Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
2.
JCI Insight ; 6(23)2021 12 08.
Article in English | MEDLINE | ID: mdl-34609964

ABSTRACT

Controlled human malaria infection (CHMI) provides a highly informative means to investigate host-pathogen interactions and enable in vivo proof-of-concept efficacy testing of new drugs and vaccines. However, unlike Plasmodium falciparum, well-characterized P. vivax parasites that are safe and suitable for use in modern CHMI models are limited. Here, 2 healthy malaria-naive United Kingdom adults with universal donor blood group were safely infected with a clone of P. vivax from Thailand by mosquito-bite CHMI. Parasitemia developed in both volunteers, and prior to treatment, each volunteer donated blood to produce a cryopreserved stabilate of infected RBCs. Following stringent safety screening, the parasite stabilate from one of these donors (PvW1) was thawed and used to inoculate 6 healthy malaria-naive United Kingdom adults by blood-stage CHMI, at 3 different dilutions. Parasitemia developed in all volunteers, who were then successfully drug treated. PvW1 parasite DNA was isolated and sequenced to produce a high-quality genome assembly by using a hybrid assembly method. We analyzed leading vaccine candidate antigens and multigene families, including the vivax interspersed repeat (VIR) genes, of which we identified 1145 in the PvW1 genome. Our genomic analysis will guide future assessment of candidate vaccines and drugs, as well as experimental medicine studies.


Subject(s)
Genome/genetics , Malaria, Falciparum/genetics , Animals , Healthy Volunteers , Humans , Male , Plasmodium vivax
3.
Psychoneuroendocrinology ; 133: 105394, 2021 11.
Article in English | MEDLINE | ID: mdl-34474197

ABSTRACT

Preclinical models of organismal response to traumatic stress (threat of death or serious injury) can be monitored using neuroendocrine, behavioral, and structural metrics. While many rodent models of traumatic stress have provided a glimpse into select components of the physiological response to acute and chronic stressors, few studies have directly examined the potential differences between stressors and their potential outcomes. To address this gap, we conducted a multi-level comparison of the immediate and longer-term effects of two types of acute traumatic stressors. Adult male rats were exposed to either underwater trauma (UWT), predator exposure (PE), or control procedural handling conditions. Over the next 7 days, yoked cohorts underwent either serial blood sampling for neuroendocrine evaluation across the circadian cycle, or repeated behavioral testing in the elevated plus maze. In addition, a subset of brains from the latter cohort were assessed for dendritic spine changes in the prefrontal cortex and basolateral amygdala. We observed stressor-dependent patterns of response and recovery across all measures, with divergence between endocrine responses despite similar behavioral outcomes. These results demonstrate that different stressors elicit unique behavioral, neuroendocrine, and neuro-structural response profiles and suggest that specific stress models can be used to model desired responses for specific preclinical applications, such as evaluations of underlying mechanisms or therapeutic candidates.


Subject(s)
Behavior, Animal , Neurons , Neurosecretory Systems , Psychological Trauma , Stress, Psychological , Animals , Basolateral Nuclear Complex/cytology , Circadian Rhythm , Dendrites , Male , Predatory Behavior , Prefrontal Cortex/cytology , Rats
4.
ACS Infect Dis ; 7(2): 506-517, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33529014

ABSTRACT

Cutaneous leishmaniasis (CL) is the most common form of leishmaniasis affecting human populations, yet CL remains largely ignored in drug discovery programs. CL causes disfiguring skin lesions and often relapses after "clinical cure" using existing therapeutics. To expand the pool of anti-CL lead candidates, we implemented an integrated screening platform comprising three progressive Leishmania parasite life cycle forms. We identified tretazicar (CB1954, 5-(aziridin-1-yl)-2,4-dinitrobenzamide) as a potent inhibitor of Leishmania parasite viability across multiple Leishmania species, which translated into complete and prolonged in vivo suppression of CL lesion formation in BALB/c mice when used as a monotherapy and which was superior to liposomal amphotericin B. In addition, oral twice a day administration of tretazicar healed the majority of existing Leishmania major (L. major) cutaneous lesions. In drug combination studies, there was a strong potentiation when subtherapeutic doses of liposomal amphotericin B and tretazicar were simultaneously administered. This drug combination decreased L. major lesion size in mice earlier than individual monotherapy drug treatments and maintained all animals lesion free for up to 64 days after treatment cessation. In contrast, administration of subtherapeutic doses of tretazicar or amphotericin B as monotherapies resulted in no or partial lesion cures, respectively. We propose that tretazicar should be explored as a component of a systemic CL combination therapy and potentially for other diseases where amphotericin B is a first line therapy.


Subject(s)
Antiprotozoal Agents , Leishmania major , Amphotericin B , Animals , Antiprotozoal Agents/pharmacology , Aziridines , Disease Models, Animal , Mice , Mice, Inbred BALB C
5.
Malar J ; 19(1): 336, 2020 Sep 16.
Article in English | MEDLINE | ID: mdl-32938444

ABSTRACT

BACKGROUND: Malaria remains the top infectious disease threat facing the U.S. military in many forward operating environments. Compliance with malaria chemoprophylaxis remains a critical component in preventing malaria in the deployed Service Member. Studies of previous military operations show that compliance is consistently higher with weekly versus daily dosing regimens. Current FDA approved weekly chemoprophylaxis options have contraindications that can limit prescribing. The combination of chloroquine (CQ) with azithromycin (AZ) has previously been shown to be an efficacious treatment option for malaria, has pharmacokinetics compatible with weekly dosing, and has shown synergy when combined in vitro. METHODS: In this open label study, 18 healthy volunteers, aged 18-50 years (inclusive), were randomly assigned to receive either 300 mg CQ or 300 mg CQ and 2 gm azithromycin (CQAZ) of directly observed therapy, weekly for 3 weeks prior to undergoing mosquito bite challenge with chloroquine-resistant Plasmodium falciparum. Volunteers that remained asymptomatic and had no evidence of parasitaemia continued to receive weekly post-exposure chemoprophylaxis for 3 weeks following malaria challenge. The primary endpoint was the number of volunteers that remained asymptomatic and had no evidence of parasitaemia 28 days after the malaria challenge. RESULTS: All 6 (100%) volunteers randomized to the CQ control group became symptomatic with parasitaemia during the 28-day post-challenge period. Only 1/12 (8.3%) of volunteers in the CQAZ group developed symptoms and parasitaemia during the 28-day post-challenge period. However, after chemoprophylaxis was discontinued an additional 6 volunteers developed parasitaemia between days 28-41 after challenge, with 4 of 6 experiencing symptoms. 80% of subjects in the CQAZ group experienced treatment related gastrointestinal adverse events (including 13% that experienced severe nausea) compared to 38% in the CQ group. A comparison of the pharmacokinetics in the CQAZ group demonstrated higher azithromycin Cmax (p = 0.03) and AUC (p = 0.044) levels in those volunteers who never became parasitaemic compared to those who did. CONCLUSION: Given the high rate of side effects and poor efficacy when administered for 3 weeks before and after challenge, the combination of weekly chloroquine and azithromycin is a suboptimal regimen combination for weekly malaria chemoprophylaxis. Trial registration ClinicalTrials.gov NCT03278808.


Subject(s)
Antimalarials/adverse effects , Azithromycin/adverse effects , Chloroquine/adverse effects , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Adult , Drug Combinations , Drug Resistance , Female , Humans , Male , Middle Aged , Young Adult
6.
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
7.
Drug Metab Pers Ther ; 35(1)2020 03 05.
Article in English | MEDLINE | ID: mdl-32134728

ABSTRACT

Background Probiotics are live microbial organisms that provide benefit to the host while co-habitating in the gastrointestinal tract. Probiotics are safe, available over the counter, and have clinical benefit by reducing the number of antibiotic-associated diarrhea days. Prescriptions from providers and direct consumer demand of probiotics appear to be on the rise. Several recent animal studies have demonstrated that probiotics may have significant effect on absorption of co-administered drugs. However, to date, most probiotic-drug interaction studies in animal models have been limited to bacterial probiotics and nonantibiotic drugs. Methods We performed a traditional pharmacokinetic mouse study examining the interactions between a common commercially available yeast probiotic, Saccharomyces boulardii CNCM I-745 (Florastor®) and an orally administered amoxicillin. Results We showed that there were no significant differences in pharmacokinetic parameters (half-life, area under the curve, peak concentrations, time to reach maximum concentration, elimination rate constant) of amoxicillin between the probiotic treated and untreated control groups. Conclusions Altogether, our findings suggest that coadministration or concurrent use of S. boulardii probiotic and amoxicillin would not likely alter the efficacy of amoxicillin therapy.


Subject(s)
Amoxicillin/pharmacokinetics , Anti-Bacterial Agents/pharmacokinetics , Probiotics/administration & dosage , Saccharomyces boulardii/chemistry , Administration, Oral , Amoxicillin/administration & dosage , Amoxicillin/analysis , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/analysis , Dietary Supplements , Liver/chemistry , Liver/metabolism , Male , Mice , Mice, Inbred ICR
8.
Mil Med ; 185(Suppl 1): 256-262, 2020 01 07.
Article in English | MEDLINE | ID: mdl-32074328

ABSTRACT

OBJECTIVE: Infection as sequelae to explosion-related injury is an enduring threat to our troops. There are limited data on the effects of blast on antibiotic pharmacokinetics (PK), pharmacodynamics (PD), and efficacy. The observational study presented here is our Institute's first attempt to address this issue by combining our existing interdepartmental blast, infection modeling, and in vivo PK/PD capabilities and was designed to determine the PK effects of blast on the first-line antibiotic, cefazolin, in an in vivo mouse model. METHODS: A total of 160 male BALB/c mice were divided to sham and blast (exposed to blast overpressure of 19 psi) in two biological replicates. At 1 hour after blast/sham exposure, the animals received IV injection of cefazolin (328 mg/kg). Animals were euthanized at 3 minutes, 10 minutes, 15 minutes, 30 minutes, 1 hour, 3 hours, 6 hours, or 10 hours after the injection. Plasma and liver were analyzed for concentration of cefazolin using mass-spectrometry. RESULTS: We observed increases in the concentration of cefazolin in the plasma and liver of blast exposed animals at later time points and increase in elimination half-life. CONCLUSION: Our results indicate that blast-induced physiologic changes significantly influence cefazolin PK and suggest that efficacy could be affected in the context of the blast; assessment of efficacy and PD effects require further investigation. Metabolic changes resulting from blast may influence other classes of antibiotics and other therapeutics used with these injuries. Therefore, this may have important treatment considerations in other areas of military medicine.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Blast Injuries/complications , Pressure/adverse effects , Animals , Anti-Bacterial Agents/blood , Anti-Bacterial Agents/therapeutic use , Area Under Curve , Blast Injuries/blood , Blast Injuries/physiopathology , Cefazolin/blood , Cefazolin/pharmacokinetics , Cefazolin/therapeutic use , Disease Models, Animal , Explosions/statistics & numerical data , Mice , Mice, Inbred BALB C/injuries , Mice, Inbred BALB C/physiology , ROC Curve
10.
J Infect Dis ; 220(11): 1761-1770, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31549155

ABSTRACT

BACKGROUND: Plasmodium vivax malaria requires a 2-week course of primaquine (PQ) for radical cure. Evidence suggests that the hepatic isoenzyme cytochrome P450 2D6 (CYP2D6) is the key enzyme required to convert PQ into its active metabolite. METHODS: CYP2D6 genotypes and phenotypes of 550 service personnel were determined, and the pharmacokinetics (PK) of a 30-mg oral dose of PQ was measured in 45 volunteers. Blood and urine samples were collected, with PQ and metabolites were measured using ultraperformance liquid chromatography with mass spectrometry. RESULTS: Seventy-six CYP2D6 genotypes were characterized for 530 service personnel. Of the 515 personnel for whom a single phenotype was predicted, 58% had a normal metabolizer (NM) phenotype, 35% had an intermediate metabolizer (IM) phenotype, 5% had a poor metabolizer (PM) phenotype, and 2% had an ultrametabolizer phenotype. The median PQ area under the concentration time curve from 0 to ∞ was lower for the NM phenotype as compared to the IM or PM phenotypes. The novel 5,6-ortho-quinone was detected in urine but not plasma from all personnel with the NM phenotype. CONCLUSION: The plasma PK profile suggests PQ metabolism is decreased in personnel with the IM or PM phenotypes as compared to those with the NM phenotype. The finding of 5,6-ortho-quinone, the stable surrogate for the unstable 5-hydroxyprimaquine metabolite, almost exclusively in personnel with the NM phenotype, compared with sporadic or no production in those with the IM or PM phenotypes, provides further evidence for the role of CYP2D6 in radical cure. CLINICAL TRIALS REGISTRATION: NCT02960568.


Subject(s)
Antimalarials/metabolism , Cytochrome P-450 CYP2D6/genetics , Cytochrome P-450 CYP2D6/metabolism , Genotype , Primaquine/metabolism , Administration, Oral , Adolescent , Adult , Antimalarials/administration & dosage , Antimalarials/pharmacokinetics , Blood Chemical Analysis , Chromatography, High Pressure Liquid , Cohort Studies , Female , Humans , Male , Mass Spectrometry , Middle Aged , Military Personnel , Phenotype , Plasma/chemistry , Primaquine/administration & dosage , Primaquine/pharmacokinetics , United States , Urinalysis , Urine/chemistry , Young Adult
11.
Mil Med ; 184(Suppl 1): 106-113, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30901419

ABSTRACT

Particle size is an important determinant of gastrointestinal absorption of compounds administrated orally. The present study evaluates the effect of a reduction in particle size assessed by homogenization, sonication, and homogenization plus sonication on the bioavailability of imidazolidinedione (IZ), an antimalarial compound with known causal prophylactic activity and radical cure of relapsing malaria. Formulations were administrated intragastrically to mice, and blood samples were collected for LC-MS/MS analysis. The homogenization method manually decreased particle size with minimal variance, resulting in a mean particle diameter of 42.22 µm, whereas the probe sonication method evenly distributed pulses of sound to break apart particles, resulting in a mean diameter of 1.50 µm. Homogenization plus sonication resulted in a mean particle diameter of 1.44 µm, which was similar to that of the sonication method alone. The compound suspensions did not show a significant difference in mean particle size between the different vehicles. The sonically engineered microparticle delivers high sonic energy to the suspension leads to faster breakdown and stabilizing of the micronized particles when compared with homogenizer. The bioavailability of the small particle IZ formulation was 100%, compared to the 55.79% relative bioavailability of IZ with larger particle size. These initial data clearly show that a reduction in particle size of orally administered IZ with probe sonication could significantly increase bioavailability in rodent animals that is affected by a high first-pass effect.


Subject(s)
Biological Availability , Imidazolidines/pharmacokinetics , Sonication/methods , Humans , Imidazolidines/metabolism , Imidazolidines/therapeutic use , Particle Size
12.
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
13.
Stress ; 21(3): 267-273, 2018 05.
Article in English | MEDLINE | ID: mdl-29451058

ABSTRACT

Adolescence is a distinct developmental period characterized by behavioral and physiological maturation. Rapid ongoing changes during neurodevelopment in particular present potential opportunities for stress to have lasting effects on longitudinal outcomes of behavioral and neuroendocrine function. While adult stress effects on outcomes during adulthood have been characterized, little is known about the lasting effects of adolescent repeated stressor exposure on outcomes during adolescence. We have previously reported different stress responses in adolescent rats relative to adult rats, including a blunted fear response outcome in adulthood in rats stressed during adolescence. The present study characterized the ontogeny of behavioral and neuroendocrine responses to eight underwater trauma (UWT) exposures in rats over a two week poststress time period during adolescence (P34) or adulthood (P83) relative to age-matched control groups that underwent eight swimming episodes without UWT. Repeated UWT exposures starting in adolescence, but not adulthood, resulted in adverse behavioral responses on the elevated plus maze 1 day post-stress. Corticosterone responses did not differ between UWT-exposed and controls for either age group at 1 day or at 7 days poststress, although there was an effect of age on corticosterone levels. We conclude that repeated UWT stress events have a lasting, negative behavioral effect on adolescent rats that is not observed in adult rats after the two-week exposure window. These results suggest that neurophysiological mechanisms underlying recovery from a repeated stressor are immature in adolescence relative to adulthood in rats.


Subject(s)
Immersion/physiopathology , Stress, Psychological/psychology , Wounds and Injuries/physiopathology , Aging/psychology , Animals , Anxiety/psychology , Behavior, Animal , Corticosterone/blood , Exploratory Behavior , Rats , Rats, Sprague-Dawley , Reflex, Startle , Swimming/psychology , Water
14.
J Infect Dis ; 217(5): 693-702, 2018 02 14.
Article in English | MEDLINE | ID: mdl-29216395

ABSTRACT

Background: DSM265 is a selective inhibitor of Plasmodium dihydroorotate dehydrogenase that fully protected against controlled human malarial infection (CHMI) by direct venous inoculation of Plasmodium falciparum sporozoites when administered 1 day before challenge and provided partial protection when administered 7 days before challenge. Methods: A double-blinded, randomized, placebo-controlled trial was performed to assess safety, tolerability, pharmacokinetics, and efficacy of 1 oral dose of 400 mg of DSM265 before CHMI. Three cohorts were studied, with DSM265 administered 3 or 7 days before direct venous inoculation of sporozoites or 7 days before 5 bites from infected mosquitoes. Results: DSM265-related adverse events consisted of mild-to-moderate headache and gastrointestinal symptoms. DSM265 concentrations were consistent with pharmacokinetic models (mean area under the curve extrapolated to infinity, 1707 µg*h/mL). Placebo-treated participants became positive by quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and were treated 7-10 days after CHMI. Among DSM265-treated subjects, 2 of 6 in each cohort were sterilely protected. DSM265-treated recipients had longer times to development of parasitemia than placebo-treated participants (P < .004). Conclusions: This was the first CHMI study of a novel antimalarial compound to compare direct venous inoculation of sporozoites and mosquito bites. Times to qRT-PCR positivity and treatment were comparable for both routes. DSM265 given 3 or 7 days before CHMI was safe and well tolerated but sterilely protected only one third of participants.


Subject(s)
Antimalarials/administration & dosage , Chemoprevention/methods , Malaria, Falciparum/prevention & control , Pyrimidines/administration & dosage , Triazoles/administration & dosage , Adolescent , Adult , Animals , Antimalarials/adverse effects , Antimalarials/pharmacokinetics , Double-Blind Method , Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/pathology , Female , Humans , Male , Middle Aged , Parasitemia/prevention & control , Placebos/administration & dosage , Plasmodium falciparum/genetics , Plasmodium falciparum/isolation & purification , Pyrimidines/adverse effects , Pyrimidines/pharmacokinetics , Real-Time Polymerase Chain Reaction , Treatment Outcome , Triazoles/adverse effects , Triazoles/pharmacokinetics , Young Adult
15.
Ann Pharmacother ; 52(3): 251-256, 2018 03.
Article in English | MEDLINE | ID: mdl-29047306

ABSTRACT

BACKGROUND: There is no established method for monitoring the anticoagulant effects of apixaban and rivaroxaban. Linear correlation between serum levels and anti-Xa activity has been shown, with r2 ranging from 0.88 to 0.99. However, there are minimal data in patients receiving apixaban 5 mg twice daily or rivaroxaban 20 mg once daily. OBJECTIVE: To evaluate the anti-Xa activity and serum levels at those doses and compare the trough anti-Xa activity. METHODS: This was a single-center prospective study,approved by the institutional review board. Patients on an inappropriate dose or receiving an interacting drug were excluded. Blood samples were drawn 0.5 to 3 hours before a dose for both agents, 2 to 3 hours after an apixaban dose, and 12 to 16 hours after a rivaroxaban dose. Anti-Xa activity and serum levels were determined, and correlation was done via regression analysis. Trough anti-Xa activity was compared using a t-test. RESULTS: The study enrolled 88 patients receiving each drug. The r2 values were 0.79 and 0.87 for apixaban and rivaroxaban, respectively. The mean trough anti-Xa activity was 1.79 ± 0.96 IU/mL for apixaban and 1.25 ± 0.88 IU for rivaroxaban ( P < 0.01). The trough sample was drawn a mean of 1.3 and 1.8 hours prior to the next dose for apixaban and rivaroxaban, respectively ( P < 0.01). CONCLUSIONS: Good correlation was shown between anti-Xa activity and serum levels. The clinical utility of monitoring anti-Xa activity and the significance of the difference in trough anti-Xa activity for these agents remains to be established.


Subject(s)
Factor Xa Inhibitors/blood , Factor Xa/analysis , Pyrazoles/blood , Pyridones/blood , Rivaroxaban/blood , Aged , Factor Xa Inhibitors/pharmacokinetics , Factor Xa Inhibitors/therapeutic use , Female , Humans , Male , Middle Aged , Pyrazoles/pharmacokinetics , Pyrazoles/therapeutic use , Pyridones/pharmacokinetics , Pyridones/therapeutic use , Rivaroxaban/pharmacokinetics , Rivaroxaban/therapeutic use
16.
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.

17.
J Neurotrauma ; 34(19): 2768-2789, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28326890

ABSTRACT

Closed-head concussive injury is one of the most common causes of traumatic brain injury (TBI). Isolated concussions frequently produce acute neurological impairments, and individuals typically recover spontaneously within a short time frame. In contrast, brain injuries resulting from multiple concussions can result in cumulative damage and elevated risk of developing chronic brain pathologies. Increased attention has focused on identification of diagnostic markers that can prognostically serve as indices of brain health after injury, revealing the temporal profile of vulnerability to a second insult. Such markers may demarcate adequate recovery periods before concussed patients can return to required activities. We developed a noninvasive closed-head impact model that captures the hallmark symptoms of concussion in the absence of gross tissue damage. Animals were subjected to single or repeated concussive impact and examined using a battery of neurological, vestibular, sensorimotor, and molecular metrics. A single concussion induced transient, but marked, acute neurological impairment, gait alterations, neuronal death, and increased glial fibrillary acidic protein (GFAP) expression in brain tissue. As expected, repeated concussions exacerbated sensorimotor dysfunction, prolonged gait abnormalities, induced neuroinflammation, and upregulated GFAP and tau. These animals also exhibited chronic functional neurological impairments with sustained astrogliosis and white matter thinning. Acute changes in molecular signatures correlated with behavioral impairments, whereas increased times to regaining consciousness and balance impairments were associated with higher GFAP and neuroinflammation. Overall, behavioral consequences of either single or repeated concussive impact injuries appeared to resolve more quickly than the underlying molecular, metabolic, and neuropathological abnormalities. This observation, which is supported by similar studies in other mTBI models, underscores the critical need to develop more objective prognostic measures for guiding return-to-play decisions.


Subject(s)
Brain Concussion , Disease Models, Animal , Animals , Brain Concussion/complications , Brain Concussion/pathology , Brain Concussion/physiopathology , Male , Rats , Rats, Sprague-Dawley
18.
Antimicrob Agents Chemother ; 60(10): 5906-13, 2016 10.
Article in English | MEDLINE | ID: mdl-27458212

ABSTRACT

Individuals with glucose-6-phosphate dehydrogenase (G6PD) deficiency (G6PDd) are at risk for developing hemolytic anemia when given the antimalarial drug primaquine (PQ). The WHO Evidence Review Group released a report suggesting that mass administration of a single dose of PQ at 0.25 mg of base/kg of body weight (mpk) (mouse equivalent of 3.125 mpk) could potentially reduce malaria transmission based on its gametocytocidal activity and could be safely administered to G6PD-deficient individuals, but there are limited safety data available confirming the optimum single dose of PQ. A single-dose administration of PQ was therefore assessed in our huRBC-SCID mouse model used to predict hemolytic toxicity with respect to G6PD deficiency. In this model, nonobese diabetic (NOD)/SCID mice are engrafted with human red blood cells (huRBC) from donors with the African or Mediterranean variant of G6PDd (A-G6PDd or Med-G6PDd, respectively) and demonstrate dose-dependent sensitivity to PQ. In mice engrafted with A-G6PD-deficient huRBC, single-dose PQ at 3.125, 6.25, or 12.5 mpk had no significant loss of huRBC compared to the vehicle control group. In contrast, in mice engrafted with Med-G6PDd huRBC, a single dose of PQ at 3.125, 6.25, or 12.5 mpk resulted in a significant, dose-dependent loss of huRBC compared to the value for the vehicle control group. Our data suggest that administration of a single low dose of 0.25 mpk of PQ could induce hemolytic anemia in Med-G6PDd individuals but that use of single-dose PQ at 0.25 mpk as a gametocytocidal drug to block transmission would be safe in areas where A-G6PDd predominates.


Subject(s)
Antimalarials/administration & dosage , Glucosephosphate Dehydrogenase Deficiency/parasitology , Malaria/transmission , Primaquine/administration & dosage , Animals , Antimalarials/pharmacokinetics , Disease Models, Animal , Erythrocyte Transfusion , Erythrocytes/drug effects , Humans , Mice, SCID , Primaquine/analogs & derivatives , Primaquine/pharmacokinetics
19.
Malar J ; 15(1): 280, 2016 05 17.
Article in English | MEDLINE | ID: mdl-27188854

ABSTRACT

BACKGROUND: The liver-stage anti-malarial activity of primaquine and other 8-aminoquinoline molecules has been linked to bio-activation through CYP 2D6 metabolism. Factors such as CYP 2D6 poor metabolizer status and/or co-administration of drugs that inhibit/interact with CYP 2D6 could alter the pharmacological properties of primaquine. METHODS: In the present study, the inhibitory potential of the selective serotonin reuptake inhibitor (SSRI) and serotonin norepinephrine reuptake inhibitor (SNRI) classes of antidepressants for CYP 2D6-mediated primaquine metabolism was assessed using in vitro drug metabolism and in vivo pharmacological assays. RESULTS: The SSRI/SNRI classes of drug displayed a range of inhibitory activities on CYP 2D6-mediated metabolism of primaquine in vitro (IC50 1-94 µM). Fluoxetine and paroxetine were the most potent inhibitors (IC50 ~1 µM) of CYP 2D6-mediated primaquine metabolism, while desvenlafaxine was the least potent (IC50 ~94 µM). The most potent CYP 2D6 inhibitor, fluoxetine, was chosen to investigate the potential pharmacological consequences of co-administration with primaquine in vivo. The pharmacokinetics of a CYP 2D6-dependent primaquine metabolite were altered upon co-administration with fluoxetine. Additionally, in a mouse malaria model, co-administration of fluoxetine with primaquine reduced primaquine anti-malarial efficacy. CONCLUSIONS: These results are the first from controlled pre-clinical experiments that indicate that primaquine pharmacological properties can be modulated upon co-incubation/administration with drugs that are known to interact with CYP 2D6. These results highlight the potential for CYP 2D6-mediated drug-drug interactions with primaquine and indicate that the SSRI/SNRI antidepressants could be used as probe molecules to address the primaquine-CYP 2D6 DDI link in clinical studies. Additionally, CYP 2D6-mediated drug-drug interactions can be considered when examining the possible causes of human primaquine therapy failures.


Subject(s)
Antidepressive Agents/pharmacokinetics , Antimalarials/pharmacokinetics , Cytochrome P-450 CYP2D6/metabolism , Drug Interactions , Primaquine/pharmacokinetics , Serotonin and Noradrenaline Reuptake Inhibitors/pharmacokinetics , Animals , Antidepressive Agents/administration & dosage , Antidepressive Agents/metabolism , Antimalarials/administration & dosage , Antimalarials/metabolism , Cells, Cultured , Disease Models, Animal , Female , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Inhibitory Concentration 50 , Malaria/drug therapy , Male , Mice , Mice, Inbred C57BL , Primaquine/administration & dosage , Primaquine/metabolism , Serotonin and Noradrenaline Reuptake Inhibitors/administration & dosage , Serotonin and Noradrenaline Reuptake Inhibitors/metabolism , Treatment Outcome
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...