discovery/methods , Acridones/therapeutic use , Animals , Antimalarials/therapeutic use , Disease Models, Animal , Hep G2 Cells , Humans , Malaria/drug therapy , Mice , Plasmodium/classification , Plasmodium/drug effects , Species Specificity , Structure-Activity Relationship
4.
Clin Infect Dis ; 69(3): 480-486, 2019 07 18.
Article in English | MEDLINE | ID: mdl-30388194

ABSTRACT

BACKGROUND: Tafenoquine was recently approved for chemoprophylaxis of malaria. Its specific activity against liver and blood stages of Plasmodium species has been separately characterized in animals but not in humans. METHODS: In this randomized, double-blind, placebo-controlled study, 16 malaria-naive, glucose-6-phosphate dehydrogenase-normal participants aged 20-42 years received tafenoquine chemoprophylaxis prior to challenge with blood stage Plasmodium falciparum. Participants were randomly assigned to either tafenoquine (n = 12) or placebo (n = 4) and took blinded study medication (single 200-mg dose) on days 1, 2, 3, and 10, followed by intravenous inoculation with approximately 2800 P. falciparum parasitized erythrocytes on day 13. The primary endpoint was the number of participants requiring rescue treatment with artemether/lumefantrine due to the onset of parasitemia as determined by quantitative polymerase chain reaction. RESULTS: None of the 12 participants who received tafenoquine developed parasitemia, whereas all placebo participants developed parasitemia (P = .0005). Two cases of mild hemoglobin decrease and a single case of mild hyperbilirubinemia occurred in the tafenoquine group. CONCLUSIONS: Tafenoquine chemoprophylaxis is safe and effective in preventing malaria in healthy nonimmune participants challenged with blood stage P. falciparum. CLINICAL TRIALS REGISTRATION: Australian and New Zealand Clinical Trials Registry (ANZCTR): ACTRN12617000102370.


Subject(s)
Aminoquinolines/administration & dosage , Antimalarials/administration & dosage , Malaria, Falciparum/prevention & control , Adult , Chemoprevention/methods , Double-Blind Method , Female , Healthy Volunteers , Humans , Male , Parasitemia/prevention & control , Placebos , Plasmodium falciparum/drug effects , Schizonts/drug effects , Young Adult
5.
Malar Res Treat ; 2017: 7508291, 2017.
Article in English | MEDLINE | ID: mdl-28491482

ABSTRACT

Decoquinate nanoparticle and microparticle suspended in an oily vehicle to retard drug release are evaluated for long-term malaria prophylaxis. Pharmacokinetic studies in normal animals and antimalarial efficacy in liver stage malaria mice were conducted at various single intramuscular-decoquinate doses for 2, 4, 6, or 8 weeks prior to infection with P. berghei sporozoites. The liver stage efficacy evaluation was monitored by using an in vivo imaging system. Full causal prophylaxis was shown in mice with a single intramuscular dose at 120 mg/kg of nanoparticle decoquinate (0.43 µm) for 2-3 weeks and with microparticle decoquinate (8.31 µm) injected 8 weeks earlier than inoculation. The time above MIC of 1,375 hr observed with the microparticle formulation provided a 2.2-fold longer drug exposure than with the nanoparticle formulation (624 hr). The prophylactic effect of the microparticle formulation observed in mice was shown to be 3-4 times longer than the nanoparticle decoquinate formulation.

6.
Travel Med Infect Dis ; 17: 19-27, 2017.
Article in English | MEDLINE | ID: mdl-28495354

ABSTRACT

BACKGROUND: Tafenoquine is a new prophylactic antimalarial drug. The current analysis presents an integrated safety assessment of the Tafenoquine Anticipated Clinical Regimen (Tafenoquine ACR) from 5 clinical trials, including 1 conducted in deployed military personnel and 4 in non-deployed residents, which also incorporated placebo and mefloquine comparator groups. METHODS: Adverse events (AEs) were coded according to the Medical Dictionary for Regulatory Activities (MedDRA®, Version 15.0) and summarized. Among all subjects who had received the Tafenoquine ACR, safety findings were compared for subjects who were deployed military personnel from the Australian Defence Force (Deployed ADF) versus non-deployed residents (Resident Non-ADF). RESULTS: The incidence of at least one AE was 80.6%, 64.1%, 67.6% and 94.9% in the mefloquine, placebo, tafenoquine Resident Non-ADF and tafenoquine Deployed ADF groups, respectively. The latter group had a higher incidence of AEs related to military deployment. AEs that occurred at ≥ 1% incidence in both tafenoquine sub-groups and at a higher frequency than placebo included diarrhea, nausea, vomiting, gastroenteritis, nasopharyngeal tract infections, and back/neck pain. CONCLUSIONS: Weekly administration of tafenoquine for up to six months increased the incidence of gastrointestinal AEs, certain infections, and back/neck pain, but not the overall incidence of AEs versus placebo. CLINICAL TRIAL REGISTRATION NUMBERS/CLINICALTRIALS. GOV IDENTIFIERS: NCT02491606; NCT02488980; NCT02488902.


Subject(s)
Aminoquinolines/adverse effects , Antimalarials/adverse effects , Drug-Related Side Effects and Adverse Reactions/epidemiology , Malaria/drug therapy , Malaria/prevention & control , Adolescent , Adult , Aged , Aminoquinolines/therapeutic use , Antimalarials/therapeutic use , Female , Humans , Incidence , Male , Middle Aged , Military Personnel , Randomized Controlled Trials as Topic , Young Adult
7.
Mil Med ; 182(S1): 360-368, 2017 03.
Article in English | MEDLINE | ID: mdl-28291500

ABSTRACT

BACKGROUND: The liver stages of Plasmodium parasites are important targets for the discovery and development of prophylactic drugs. METHODS: A real-time in vivo imaging system was used to determine the level of luminescence measured from firefly luciferase expression by sporozoites developing in hepatocytes in different strains of mice. RESULTS: The luminescence values (photon counts/sec) measured from the anatomical liver location in the untreated mice infected with 10,000 Plasmodium berghei sporozoites were 8.15 × 105 for C57BL/6 Albino, 2.12 × 105 for C3H/HeNCrL, 0.91 × 105 for C57BL/6 WT, 0.28 × 105 for BALB/c, and 0.16 × 105 for ICR/CD-1 mice. This data suggests that the C57BL/6 Albino strain is most susceptible to luminescent photon, mainly because the less light scattering and absorption from deeper tissues and the skin in the strain of mouse. The photon count observed in black C57BL/6 wild type mice was shown to be 88.83% lower compared to C57BL/6 Albino mice. Although the highest growth rate of sporozoites in hepatocytes was found for C57BL/6 wild type mice in this study, the black skin of this mouse significantly reduced parasite-associated bioluminescence. CONCLUSIONS: The minimal light scattering and absorption and also enhanced susceptibility to liver infection of C57BL/6 Albino mice makes this strain preferable sensitivity for discovery and development of prophylactic antimalarial drugs.


Subject(s)
Disease Susceptibility/physiopathology , Liver/physiopathology , Mice/parasitology , Plasmodium berghei/pathogenicity , Animals , Female , Male , Mice, Inbred BALB C/parasitology , Mice, Inbred C3H/parasitology , Mice, Inbred C57BL/parasitology , Mice, Inbred ICR/parasitology
8.
Article in English | MEDLINE | ID: mdl-28137819

ABSTRACT

In any drug discovery and development effort, a reduction in the time of the lead optimization cycle is critical to decrease the time to license and reduce costs. In addition, ethical guidelines call for the more ethical use of animals to minimize the number of animals used and decrease their suffering. Therefore, any effort to develop drugs to treat cutaneous leishmaniasis requires multiple tiers of in vivo testing that start with higher-throughput efficacy assessments and progress to lower-throughput models with the most clinical relevance. Here, we describe the validation of a high-throughput, first-tier, noninvasive model of lesion suppression that uses an in vivo optical imaging technology for the initial screening of compounds. A strong correlation between luciferase activity and the parasite load at up to 18 days postinfection was found. This correlation allows the direct assessment of the effects of drug treatment on parasite burden. We demonstrate that there is a strong correlation between drug efficacy measured on day 18 postinfection and the suppression of lesion size by day 60 postinfection, which allows us to reach an accurate conclusion on drug efficacy in only 18 days. Compounds demonstrating a significant reduction in the bioluminescence signal compared to that in control animals can be tested in lower-throughput, more definitive tests of lesion cure in BALB/c mice and Golden Syrian hamsters (GSH) using Old World and New World parasites.


Subject(s)
Antiprotozoal Agents/pharmacology , High-Throughput Screening Assays , Leishmania major/drug effects , Leishmaniasis, Cutaneous/drug therapy , Organisms, Genetically Modified , Amphotericin B/pharmacology , Animals , Cell Line , Cell Survival/drug effects , Drug Evaluation, Preclinical/economics , Drug Evaluation, Preclinical/methods , Female , Firefly Luciferin/administration & dosage , Fluconazole/pharmacology , Genes, Reporter , Leishmania major/genetics , Leishmania major/growth & development , Leishmaniasis, Cutaneous/parasitology , Leishmaniasis, Cutaneous/pathology , Luciferases/genetics , Luciferases/metabolism , Luminescent Measurements , Macrophages/cytology , Macrophages/drug effects , Meglumine/pharmacology , Meglumine Antimoniate , Mesocricetus , Mice , Mice, Inbred BALB C , Ofloxacin/pharmacology , Optical Imaging , Organometallic Compounds/pharmacology , Triazoles/pharmacology
9.
Malar J ; 15(1): 588, 2016 Dec 07.
Article in English | MEDLINE | ID: mdl-27923405

ABSTRACT

BACKGROUND: Due to the ability of the 8-aminoquinolines (8AQs) to kill different stages of the malaria parasite, primaquine (PQ) and tafenoquine (TQ) are vital for causal prophylaxis and the eradication of erythrocytic Plasmodium sp. parasites. Recognizing the potential role of cytochrome (CYP) 450 2D6 in the metabolism and subsequent hepatic efficacy of 8-aminoquinolines, studies were designed to explore whether CYP2D-mediated metabolism was related to the ability of single-dose PQ and TQ to eliminate the asexual and sexual erythrocytic stages of Plasmodium berghei. METHODS: An IV P. berghei sporozoite murine challenge model was utilized to directly compare causal prophylactic and erythrocytic activity (asexual and sexual parasite stages) dose-response relationships in C57BL/6 wild-type (WT) mice and subsequently compare the erythrocytic activity of PQ and TQ in WT and CYP2D knock-out (KO) mice. RESULTS: Single-dose administration of either 25 mg/kg TQ or 40 mg/kg PQ eradicated the erythrocytic stages (asexual and sexual) of P. berghei in C57BL WT and CYP2D KO mice. In WT animals, the apparent elimination of hepatic infections occurs at lower doses of PQ than are required to eliminate erythrocytic infections. In contrast, the minimally effective dose of TQ needed to achieve causal prophylaxis and to eradicate erythrocytic parasites was analogous. CONCLUSION: The genetic deletion of the CYP2D cluster does not affect the ability of PQ or TQ to eradicate the blood stages (asexual and sexual) of P. berghei after single-dose administration.


Subject(s)
Aminoquinolines/pharmacology , Antimalarials/pharmacology , Cytochrome P-450 CYP2D6/metabolism , Malaria/drug therapy , Plasmodium berghei/drug effects , Primaquine/pharmacology , Aminoquinolines/administration & dosage , Animals , Antimalarials/administration & dosage , Chemoprevention/methods , Cytochrome P-450 CYP2D6/deficiency , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Therapy/methods , Female , Male , Mice, Inbred C57BL , Mice, Knockout , Primaquine/administration & dosage , Treatment Outcome
10.
Article in English | MEDLINE | ID: mdl-28077950

ABSTRACT

This study was designed to determine whether yoga might alleviate symptoms of pain, sleep disturbance, anxiety, and depression in children with cystic fibrosis (CF). CF is the most common genetic, life-limiting chronic disease among Caucasian populations. It primarily affects the lungs but also many other secretory organs and consequently leads to significant morbidities. Research has shown that children with CF have significantly increased depression, anxiety, and pain compared to their healthy counterparts. Subjects participated in six one-on-one sessions over a 10-week period with a certified instructor who designed each yoga practice based on a preestablished list of 30 yoga asanas. Questionnaires evaluating pain, sleep disturbance, sustained anxiety, immediate anxiety, and depression were administered. Differences between premeasures and postmeasures were evaluated using a two-sided test. Twenty subjects were assessed (12 females/8 males), median age of 11 (7-20) years. Mean immediate anxiety scores decreased (before session to after session 29 to 23.6, respectively, p < 0.001). Joint pain improved (3.25 to 3.65, p = 0.028). CFQ-R emotion subscale improved from 79.2 to 85 (p = 0.073), and the respiratory subscale improved from 66.7 to 79.2 (p = 0.076). Other results were less notable. We conclude that yoga may reduce immediate anxiety and joint pain in patients with CF.

11.
Am J Trop Med Hyg ; 94(2): 340-347, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26503273

ABSTRACT

Leishmaniasis is a complex tropical disease caused by kinetoplastid parasitic protozoa of the genus Leishmania and is transmitted by the sand fly insect vector. Cutaneous leishmaniasis (CL) is the most common form of this disease, and CL infections often result in serious skin lesions and scars. CL remains a public health problem in many endemic countries worldwide because of the absence of effective, safe, and cost-effective drugs for treatment. One of the strategies we chose to use to find novel chemical entities worthy of further development as antileishmanials involved screening synthetic and natural products libraries. In our study, we developed a Leishmania major intracellular amastigote assay that uses the activity of luciferase as a measure of parasite proliferation and used this assay to screen a collection of 400 compounds obtained from Medicines for Malaria Venture (MMV) for their antileishmanial activity. Our results showed that 14 compounds identified by MMV as antimalarial drugs have antileishmanial activity and can potentially be optimized for CL drug development.


Subject(s)
Antiprotozoal Agents/pharmacology , Drug Evaluation, Preclinical/methods , Leishmania major/drug effects , Antiprotozoal Agents/chemistry , Cell Survival , Hep G2 Cells , Humans , Inhibitory Concentration 50 , Molecular Structure
12.
Antimicrob Agents Chemother ; 59(7): 3864-9, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25870069

ABSTRACT

Cytochrome P450 (CYP) 2D metabolism is required for the liver-stage antimalarial efficacy of the 8-aminoquinoline molecule tafenoquine in mice. This could be problematic for Plasmodium vivax radical cure, as the human CYP 2D ortholog (2D6) is highly polymorphic. Diminished CYP 2D6 enzyme activity, as in the poor-metabolizer phenotype, could compromise radical curative efficacy in humans. Despite the importance of CYP 2D metabolism for tafenoquine liver-stage efficacy, the exact role that CYP 2D metabolism plays in the metabolism and pharmacokinetics of tafenoquine and other 8-aminoquinoline molecules has not been extensively studied. In this study, a series of tafenoquine pharmacokinetic experiments were conducted in mice with different CYP 2D metabolism statuses, including wild-type (WT) (reflecting extensive metabolizers for CYP 2D6 substrates) and CYPmouse 2D knockout (KO) (reflecting poor metabolizers for CYP 2D6 substrates) mice. Plasma and liver pharmacokinetic profiles from a single 20-mg/kg of body weight dose of tafenoquine differed between the strains; however, the differences were less striking than previous results obtained for primaquine in the same model. Additionally, the presence of a 5,6-ortho-quinone tafenoquine metabolite was examined in both mouse strains. The 5,6-ortho-quinone species of tafenoquine was observed, and concentrations of the metabolite were highest in the WT extensive-metabolizer phenotype. Altogether, this study indicates that CYP 2D metabolism in mice affects tafenoquine pharmacokinetics and could have implications for human tafenoquine pharmacokinetics in polymorphic CYP 2D6 human populations.


Subject(s)
Aminoquinolines/pharmacokinetics , Antimalarials/pharmacokinetics , Cytochrome P-450 CYP2D6/genetics , Aminoquinolines/blood , Animals , Antimalarials/blood , Area Under Curve , Biotransformation , Cytochrome P-450 CYP2D6/metabolism , Half-Life , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Primaquine/pharmacokinetics
13.
Antimicrob Agents Chemother ; 59(4): 2380-7, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25645856

ABSTRACT

Primaquine (PQ) metabolism by the cytochrome P450 (CYP) 2D family of enzymes is required for antimalarial activity in both humans (2D6) and mice (2D). Human CYP 2D6 is highly polymorphic, and decreased CYP 2D6 enzyme activity has been linked to decreased PQ antimalarial activity. Despite the importance of CYP 2D metabolism in PQ efficacy, the exact role that these enzymes play in PQ metabolism and pharmacokinetics has not been extensively studied in vivo. In this study, a series of PQ pharmacokinetic experiments were conducted in mice with differential CYP 2D metabolism characteristics, including wild-type (WT), CYP 2D knockout (KO), and humanized CYP 2D6 (KO/knock-in [KO/KI]) mice. Plasma and liver pharmacokinetic profiles from a single PQ dose (20 mg/kg of body weight) differed significantly among the strains for PQ and carboxy-PQ. Additionally, due to the suspected role of phenolic metabolites in PQ efficacy, these were probed using reference standards. Levels of phenolic metabolites were highest in mice capable of metabolizing CYP 2D6 substrates (WT and KO/KI 2D6 mice). PQ phenolic metabolites were present in different quantities in the two strains, illustrating species-specific differences in PQ metabolism between the human and mouse enzymes. Taking the data together, this report furthers understanding of PQ pharmacokinetics in the context of differential CYP 2D metabolism and has important implications for PQ administration in humans with different levels of CYP 2D6 enzyme activity.


Subject(s)
Antimalarials/pharmacokinetics , Cytochrome P-450 CYP2D6/metabolism , Primaquine/pharmacokinetics , Animals , Area Under Curve , Biotransformation , Cytochrome P-450 CYP2D6/genetics , Half-Life , Humans , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
14.
Infect Immun ; 82(12): 5308-16, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25287923

ABSTRACT

Enterotoxigenic Escherichia coli (ETEC) is a significant cause of diarrheal disease and death, especially in children in developing countries. ETEC causes disease by colonizing the small intestine and producing heat-labile toxin (LT), heat-stable toxin (ST), or both LT and ST (LT+ST). The majority of ETEC strains produce both ST and LT. Despite the prevalence of LT+ST-producing organisms, few studies have examined the physiologic or immunologic consequences of simultaneous exposure to these two potent enterotoxins. In the current report, we demonstrate that when LT and ST are both present, they increase water movement into the intestinal lumen over and above the levels observed with either toxin alone. As expected, cultured intestinal epithelial cells increased their expression of intracellular cyclic GMP (cGMP) when treated with ST and their expression of intracellular cyclic AMP (cAMP) when treated with LT. When both toxins were present, cGMP levels but not cAMP levels were synergistically elevated compared with the levels of expression caused by the corresponding single-toxin treatment. Our data also demonstrate that the levels of inflammatory cytokines produced by intestinal epithelial cells in response to LT are significantly reduced in animals exposed to both enterotoxins. These findings suggest that there may be complex differences between the epithelial cell intoxication and, potentially, secretory outcomes induced by ETEC strains expressing LT+ST compared with strains that express LT or ST only. Our results also reveal a novel mechanism wherein ST production may reduce the hosts' ability to mount an effective innate or adaptive immune response to infecting organisms.


Subject(s)
Bacterial Toxins/toxicity , Cytokines/metabolism , Enterotoxigenic Escherichia coli/physiology , Enterotoxins/toxicity , Epithelial Cells/drug effects , Escherichia coli Proteins/toxicity , Nucleotides, Cyclic/metabolism , Water/metabolism , Animals , Cell Line , Enterotoxigenic Escherichia coli/immunology , Epithelial Cells/metabolism , Female , Humans , Mice , Mice, Inbred BALB C
15.
Malar J ; 13: 2, 2014 Jan 03.
Article in English | MEDLINE | ID: mdl-24386891

ABSTRACT

BACKGROUND: Tafenoquine (TQ) is an 8-aminoquinoline (8AQ) that has been tested in several Phase II and Phase III clinical studies and is currently in late stage development as an anti-malarial prophylactic agent. NPC-1161B is a promising 8AQ in late preclinical development. It has recently been reported that the 8AQ drug primaquine requires metabolic activation by CYP 2D6 for efficacy in humans and in mice, highlighting the importance of pharmacogenomics in the target population when administering primaquine. A logical follow-up study was to determine whether CYP 2D activation is required for other compounds in the 8AQ structural class. METHODS: In the present study, the anti-malarial activities of NPC-1161B and TQ were assessed against luciferase expressing Plasmodium berghei in CYP 2D knock-out mice in comparison with normal C57BL/6 mice (WT) and with humanized/CYP 2D6 knock-in mice by monitoring luminescence with an in vivo imaging system. These experiments were designed to determine the direct effects of CYP 2D metabolic activation on the anti-malarial efficacy of NPC-1161B and TQ. RESULTS: NPC-1161B and TQ exhibited no anti-malarial activity in CYP 2D knock-out mice when dosed at their ED100 values (1 mg/kg and 3 mg/kg, respectively) established in WT mice. TQ anti-malarial activity was partially restored in humanized/CYP 2D6 knock-in mice when tested at two times its ED100. CONCLUSIONS: The results reported here strongly suggest that metabolism of NPC-1161B and TQ by the CYP 2D enzyme class is essential for their anti-malarial activity. Furthermore, these results may provide a possible explanation for therapeutic failures for patients who do not respond to 8AQ treatment for relapsing malaria. Because CYP 2D6 is highly polymorphic, variable expression of this enzyme in humans represents a significant pharmacogenomic liability for 8AQs which require CYP 2D metabolic activation for efficacy, particularly for large-scale prophylaxis and eradication campaigns.


Subject(s)
Aminoquinolines/metabolism , Antimalarials/metabolism , Cytochrome P-450 CYP2D6/metabolism , Malaria/drug therapy , Plasmodium berghei/drug effects , Succinates/metabolism , Animals , Cytochrome P-450 CYP2D6/genetics , Dose-Response Relationship, Drug , Malaria/parasitology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
16.
JIMD Rep ; 11: 125-32, 2013.
Article in English | MEDLINE | ID: mdl-23657977

ABSTRACT

Mucopolysaccharidosis type VI (MPS VI, Maroteaux-Lamy syndrome, MIM 253200 ) is an autosomal recessive lysosomal storage disease (LSD) caused by decreased activity of arylsulfatase B (N-acetylgalactosamine 4-sulfatase) enzyme resulting in dermatan sulfate accumulation; mucopolysaccharidosis type IVA (MPS IVA, Morquio syndrome A, MIM 253000 ) by decreased activity of N-acetylgalactosamine 6-sulfatase enzyme resulting in accumulation of keratan sulfate. Clinical symptoms include coarse facial features, joint stiffness, hepatosplenomegaly, hip osteonecrosis, and dysostosis multiplex. MPS IVA symptoms are similar but with joint hypermobility.With suspicion of MPS disease, clinicians request urine studies for quantitative and qualitative glycosaminoglycans (GAGs). Diagnosis is confirmed by decreased enzyme activity in leukocytes or cultured skin fibroblasts. Further confirmation is obtained with identification of two mutations in the ARSB gene for MPS VI or mutations in the GALNS gene for MPS IVA.We report slowly progressing patients, one with MPS VI and two with MPS IVA, who presented with skeletal changes and hip findings resembling Legg-Calvé-Perthes disease or spondyloepiphyseal dysplasia and normal/near normal urine GAG levels. The urine analysis data presented suggest that present screening techniques for MPS are inadequate in milder patients and result in delayed or missed diagnoses. The patients presented in this paper emphasize the importance of enzymatic and molecular testing.

SELECTION OF CITATIONS
SEARCH DETAIL
...