Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Hum Gene Ther Clin Dev ; 25(1): 16-27, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24649838

ABSTRACT

Herpes simplex virus type 1 (HSV-1) mutants lacking the γ(1)34.5 neurovirulence loci are promising agents for treating malignant glioma. Arming oncolytic HSV-1 to express immunostimulatory genes may potentiate therapeutic efficacy. We have previously demonstrated improved preclinical efficacy, biodistribution, and safety of M002, a γ(1)34.5-deleted HSV-1 engineered to express murine IL-12. Herein, we describe the safety and biodistribution of M032, a γ(1)34.5-deleted HSV-1 virus that expresses human IL-12 after intracerebral administration to nonhuman primates, Aotus nancymae. Cohorts were administered vehicle, 10(6), or 10(8) pfu of M032 on day 1 and subjected to detailed clinical observations performed serially over a 92-day trial. Animals were sacrificed on days 3, 31, and 91 for detailed histopathologic assessments of all organs and to isolate and quantify virus in all organs. With the possible exception of one animal euthanized on day 16, neither adverse clinical signs nor sex- or dose-related differences were attributed to M032. Elevated white blood cell and neutrophil counts were observed in virus-injected groups on day 3, but no other significant changes were noted in clinical chemistry or coagulation parameters. Minimal to mild inflammation and fibrosis detected, primarily in meningeal tissues, in M032-injected animals on days 3 and 31 had mostly resolved by day 91. The highest viral DNA levels were detected at the injection site and motor cortex on day 3 but decreased in central nervous system tissues over time. These data demonstrate the requisite safety of intracerebral M032 administration for consideration as a therapeutic for treating malignant brain tumors.


Subject(s)
Glioma/therapy , Herpesvirus 1, Human/genetics , Infusions, Intraventricular , Interleukin-12/genetics , Oncolytic Virotherapy/methods , Animals , Aotidae , Brain Neoplasms/therapy , Drug Administration Routes , Female , Interleukin-12/biosynthesis , Male , Virus Replication
2.
Toxicology ; 293(1-3): 30-40, 2012 Mar 11.
Article in English | MEDLINE | ID: mdl-22239858

ABSTRACT

In a prior 28-day dietary study in rats with 20 and 100 ppm K⁺ PFOS, activation of PPARα and CAR/PXR were concluded to be etiological factors in K⁺ PFOS-induced hepatomegaly and hepatic tumorigenesis. The objective of this study was to evaluate persistence/resolution of K⁺ PFOS-induced, liver-related effects in male Sprague Dawley rats following a 7-day dietary exposure to K⁺ PFOS at 20 or 100 ppm. Groups of 10 rats per treatment were observed on recovery Day(s) 1, 28, 56, and 84 following treatment. Changes consistent with hepatic PPARα and CAR/PXR activation noted on recovery Day 1 included: increased liver weight; decreased plasma cholesterol, alanine aminotransferase, and triglycerides; decreased liver DNA concentration and increased hepatocellular cytosolic CYP450 concentration; increased liver activity of acyl CoA oxidase, CYP4A, CYP2B, and CYP3A; increased liver proliferative index and decreased liver apoptotic index; decreased hepatocellular glycogen-induced vacuoles; increased centrilobular hepatocellular hypertrophy. Most effects resolved to control levels during recovery. Effects on plasma cholesterol, hepatocellular cytosolic CYP450 concentrations, liver apoptotic index, CYP3A, and centrilobular hepatocellular hypertrophy persisted through the end of the recovery period. Thyroid parameters (histology, apoptosis, and proliferation) were unaffected at all time points. Mean serum PFOS concentrations on recovery Day 1 were 39 and 140 µg/mL (20 ppm and 100 ppm K⁺ PFOS, respectively), decreasing to 4 and 26 µg/mL by recovery Day 84. Thus, hepatic effects in male rats resulting from K⁺ PFOS-induced activation of PPARα and CAR/PXR resolved slowly or were still present after 84-days following a 7-day dietary treatment, consistent with the slow elimination rate of PFOS.


Subject(s)
Alkanesulfonic Acids/pharmacokinetics , Alkanesulfonic Acids/toxicity , Carcinogens/pharmacokinetics , Carcinogens/toxicity , Fluorocarbons/pharmacokinetics , Fluorocarbons/toxicity , Hepatomegaly/chemically induced , Liver/drug effects , Thyroid Gland/drug effects , Alkanesulfonic Acids/administration & dosage , Alkanesulfonic Acids/metabolism , Animals , Apoptosis/drug effects , Carcinogens/administration & dosage , Carcinogens/metabolism , Cell Proliferation/drug effects , Constitutive Androstane Receptor , Cytochrome P-450 Enzyme System/metabolism , Dose-Response Relationship, Drug , Environmental Pollutants/administration & dosage , Environmental Pollutants/metabolism , Environmental Pollutants/pharmacokinetics , Environmental Pollutants/toxicity , Fluorocarbons/administration & dosage , Fluorocarbons/metabolism , Half-Life , Hepatomegaly/blood , Hepatomegaly/metabolism , Hepatomegaly/pathology , Liver/metabolism , Liver/pathology , Male , PPAR alpha/metabolism , Pregnane X Receptor , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/metabolism , Thyroid Gland/pathology , Toxicity Tests, Subacute
3.
Reprod Toxicol ; 33(4): 441-451, 2012 Jul.
Article in English | MEDLINE | ID: mdl-21856411

ABSTRACT

Perfluorohexanesulfonate (PFHxS) has been found in biological samples from wildlife and humans. The human geometric mean serum PFHxS elimination half-life has been estimated to be 2665days. A series of studies was undertaken to establish pharmacokinetic parameters for PFHxS in rats, mice, and monkeys after single administration with pharmacokinetic parameters determined by WinNonlin(®) software. Rats and mice appeared to be more effective at eliminating PFHxS than monkeys. With the exception of female rats, which had serum PFHxS elimination half-life of approximately 2 days, the serum elimination half-lives in the rodent species and monkeys approximated 1month and 4months, respectively, when followed over extended time periods (10-24weeks). Collectively, these studies provide valuable insight for human health risk assessment regarding the potential for accumulation of PFHxS in humans.


Subject(s)
Environmental Pollutants/pharmacokinetics , Sulfonic Acids/pharmacokinetics , Administration, Oral , Animals , Dose-Response Relationship, Drug , Environmental Pollutants/blood , Environmental Pollutants/urine , Feces/chemistry , Female , Fluorocarbons , Half-Life , Injections, Intravenous , Liver/metabolism , Macaca fascicularis , Male , Metabolic Clearance Rate , Mice , Mice, Inbred Strains , Rats , Rats, Sprague-Dawley , Sex Factors , Species Specificity , Sulfonic Acids/blood , Sulfonic Acids/urine , Tissue Distribution
4.
Reprod Toxicol ; 33(4): 428-440, 2012 Jul.
Article in English | MEDLINE | ID: mdl-21889587

ABSTRACT

Perfluorooctanesulfonate (PFOS) has been found in biological samples in wildlife and humans. The geometric mean half-life of serum elimination of PFOS in humans has been estimated to be 4.8 years (95% CI, 4.0-5.8). A series of studies was undertaken to establish pharmacokinetic parameters for PFOS in rats, mice, and monkeys after single oral and/or IV administration of K(+)PFOS. Animals were followed for up to 23 weeks, and pharmacokinetic parameters were determined by WinNonlin® software. Rats and mice appeared to be more effective at eliminating PFOS than monkeys. The serum elimination half-lives in the rodent species were on the order of 1-2 months; whereas, in monkeys, the serum elimination half lives approximated 4 months. Collectively, these studies provide valuable insight for human health risk assessment regarding the potential for accumulation of body burden in humans on repeated exposure to PFOS and PFOS-generating materials.


Subject(s)
Alkanesulfonic Acids/pharmacokinetics , Environmental Pollutants/pharmacokinetics , Fluorocarbons/pharmacokinetics , Administration, Oral , Alkanesulfonic Acids/blood , Alkanesulfonic Acids/urine , Animals , Environmental Pollutants/blood , Environmental Pollutants/urine , Feces/chemistry , Female , Fluorocarbons/blood , Fluorocarbons/urine , Half-Life , Injections, Intravenous , Liver/metabolism , Macaca fascicularis , Male , Metabolic Clearance Rate , Mice , Mice, Inbred Strains , Rats , Rats, Sprague-Dawley , Species Specificity , Tissue Distribution
5.
Cancer Chemother Pharmacol ; 68(2): 513-24, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21085965

ABSTRACT

PURPOSE: Characterize the preclinical pharmacokinetics, metabolic profile, multi-species toxicology, and antitumor efficacy of azurin-p28 (NSC 745104), an amphipathic, 28 amino acid fragment (aa 50-77) of the copper containing redox protein azurin that preferentially enters cancer cells and is currently under development for treatment of p53-positive solid tumors. METHODS: An LC/MS/MS assay was developed, validated, and applied to liver microsomes, serum, and tumor cells to assess cellular uptake and metabolic stability. Pharmacokinetics was established after administration of a single intravenous dose of p28 in preclinical species undergoing chronic toxicity testing. Antitumor efficacy was assessed on human tumor xenografts. A human therapeutic dose was predicted based on efficacy and pharmacokinetic parameters. RESULTS: p28 is stable, showed tumor penetration consistent with selective entry into tumor cells and significantly inhibited p53-positive tumor growth. Renal clearance, volume of distribution, and metabolic profile of p28 was relatively similar among species. p28 was non-immunogenic and non-toxic in mice and non-human primates (NHP). The no observed adverse effect level (NOAEL) was 120 mg/kg iv in female mice. A NOAEL was not established for male mice due to decreased heart and thymus weights that was reversible and did not result in limiting toxicity. In contrast, the NOAEL for p28 in NHP was defined as the highest dose (120 mg/kg/dose; 1,440 mg/m(2)/dose) studied. The maximum-tolerated dose (MTD) for subchronic administration of p28 to mice is >240 mg/kg/dose (720 mg/m(2)/dose), while the MTD for subchronic administration of p28 to Cynomolgous sp. is >120 mg/kg (1,440 mg/m(2)/dose). The efficacious (murine) dose of p28 was 10 mg/kg ip per day. CONCLUSIONS: p28 does not exhibit preclinical immunogenicity or toxicity, has a similar metabolic profile among species, and is therapeutic in xenograft models.


Subject(s)
Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Azurin/adverse effects , Azurin/pharmacokinetics , Neoplasms/drug therapy , Peptide Fragments/pharmacokinetics , Tumor Suppressor Protein p53/antagonists & inhibitors , Animals , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Azurin/metabolism , Azurin/therapeutic use , Biotransformation , Cell Line, Tumor , Drug Evaluation, Preclinical , Female , Humans , Macaca fascicularis , Male , Mice , Mice, Nude , No-Observed-Adverse-Effect Level , Peptide Fragments/adverse effects , Peptide Fragments/metabolism , Peptide Fragments/therapeutic use , Specific Pathogen-Free Organisms , Tumor Burden/drug effects , Tumor Suppressor Protein p53/metabolism , Ubiquitination/drug effects , Xenograft Model Antitumor Assays
6.
Clin Cancer Res ; 15(12): 4131-7, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19509153

ABSTRACT

PURPOSE: The purpose of this study was to evaluate the biodistribution and toxicity of Ad5.SSTR/TK.RGD, an infectivity-enhanced adenovirus expressing a therapeutic suicide gene and somatostatin receptor type 2 (for noninvasive assessment of gene transfer with nuclear imaging) in advance of a planned phase I clinical trial for recurrent ovarian carcinoma. EXPERIMENTAL DESIGN: Cohorts of Syrian hamsters were treated i.p. for 3 consecutive days with Ad5.SSTR/TK.RGD or control buffer with or without the prodrug ganciclovir (GCV) and euthanized on day 4, 19, or 56. Tissue and serum samples were evaluated for the presence of virus using qPCR analysis and were assessed for vector-related tissue or laboratory effects. RESULTS: Levels of Ad5.SSTR/TK.RGD in blood and tissues outside of the abdominal cavity were low, indicating minimal systemic absorption. GCV did not affect Ad5.SSTR/TK.RGD biodistribution. The mean Ad5.SSTR/TK.RGD viral level was 100-fold lower on day 19 than day 4, suggesting vector elimination over time. Animals in the Ad5.SSTR/TK.RGD +/- GCV cohort had clinical laboratory parameters and microscopic lesions in the abdominal organs indicative of an inflammatory response. Toxicity in this dose cohort seemed to be reversible over time. CONCLUSIONS: These studies provide justification for planned dosing of Ad5.SSTR/TK.RGD for a planned phase I clinical trial and insights regarding anticipated toxicity.


Subject(s)
Adenoviridae/metabolism , Genetic Therapy/adverse effects , Genetic Vectors/adverse effects , Genetic Vectors/metabolism , Adenoviridae/genetics , Animals , Clinical Trials, Phase I as Topic , Cricetinae , Female , Ganciclovir/pharmacology , Gene Transfer Techniques , Genetic Vectors/genetics , Mesocricetus , Receptors, Somatostatin/metabolism
7.
Toxicology ; 256(1-2): 65-74, 2009 Feb 04.
Article in English | MEDLINE | ID: mdl-19059455

ABSTRACT

Materials derived from perfluorobutanesulfonyl fluoride (PBSF, C(4)F(9)SO(2)F) have been introduced as replacements for eight-carbon homolog products that were manufactured from perfluorooctanesulfonyl fluoride (POSF, C(8)F(17)SO(2)F). Perfluorobutanesulfonate (PFBS, C(4)F(9)SO(3)(-)) is a surfactant and potential degradation product of PBSF-derived materials. The purpose of this series of studies was to evaluate the pharmacokinetics of PFBS in rats, monkeys, and humans, thereby providing critical information for human health risk assessment. Studies included: (1) intravenous (i.v.) elimination studies in rats and monkeys; (2) oral uptake and elimination studies in rats; and (3) human serum PFBS elimination in a group of workers with occupational exposure to potassium PFBS (K(+)PFBS). PFBS concentrations were determined in serum (all species), liver (rats), urine (all species), and feces (rats). In rats, the mean terminal serum PFBS elimination half-lives, after i.v. administration of 30mg/kg PFBS, were: males 4.51+/-2.22h (standard error) and females 3.96+/-0.21h. In monkeys, the mean terminal serum PFBS elimination half-lives, after i.v. administration of 10mg/kg PFBS, were: males 95.2+/-27.1h and females 83.2+/-41.9h. Although terminal serum half-lives in male and female rats were similar, without statistical significance, clearance (CL) was significantly greater in female rats (469+/-40mL/h) than male rats (119+/-34mL/h) with the area under the curve (AUC) significantly larger in male rats (294+/-77microg.h/mL) than female rats (65+/-5microg.h/mL). These differences were not observed in male and female monkeys. Volume of distribution estimates suggested distribution was primarily extracellular in both rats and monkeys, regardless of sex, and urine appeared to be a major route of elimination. Among 6 human subjects (5 male, 1 female) followed up to 180 days, the geometric mean serum elimination half-life for PFBS was 25.8 days (95% confidence interval 16.6-40.2). Urine was observed to be a pathway of elimination in the human. Although species-specific differences exist, these findings demonstrate that PFBS is eliminated at a greater rate from human serum than the higher chain homologs of perfluorooctanesulfonate (PFOS) and perfluorohexanesulfonate (PFHxS). Thus, compared to PFOS and PFHxS, PFBS has a much lower potential for accumulation in human serum after repeated occupational, non-occupational (e.g., consumer), or environmental exposures.


Subject(s)
Fluorocarbons/pharmacokinetics , Administration, Oral , Animals , Chromatography, High Pressure Liquid , Data Interpretation, Statistical , Feces/chemistry , Female , Fluorocarbons/administration & dosage , Half-Life , Humans , Injections, Intravenous , Macaca fascicularis , Male , Mass Spectrometry , Rats , Rats, Sprague-Dawley , Species Specificity
8.
J Pharm Pharmacol ; 60(11): 1441-8, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18957164

ABSTRACT

Phor21-betaCG(ala), a 36-amino acid peptide comprised of a lytic peptide (Phor21) conjugated to a modified 15-amino acid segment of the beta-chain of chorionic gonadotropin (betaCG(ala)), selectively kills cancer cells that over-express luteinizing hormone/chorionic gonadotropin (LH/CG) receptors by disrupting cellular membrane structure. These studies were designed to further characterize its in-vitro inhibition and in-vivo destruction of prostate cancer cells, biostability and pharmacokinetics to determine its pharmacokinetic and pharmacodynamic profile. Inhibitory effects of Phor21-betaCG(ala) were tested in PC-3 and Caco-2 cells as well as in nude mice bearing PC-3 cells transfected with the luciferase gene (PC-3.luc). Plasma stability, protease hydrolysis and pharmacokinetics of Phor21-betaCG(ala) were measured by using liquid chromatography mass spectrometry (LC/MS/MS). Phor21-betaCG(ala) selectively inhibited proliferation in-vitro and in-vivo metastases of PC-3 cells. Phor21-betaCG(ala) was relatively stable in mouse, rat, dog and human plasma. Its degradation was partially due to protease hydrolysis and thermodynamic catalysis. Intravenous administration of Phor21-betaCG(ala) showed its blood C(max) and AUC(0-->infinity) around the in-vitro effective levels. In the tested rodents, Phor21-betaCG(ala) displayed a moderate volume of distribution at steady state (Vd(ss)) and slow clearance (Cl) in the rodents. In conclusion, Phor21-betaCG(ala) displayed promising in-vitro and in-vivo anti-cancer activity with favourable pharmacokinetics, and may offer a novel approach to metastatic cancer chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Peptides/pharmacology , Prostatic Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacokinetics , Area Under Curve , Caco-2 Cells , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatography, Liquid , Dogs , Humans , Luciferases/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Neoplasm Metastasis/drug therapy , Peptides/pharmacokinetics , Rats , Rats, Inbred F344 , Species Specificity , Tandem Mass Spectrometry , Tissue Distribution , Transfection
9.
Toxicol Sci ; 104(1): 40-53, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18353799

ABSTRACT

Perfluorobutyrate (PFBA) has been detected in precipitation, surface waters, water treatment effluent, and in public and private wells in Minnesota at up to low microg/l concentrations. We evaluated the pharmacokinetics of PFBA in rats, mice, monkeys, and humans to provide a rational basis for dose selection in toxicological studies and to aid in human-health-risk assessment. Studies included (1) rats--iv and oral; (2) mice--oral; (3) monkeys--iv; and (4) humans--occupationally exposed volunteers. PFBA was determined in serum (all species), liver (rats and mice), urine (rats, mice, and monkeys), and feces (rats and mice). In addition, we characterized serum PFBA concentrations in 177 individuals with potential exposure to PFBA through drinking water. Mean terminal serum PFBA elimination half-lives for males (M) and females (F), respectively, in h were (1) for rats given 30 mg/kg, 9.22 and 1.76 (oral), and 6.38 and 1.03 (iv); (2) for mice given oral doses of 10, 30, or 100 mg/kg ammonium PFBA, 13.34 and 2.87 at 10 mg/kg, 16.25 and 3.08 at 30 mg/kg; and 5.22 and 2.79 at 100 mg/kg; (3) for monkeys given 10 mg/kg iv, 40.32 and 41.04; and (4) for humans, 72.16 and 87.00 (74.63 combined). Volume of distribution estimates indicated primarily extracellular distribution. Among individuals with plausible exposure via drinking water, 96% of serum PFBA concentrations were < 2 ng/ml (maximum 6 ng/ml). These findings demonstrate that PFBA is eliminated efficiently from serum with a low potential for accumulation from repeated exposure.


Subject(s)
Fluorocarbons/pharmacokinetics , Occupational Exposure/analysis , Water Pollutants, Chemical/pharmacokinetics , Water Supply , Animals , Environmental Monitoring , Feces/chemistry , Female , Fluorocarbons/blood , Fluorocarbons/urine , Haplorhini , Humans , Liver/metabolism , Macaca fascicularis , Male , Mice , Rats , Rats, Sprague-Dawley , Risk Assessment , Water Pollutants, Chemical/blood , Water Pollutants, Chemical/urine
10.
Cancer Chemother Pharmacol ; 61(1): 63-73, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17356822

ABSTRACT

PURPOSE: To characterize the stability, pharmacokinetics and metabolism of analogs of gossypol, apogossypol and apogossypol hexaacetate to provide a basis for comparison. METHODS: Gossypol, apogossypol and apogossypol hexaacetate were incubated in plasma or liver microsomes from various species, or administered to mice, respectively, from which the stability, metabolism and pharmacokinetic profiles of these analogs were quantitatively determined using a liquid chromatography-mass spectrometry (LC/MS/MS) method. RESULTS: In various species of plasma, apogossypol and gossypol exhibited similar stability, while 20-40% of apogossypol hexaacetate was converted into apogossypol with concurrent formation of the corresponding di-, tri-, tetra-, and penta-acetates of apogossypol. (+/-)-Gossypol and (-)-gossypol showed comparable pharmacokinetic profile and oral bioavailability (12.2-17.6%) with some variations of clearance and V (ss) following oral and intravenous administration to mice. At the same molar dose, apogossypol showed delayed T (max)(1 h), a slower clearance rate and less distribution after administration to mice. Mono- and di-glucuronide conjugates of apogossypol were readily observed in mouse plasma following administration. Apogossypol formulated in sesame oil appeared to possess larger AUC and thus higher oral bioavailability than that formulated in cremophor EL:ethanol:saline. In contrast, intravenous apogossypol hexaacetate exhibited highest clearance rate partially due to its conversion into apogossypol. Concomitant with disappearance of apogossypol hexaacetate (iv), apogossypol converted from apogossypol hexaacetate was quantitatively detected, and accounted for approximately 30% of total plasma apogossypol hexaacetate. Oral apogossypol hexaacetate showed no bioavailability with little apogossypol occurring in the plasma. In human and mouse liver microsomes, glucuronide conjugates of apogossypol and its acetates were readily identified with the exception of gossypol glucuronidation. Apogossypol appeared more stable in human and mouse liver microsomal preparations than gossypol and apogossypol hexaacetate. CONCLUSIONS: Apogossypol and gossypol show similar oral and intravenous pharmacokinetic profiles and in vitro stability although apogossypol appears to have a slower clearance rate, larger AUC, and better microsomal stability. Apogossypol hexaacetate converts to apogossypol in both in vitro and in vivo settings and lacks any quantifiable oral bioavailability.


Subject(s)
Acetates/pharmacokinetics , Contraceptive Agents, Male/pharmacokinetics , Gossypium/chemistry , Gossypol/analogs & derivatives , Gossypol/pharmacokinetics , Administration, Oral , Animals , Area Under Curve , Biological Availability , Chromatography, Liquid , Contraceptive Agents, Male/administration & dosage , Dogs , Drug Carriers , Drug Stability , Gossypol/administration & dosage , Humans , Injections, Intravenous , Male , Mice , Microsomes, Liver/drug effects , Rats , Species Specificity , Tandem Mass Spectrometry , Tissue Distribution
11.
Food Chem Toxicol ; 46(1): 203-11, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17868966

ABSTRACT

1-methyl-d-tryptophan (D-1MT) reverses the immunosuppressive effect of indoleamine 2,3-dioxygenase (IDO), and it is currently being developed both as a vaccine adjuvant and as an immunotherapeutic agent for combination with chemotherapy. The present study examined the pharmacokinetics and toxicity of D-1MT in preparation for clinical trials. Incubation of D-1MT in rat plasma for 24h produced no significant degradation, with <15% of D-1MT being bound to plasma protein. Following oral administration, D-1MT exhibited a larger AUC and V(d), longer elimination t(1/2), and slower clearance in rats than in dogs. When oral doses of D-1MT exceeded levels of 600 mg/m(2)/day in rats, or 1200 mg/m(2)/day in dogs, the C(max) and AUC values decreased, resulting in a corresponding decrease in oral bioavailability. Thus, the doses were indicative of the lowest saturating doses in dogs and rats corresponding with an elimination t(1/2) of 6.0 h and 28.7 h, a T(max) of 1h and 8h, and a bioavailability of 47% and 92%, respectively. Tissue concentrations of D-1MT in mice were highest in the kidney, followed by the liver, muscle, heart, lung, and spleen, respectively; 48 h post dosing, D-1MT was excreted in the urine (35.1%) and feces (13.5%). Oral administration of D-1MT in rats from 150 to 3000 mg/m(2)/day (25-500 mg/kg/day) and in dogs from 600 to 1200 mg/m(2)/day (30 and 60 mg/kg/day) for 28 consecutive days did not lead to mortality, adverse events, histopathological lesions, or significant changes in hematology, clinical chemistry, and body weight. These results suggested that 3000 and 1200 mg/m(2)/day were the no-observed-adverse-effect levels in rats and dogs, respectively. Mean plasma concentrations of D-1MT (600 and 1200 mg/m(2)/day) in dogs 1h post dosing were 54.4 and 69.5 microg/ml on Day 1, respectively, and 53.1 and 66.6 microg/ml on Day 28, respectively; thus, indicating no increase in plasma D-1MT with a change in dose. In conclusion, D-1MT has little toxicity when administered orally to rats and dogs. Exceeding the saturating dose of D-1MT is unlikely to cause systemic toxicity, since any further increase in D-1MT plasma levels would be minimal.


Subject(s)
Tryptophan/analogs & derivatives , Administration, Oral , Animals , Area Under Curve , Blood Proteins/metabolism , Calibration , Dogs , Half-Life , Injections, Intravenous , Intestinal Absorption , Male , Mice , No-Observed-Adverse-Effect Level , Protein Binding , Rats , Rats, Inbred F344 , Rats, Inbred Lew , Tissue Distribution , Tryptophan/blood , Tryptophan/pharmacokinetics , Tryptophan/toxicity
12.
Am J Obstet Gynecol ; 196(4): 389.e1-9; discussion 389.e9-10, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17403430

ABSTRACT

OBJECTIVE: The purpose of this study was to evaluate the biodistribution and toxicity of the tropism-modified infectivity-enhanced conditionally replicative adenovirus, Ad5-delta24-arginine-glycine-aspartate (RGD). STUDY DESIGN: Cohorts of cotton rats were treated intravenously or intraperitoneally for 3 consecutive days with 5 x 10(8) to 5 x 10(11) particles/kg of Ad5-delta24-RGD or controls and killed on day 8, 17, or 56. For biodistribution studies, tissue samples from 14 organ sites and serum samples were evaluated for the presence of virus with the use of quantitative polymerase chain reaction analysis. For toxicity experiments, tissue samples from more than 30 organ sites and serum samples were obtained for the assessment of vector-related tissue or laboratory effects. RESULTS: Ad5-delta24-RGD was noted in tested samples at days 8 and 17 in animals that were treated intravenously and intraperitoneally with clearance by day 56. There were lower copies of vector noted in the blood and liver specimens of intraperitoneally treated animals. Mild peritonitis histopathologic findings were noted in rats that were treated intraperitoneally with Ad5-delta24-RGD; pathologic findings did not vary significantly with dose, over time, or in comparison to that noted in animals that were treated with Ad5-delta24. CONCLUSION: These studies provide critical insights regarding Ad5-delta24-RGD dosing and anticipated toxicity for a planned clinical trial for ovarian cancer.


Subject(s)
Adenoviridae , Neoplasm Recurrence, Local/therapy , Oligopeptides/pharmacology , Oncolytic Virotherapy/methods , Ovarian Neoplasms/therapy , Analysis of Variance , Animals , Disease Models, Animal , Female , Gene Transfer Techniques , Genetic Vectors/pharmacology , Injections, Intraperitoneal , Injections, Intravenous , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/mortality , Ovarian Neoplasms/mortality , Polymerase Chain Reaction , Probability , Random Allocation , Rats , Rats, Inbred Strains , Reference Values , Sensitivity and Specificity , Sigmodontinae , Survival Analysis
13.
Br J Pharmacol ; 147(5): 476-85, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16432511

ABSTRACT

This study aimed at characterizing the interspecies absorption, distribution, metabolism and elimination (ADME) profile of N-geranyl-N'-(2-adamantyl)ethane-1,2-diamine (SQ109), a new diamine-based antitubercular drug. Single doses of SQ109 were administered (intravenously (i.v.) and per os (p.o.)) to rodents and dogs and blood samples were analyzed by liquid chromatography tandem mass spectrometry (LC/MS/MS). Based on i.v. equivalent body surface area dose, the terminal half-life (t1/2) of SQ109 in dogs was longer than that in rodents, reflected by a larger volume of distribution (Vss) and a higher clearance rate of SQ109 in dogs, compared to that in rodents. The oral bioavailability of SQ109 in dogs, rats and mice were 2.4-5, 12 and 3.8%, respectively. After oral administration of [14C]SQ109 to rats, the highest level of radioactivity was in the liver, followed by the lung, spleen and kidney. Tissue-to-blood ratios of [14C]SQ109 were greater than 1. Fecal elimination of [14C]SQ109 accounted for 22.2% of the total dose of [14C]SQ109, while urinary excretion accounted for only 5.6%. The binding of [14C]SQ109 (0.1-2.5 microg ml-1) to plasma proteins varied from 6 to 23% depending on the species (human, mouse, rat and dog). SQ109 was metabolized by rat, mouse, dog and human liver microsomes, resulting in 22.8, 48.4, 50.8 or 58.3%, respectively, of SQ109 remaining after a 10-min incubation at 37 degrees C. The predominant metabolites in the human liver microsomes gave intense ion signals at 195, 347 and 363m/z, suggesting the oxidation, epoxidation and N-dealkylation of SQ109. P450 reaction phenotyping using recombinant cDNA-expressed human CYPs in conjunction with specific CYP inhibitors indicated that CYP2D6 and CYP2C19 were the predominant CYPs involved in SQ109 metabolism.


Subject(s)
Adamantane/analogs & derivatives , Antitubercular Agents/pharmacokinetics , Ethylenediamines/pharmacokinetics , Adamantane/pharmacokinetics , Animals , Aryl Hydrocarbon Hydroxylases/physiology , Blood Proteins/metabolism , Cytochrome P-450 CYP2C19 , Cytochrome P-450 CYP2D6/physiology , Dogs , Enzyme Inhibitors/pharmacology , Female , Humans , Male , Metabolic Clearance Rate , Mice , Mixed Function Oxygenases/physiology , Protein Binding , Rats , Rats, Inbred F344 , Species Specificity , Tissue Distribution
14.
J Pharmacol Exp Ther ; 315(2): 905-11, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16085758

ABSTRACT

The present study was aimed at fingerprinting pharmacoproteomic alterations of the Mycobacterium tuberculosis H37Rv strain induced by antitubercular drugs isoniazid (INH), ethambutol (EMB), and SQ109 [N-geranyl-N'-(2-adamantyl)ethane-1,2-diamine, a novel 1,2-diamine-based EMB analog], providing new understanding of pharmacoproteomic mechanisms of each and exploring new drug targets. The three drugs produced significant down-regulation of 13 proteins, including immunogenic ModD, Mpt64, with proteins from the Pro-Glu family being inhibited the most. Alternatively, the three drugs up-regulated 17 proteins, including secreted antigenic proteins ESAT-6 and CFP-10. Among these, ESAT-6 and AphC were most affected by INH, whereas EMB had the greatest effect on ESAT-6. All three drugs produced only moderate up-regulation of aerobic and iron metabolism proteins, i.e., electron transfer flavoprotein Fix A and Fix B, and ferritin-like protein BfrB, suggesting that the interruption of microbacterial energy metabolism is not a primary mechanism of action. INH suppressed ATP-dependent DNA/RNA helicase, but up-regulated beta-ketoacyl-acyl carrier protein synthase. These effects may contribute to its bactericidal effects. In contrast, EMB and SQ109 did just the opposite: these drugs up-regulated the helicase and down-regulated the synthase. For most of the H37Rv proteins, similar pharmacoproteomic patterns were found for both EMB and SQ109. None of the drugs significantly regulated expression of chaperonins GroES, GroEL2, and Dnak, suggesting that these drugs do not affect chaperone-mediated nascent polypeptide folding and sorting. The present study identified proteins directly modulated by the actions of INH, EMB, and SQ109 and distinguished INH activity from the diamine antitubercular compounds that inhibit M. tuberculosis H37Rv.


Subject(s)
Adamantane/analogs & derivatives , Antitubercular Agents/pharmacology , Ethambutol/pharmacology , Ethylenediamines/pharmacology , Isoniazid/pharmacology , Mycobacterium tuberculosis/drug effects , Mycobacterium tuberculosis/genetics , Adamantane/pharmacology , Amino Acid Sequence , Databases, Genetic , Electrophoresis, Polyacrylamide Gel , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Peptide Mapping , Protein Hydrolysates/chemistry , Protein Hydrolysates/metabolism , Proteome/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Trypsin/chemistry
15.
J Pharm Biomed Anal ; 37(4): 793-9, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15797803

ABSTRACT

Integrating combinatorial lead optimization of [1,2]-diamine core structure based on ethambutol with high-throughput screening has led us to focus on three promising analogs (SQ37, SQ59 and SQ109) as potential anti-tubercular drug candidates from thousands of synthesized diamine analogs for further characterization of their biopharmaceutical and pharmacokinetic properties by using liquid chromatography/tandem mass spectrometry (LC/MS/MS) and cassette dosing for pharmacokinetic screening. Simultaneous separation of the three analogs was achieved on reversed phase HPLC using a gradient mobile phase composed of MeOH/CH(3)COONH(4) (5mM)/trifluoroacetic acid: 80/20/0.1 (v/v/v). After extraction with acetonitrile from biomatrices, samples were analyzed on the LC/MS/MS system in the positive mode using an electrospray ion source. The retention time for the analogs ranged from 3.70 to 4.48 min. Incubation of SQ37 with plasma at 37 degrees C for 6h resulted in its degradation in human and rat plasma (20-35%), but no significant degradation was observed in mouse and dog plasma. SQ59 was relatively stable in the plasma of the four species. SQ109 was degraded in human and dog plasma (30-40%), but stable in mouse and rat plasma during the 6h incubation. A rapid multiple pharmacokinetic screening was taken by cassette dosing of the three analogs to mice and simultaneous analysis of their plasma concentrations. The analogs showed large Vd(ss) ranging from 11,300 (SQ37), 12,800 (SQ109) to 63,900 ml/kg (SQ59). The clearance ranged from 3240 (SQ109), 3530 (SQ37) and 8043 ml/kg/h (SQ59). The elimination t(1/2) ranged from 4.4 to 21.1h dependent on the routes. The oral bioavailability was 5.1 (SQ59), 20.1 (SQ37) and 7.8% (SQ109), respectively. Both SQ37 and SQ109 possess good pharmacokinetic properties.


Subject(s)
Antitubercular Agents/pharmacokinetics , Ethambutol/analogs & derivatives , Ethambutol/pharmacokinetics , Animals , Antitubercular Agents/blood , Biopharmaceutics , Chromatography, High Pressure Liquid , Combinatorial Chemistry Techniques , Dogs , Drug Design , Ethambutol/blood , Humans , Mass Spectrometry , Mice , Rats , Reference Standards , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL
...