Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 67
Filter
1.
Cancer Chemother Pharmacol ; 48(3): 202-8, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11592341

ABSTRACT

PURPOSE: Chloroethylnitrosoureas are among the most widely used chemotherapeutic agents for the treatment of brain tumors. SarCNU (1-(2-chloroethyl)3-sarcosinamide-1-nitrosourea) is an investigational nitrosourea analogue that has shown greater antitumor activity and a more favorable toxicity profile than 1,3-bis(2-chloroethyl)-1-nitrosourea in preclinical studies. The purpose of the present study was to characterize the plasma pharmacokinetics and oral bioavailability of SarCNU in mice and dogs treated by intravenous infusion and gastric intubation. METHODS: SarCNU was administered to mice by i.v. injection or orally at doses ranging from 10 to 100 mg/kg. Plasma samples were obtained from groups of five animals at each time-point at intervals ranging from 3 min to 2.5 h after dosing. A group of three male beagle dogs were treated with Sar CNU 10 mg/kg given both by i.v. infusion and orally in a crossover design. The concentration of SarCNU in plasma was measured by high-performance liquid chromatography. RESULTS: During the initial 90 min after i.v. injection to mice, SarCNU was eliminated from plasma in a monoexponential manner with a mean half-life of 9.8 +/- 0.8 min. The total plasma clearance was 47.3 +/- 8.7 ml/min per kg and the apparent volume of distribution was 0.7 +/- 0.1 l/kg. SarCNU exhibited linear pharmacokinetic behavior following both i.v. and oral administration of doses ranging from approximately 10 to 100 mg/kg. Peak plasma levels provided by a dose of 100 mg/kg given by the i.v. and oral routes were 142.4 microg/ml (0.5 min) and 27.8 microg/ml (9.8 min), respectively. The mean oral bioavailability of the drug was 57.3 +/- 12.6% in mice. In comparison, the disposition of SarCNU in dogs after rapid i.v. injection was biexponential, with half-lives of 5.4 +/- 8.4 min and 40.8 +/- 9.0 min for the initial and terminal disposition phases, respectively. Mean values of the total plasma clearance and apparent volume of distribution were 17.8 +/- 1.8 ml/min per kg and 1.1 +/- 0.3 l/kg, respectively. The Cmax was 18.5 +/- 6.5 microg/ml after i.v. injection and 8.5 0.4 microg/ml after oral administration of a 10 mg/kg dose. Oral bioavailability of the drug in dogs (71.7 +/- 21.2%) was greater than that observed in mice. CONCLUSIONS: SarCNU exhibited linear and consistent pharmacokinetics in mice and dogs with very good oral bioavailability in both species. These findings support the rationale for evaluating SarCNU given by the oral route of administration in phase I clinical trials.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Carmustine/pharmacokinetics , Administration, Oral , Animals , Antineoplastic Agents/administration & dosage , Area Under Curve , Biological Availability , Carmustine/administration & dosage , Carmustine/analogs & derivatives , Chromatography, High Pressure Liquid , Dogs , Drug Administration Schedule , Drug Evaluation, Preclinical , Infusions, Intravenous , Male , Maximum Tolerated Dose , Metabolic Clearance Rate , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Protein Binding , Proteins/metabolism
2.
J Pharm Biomed Anal ; 23(2-3): 395-402, 2000 Aug 15.
Article in English | MEDLINE | ID: mdl-10933532

ABSTRACT

Zinc finger motifs have been found to be important in a variety of protein structures including transcription factors and viral nucleocapsid proteins. Recently, it was demonstrated that various aromatic disulfides effectively remove the metal ion from the zinc finger, resulting in an alteration of tertiary structure in this region of the protein, thereby inhibiting transcription. Among these compounds, 2,2'-dithiobis[N-isoleucylbenzamide] exhibits activity against human immunodeficiency virus (HIV)-type 1 in vitro and has been selected for preclinical development as an anti-HIV agent. Analysis of this agent by reversed-phase high-performance liquid chromatography (HPLC) indicated a significant quantity of two additional compounds. Identifying the parent disulfide was accomplished by scanning eluting peaks with positive ion thermospray ionization (TSP) mass spectrometry (MS). Solution-induced disproportionation of the disulfide into its sulfhydryl monomer was demonstrated by treating the drug with dithiothreitol (DTT) prior to HPLC analysis. TSP-MS analysis of the remaining chromatographic peak suggested a molecular weight of 265, which, with 1H-nuclear magnetic resonance (NMR) data of the isolated material, allowed us to elucidate the chemical structure as N-isoleucyl-benzisothiazolone. Contact with stainless steel, such as that employed in an HPLC system, was found to accelerate degradation of the parent disulfide to the benzisothiazolone.


Subject(s)
Anti-HIV Agents/analysis , Benzamides/analysis , Isoleucine/analysis , Zinc Fingers , Anti-HIV Agents/pharmacology , Benzamides/pharmacology , Chromatography, High Pressure Liquid , Isoleucine/analogs & derivatives , Isoleucine/pharmacology , Magnetic Resonance Spectroscopy , Mass Spectrometry , Spectrophotometry, Ultraviolet
3.
J Pharm Biomed Anal ; 16(8): 1301-9, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9777604

ABSTRACT

Breflate is a water soluble prodrug developed to facilitate parenteral administration of the investigational antineoplastic agent brefeldin A (BFA). Previously, using analytical methods based upon reversed-phase high performance liquid chromatography (HPLC) with low wavelength UV detection or gas chromatography (GC) with electron capture detection following derivatization with heptafluorobutyrylimidazole, it was demonstrated that breflate undergoes rapid and efficient conversion to BFA following bolus i.v. injection in mice and dogs. However, plasma concentrations of the drug and prodrug achieved during the administration of nontoxic doses of breflate to beagle dogs as a 72 h continuous i.v. infusion were undetectable (< 0.1 microgram ml-1) by these methods. The sensitivity and specificity required for therapeutic drug level monitoring were achieved by GC with selected-ion mass spectrometry (MS) detection. Breflate, BFA and 1-eicosanol, the latter added to the sample as an internal standard (IS), were extracted from plasma into tert-butyl methyl ether (TBME) and esterified with trifluoroacetic anhydride. Methanol was added to the reaction mixture to effect the convenient removal of excess reagent as the volatile methyl ester during evaporation of the solvent. The residual material was analyzed directly upon reconstitution by capillary GC with automated splitless injection. Electron-ionization (70 eV) MS detection was performed by sequentially scanning ions at m/z 58, 202 and 325. The lowest concentration of either analyte quantified with acceptable reproducibility, as defined by an inter-day R.S.D. of about 20%, was near 10 ng ml-1 in plasma using a sample volume of 100 microliters. The assay has proven to be sufficiently sensitive, specific and reproducible for the routine analysis of pharmacokinetic specimens acquired during IND (investigational new drug)-directed toxicology studies in dogs.


Subject(s)
Anti-Bacterial Agents/blood , Brefeldin A/analogs & derivatives , Brefeldin A/blood , Chromatography, High Pressure Liquid/methods , Gas Chromatography-Mass Spectrometry/methods , Prodrugs/metabolism , Animals , Anti-Bacterial Agents/pharmacokinetics , Brefeldin A/pharmacokinetics , Dogs , Macrolides , Male , Mice , Sensitivity and Specificity
4.
Cancer Res ; 57(18): 3895-8, 1997 Sep 15.
Article in English | MEDLINE | ID: mdl-9307267

ABSTRACT

Nitrosoureas are among the most widely used agents used in the treatment of malignant gliomas. Here, the activity of 2-chloroethyl-3-sarcosinamide-1-nitrosourea (SarCNU) was compared with that of 1,3-bis-(2-chloroethyl)-1-nitrosourea (BCNU), in vivo against s.c. implanted SF-295 and U-251 central nervous system (CNS) tumor xenografts. When given i.v., q4d for 3 doses, to athymic mice bearing s.c. SF-295 tumors, SarCNU, at an optimum of 167 mg/kg/dose, produced 9 tumor-free animals of 10 total animals, 1 regression, and no evidence of overt toxicity (> or =20% body weight loss). With a similar dosing schedule, BCNU produced no tumor-free animals, six regressions, and one drug-related death at its optimum of 30 mg/kg/dose. Furthermore, SarCNU retained high antitumor activity at two lower dose levels, 66 and 45% of the optimal dose, whereas BCNU demonstrated a progressive loss of antitumor activity at lower doses. Following p.o. administration, SarCNU similarly demonstrated antitumor activity that was superior to that of BCNU. In the U-251 CNS tumor model, SarCNU yielded six of six tumor-free animals at 80 mg/kg/dose with i.p. administration q.d. for 5 days, starting on day 14, whereas BCNU, at 9 mg/kg/dose, yielded three of six tumor-free mice and one drug-related death. Again, SarCNU resulted in tumor-free animals at 66 and 45% of its optimal dose and was relatively nontoxic, in contrast to BCNU. Results of testing to date indicate that SarCNU is clearly more effective than BCNU against the human CNS tumors SF-295 and U-251 in vivo. These results encourage the initiation of clinical trials for SarCNU, in an effort to improve therapeutic approaches to glioma, but clinical trials must determine whether superiority of SarCNU in preclinical models can be extrapolated to patients.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Carmustine/analogs & derivatives , Carmustine/administration & dosage , Glioma/drug therapy , Administration, Oral , Animals , Drug Administration Schedule , Female , Humans , Injections, Intraperitoneal , Male , Mice , Mice, Nude , Neoplasm Transplantation , Transplantation, Heterologous
5.
AIDS Res Hum Retroviruses ; 13(9): 789-96, 1997 Jun 10.
Article in English | MEDLINE | ID: mdl-9171223

ABSTRACT

A series of compounds related to oxathiin carboxanilide has been identified as nonnucleoside reverse transcriptase inhibitors (NNRTIs) of HIV-1, and structure-activity relationships have been described (Buckheit RW, et al.: Antimicrob Agents Chemother 1995;39:2718-2727). Three new analogs (UC040, UC82, and UC781) inhibited laboratory and clinical isolates of HIV-1, including isolates representative of the various clades of HIV-1 found worldwide, in both established and fresh human cells. Virus isolates with the amino acid changes L100I, K103N, V106I, and Y181C in the reverse transcriptase were partially resistant to these compounds. However, UC781 inhibited these virus isolates at low nontoxic concentrations, presenting a broad in vitro therapeutic index. As with other NNRTIs, each of the compounds synergistically interacted with AZT to inhibit HIV-1 replication. UC781 possesses a favorable pharmacokinetic profile in mice with a high level of oral bioavailability. Plasma concentrations reached maximum levels within 2 to 4 hr of oral administration and remained in excess of those required for in vitro anti-HIV activity for at least 24 hr after a single oral dose. When evaluated in a murine hollow fiber implant model of HIV infection, UC781 dosed orally or parenterally was able to suppress HIV replication completely in this model system, providing evidence of the in vivo efficacy of the compound.


Subject(s)
Anilides/pharmacology , Anilides/pharmacokinetics , Anti-HIV Agents/pharmacology , Anti-HIV Agents/pharmacokinetics , Furans/pharmacology , Furans/pharmacokinetics , HIV-1/drug effects , HIV-2/drug effects , Reverse Transcriptase Inhibitors/pharmacokinetics , Administration, Oral , Animals , Biological Availability , HIV Core Protein p24/analysis , HIV Reverse Transcriptase/antagonists & inhibitors , HIV Reverse Transcriptase/genetics , HIV-1/enzymology , HIV-1/genetics , Humans , Male , Metabolic Clearance Rate , Mice , Mice, Inbred Strains , Microbial Sensitivity Tests , Point Mutation , Reverse Transcriptase Inhibitors/pharmacology , Thioamides
6.
Clin Cancer Res ; 3(3): 455-63, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9815705

ABSTRACT

The spicamycin analogue KRN5500 (NSC 650426; SPA) is derived from Streptomyces alanosinicus. The unique structure contains a purine, an aminoheptose sugar, glycine, and a tetradecadiene fatty acid. SPA potently inhibits the growth of certain human tumor cell lines in vitro (IC50 for growth <100 nM) and displays marked activity in vivo in Colo 205 colon carcinoma xenografts. Selective inhibition of labeled precursor incorporation was not evident at 1 or 4 h of exposure to the drug, but at 8 h, [3H] leucine incorporation was inhibited by approximately 40% at or below the IC50 for cell growth. Because of the structural similarity of SPA to inhibitors of glycoprotein processing, we examined the effect of SPA on indicators of glycoprotein synthesis and processing in HL60TB promyelocytic leukemia and Colo 205 colon carcinoma cells. Brief periods of exposure ( approximately 30 min) to SPA at the IC50 for growth increased incorporation of [3H]mannose. When examined by Western blotting after prolonged (40-48 h) incubation with lectins that target mannose-containing carbohydrates, Galanthus nivalis agglutinin and concanavalin A, a qualitative change in the pattern of mannose-containing glycoproteins was observed in HL60TB cells. Significant changes in the pattern of surface glycoprotein expression in intact cells were demonstrated by flow cytometry using fluorescence-labeled lectins. An increase in the number of cells binding G. nivalis agglutinin (indicating terminal mannose) was noted, but a decrease in the amount of lectin bound per cell was noted for wheat germ agglutinin (detecting sialic acid and terminal beta-N-acetyl glucosamine residues). Electron microscopy revealed loss of microvilli, and the Golgi apparatus appeared inflated. Our findings, therefore, raise the possibility that cells exposed to SPA have altered glycoprotein processing after exposure to low concentrations of drug, prior to the occurrence of overt cytotoxicity. These effects are consistent with a prominent early effect of SPA on the enzymatic machinery or organelles important for proper glycoprotein processing and emphasize the novelty of this agent's likely mechanism of action.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Cell Division/drug effects , Colonic Neoplasms/drug therapy , Glycoproteins/biosynthesis , Membrane Glycoproteins/biosynthesis , Animals , Antibiotics, Antineoplastic/therapeutic use , Colonic Neoplasms/pathology , Colonic Neoplasms/ultrastructure , Drug Screening Assays, Antitumor , Flow Cytometry , Galanthus , HL-60 Cells , Humans , Lectins , Mannose/metabolism , Mice , Mice, Nude , Plant Lectins , Purine Nucleosides/therapeutic use , Purine Nucleosides/toxicity , Transplantation, Heterologous , Tumor Cells, Cultured
7.
Clin Cancer Res ; 3(2): 273-9, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9815683

ABSTRACT

We have selected a panel of human tumor xenografts for in vitro and in vivo studies that allows an indication of selectivity of action of novel chemotherapeutic agents. We report here the antitumor activity of the flavone flavopiridol (previously designated L86-8275), which has been selected for further studies based in part on its behavior in the anticancer drug screening system of the United States National Cancer Institute. Eighteen human tumor and five cell line-derived xenografts established by serial passage in nude mice in our laboratory were used as tumor models for in vitro investigations using a modified double-layer soft agar assay. In vivo investigations were completed in nude mice bearing advanced-stage s.c. growing prostate cancer xenografts. Antitumor activity in vitro (test/control

Subject(s)
Antineoplastic Agents/pharmacology , Flavonoids/pharmacology , Piperidines/pharmacology , Prostatic Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Colony-Forming Units Assay , Drug Screening Assays, Antitumor , Flavonoids/therapeutic use , Humans , Male , Mice , Mice, Nude , Neoplasm Transplantation , Piperidines/therapeutic use , Tumor Cells, Cultured
8.
J Chromatogr B Biomed Appl ; 677(2): 351-62, 1996 Mar 03.
Article in English | MEDLINE | ID: mdl-8704941

ABSTRACT

A facile, sensitive and highly specific HPLC method for assaying 1-(2-chloroethyl)-3-sarcosinamide-1-nitrosourea (SarCNU) in plasma has been developed. The drug was efficiently isolated from plasma by extraction with tert.-butyl methyl ether. A structurally related compound with similar physicochemical properties served as the internal standard (I.S.). Following evaporation of the organic solvent, the extract was reconstituted with 0.05 M ammonium acetate buffer, pH 5.0, and loaded onto a 4 micron Nova-Pak C18 column (15 cm x 3.9 mm), which was preceded by a 7 micron Brownlee RP-18 precolumn (1.5 cm x 3.2 mm). Chromatography was performed at ambient temperature using a mobile phase of methanol-0.1 M ammonium formate buffer, pH 3.7 (25:75, v/v). UV absorbance of the effluent was monitored at 240 nm. A flow-rate of 1.0 ml/ min was used for analyzing mouse and dog plasma extracts. Under these conditions, the drug eluted at 4.0 min and was followed by the I.S. at 6.1 min. An automatic switching valve was employed to allow the precolumn to be flushed 1.5 min into the run, without interrupting the flow of the mobile phase to the analytical column, thereby preventing the apparent build-up of extractable, strongly retained, UV-absorbing components present in mouse and dog plasma. Operating in this manner, more than 100 samples could be analyzed during a day using a refrigerated autosampler for overnight injection. The method was readily adapted to the determination of SarCNU in human plasma by simply decreasing the eluent flow-rate to 0.6 ml/min, whereby SarCNU and the I.S. eluted at approximately 5.8 and 9.1 min, respectively. Furthermore, the switching valve was not necessary for the analysis of human plasma samples. With a 50-microliter sample volume, the lowest concentration of SarCNU included in the plasma standard curves, 0.10 micrograms/ml, was quantified with a 7.8% R.S.D. (n = 27) over a 2 month period. Plasma standards, with concentrations of 0.26 to 5.1 micrograms/ml, exhibited R.S.D. values ranging from 1.3 to 4.7%. Thermospray-ionization MS detection was used to definitively establish the specificity of the method. The sensitivity of the assay was shown by application to be more than adequate for characterizing the plasma pharmacokinetics of SarCNU in mice.


Subject(s)
Antineoplastic Agents/blood , Carmustine/analogs & derivatives , Chromatography, High Pressure Liquid/methods , Animals , Antineoplastic Agents/pharmacokinetics , Carmustine/blood , Carmustine/pharmacokinetics , Dogs , Humans , Male , Mice , Reference Standards , Reproducibility of Results , Sensitivity and Specificity , Spectrophotometry, Ultraviolet
9.
Cancer J Sci Am ; 2(1): 52-8, 1996.
Article in English | MEDLINE | ID: mdl-9166499

ABSTRACT

PURPOSE: An empiric in vitro screen of human tumor cell lines found brefeldin A inhibited the growth of immortalized human cell lines, with particular sensitivity to brefeldin in a series of immortalized melanoma cell lines and nonimmortalized prostate carcinoma explants. Brefeldin A alters the morphology and function of the Golgi apparatus, endosomal, and trans-Golgi compartments in different cell types. The studies presented here sought to obtain evidence of in vivo antitumor activity by brefeldin A. METHODS: Antiproliferative activity was studied in prostate carcinoma cells in vitro using cell counts, protein, and viable stains. Activity was also studied in vivo against subcutaneous and subrenal capsule melanoma models. RESULTS: Protracted exposures in vitro (between 24 and 72 hours) are necessary to cause persistent growth inhibition of immortalized PC3 prostate carcinoma cells. In human melanoma athymic mouse xenografts, brefeldin A showed antitumor activity in vivo when given 16 to 64 mg/kg/injection intraperitoneally q 7 h x 2, daily for 5 days. Activity was also observed in the intraperitoneal LOX IMVI (65%-100% increase in life span, with 17%-50% day 60 survivors); early-stage subcutaneous LOX IMVI and SK-MEL-5 (86%-100% growth inhibition), and subrenal capsule SK-MEL-5 and M19-MEL models. CONCLUSIONS: Brefeldin A possesses noteworthy antitumor activity in vivo and antiproliferative effects in vitro in certain cell types. Strategies to allow protracted exposure of tumor cells to brefeldin A while preserving a therapeutic index are needed to assess the clinical potential of brefeldin A.


Subject(s)
Antineoplastic Agents/therapeutic use , Brefeldin A/therapeutic use , Cell Proliferation/drug effects , Neoplasms, Experimental/drug therapy , Protein Synthesis Inhibitors/therapeutic use , Animals , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , In Vitro Techniques , Male , Melanoma/drug therapy , Mice , Mice, Nude , Prostatic Neoplasms/drug therapy , Time Factors
10.
Science ; 270(5239): 1194-7, 1995 Nov 17.
Article in English | MEDLINE | ID: mdl-7502043

ABSTRACT

Strategies for the treatment of human immunodeficiency virus-type 1 (HIV-1) infection must contend with the obstacle of drug resistance. HIV-1 nucleocapsid protein zinc fingers are prime antiviral targets because they are mutationally intolerant and are required both for acute infection and virion assembly. Nontoxic disulfide-substituted benzamides were identified that attack the zinc fingers, inactivate cell-free virions, inhibit acute and chronic infections, and exhibit broad antiretroviral activity. The compounds were highly synergistic with other antiviral agents, and resistant mutants have not been detected. Zinc finger-reactive compounds may offer an anti-HIV strategy that restricts drug-resistance development.


Subject(s)
Antiviral Agents/pharmacology , Benzamides/pharmacology , Capsid Proteins , Capsid/metabolism , Disulfides/pharmacology , Gene Products, gag/antagonists & inhibitors , HIV-1/drug effects , Viral Proteins , Zinc Fingers/drug effects , Amino Acid Sequence , Animals , Antiviral Agents/chemistry , Antiviral Agents/pharmacokinetics , Benzamides/chemistry , Benzamides/pharmacokinetics , Biological Availability , Capsid/chemistry , Cell Line , Disulfides/chemistry , Disulfides/pharmacokinetics , Drug Resistance, Microbial , Drug Synergism , Gene Products, gag/chemistry , HIV-1/physiology , Humans , Male , Mice , Molecular Sequence Data , gag Gene Products, Human Immunodeficiency Virus
11.
Antiviral Res ; 28(3): 265-79, 1995 Nov.
Article in English | MEDLINE | ID: mdl-8629818

ABSTRACT

Previous studies demonstrated that human cell lines can be cultivated in hollow fibers in the subcutaneous and intraperitoneal compartments of mice. We have extended the range of cell lines to include cells infected with the human immunodeficiency virus (HIV). Furthermore, these HIV-infected cells have been shown to replicate in the hollow fibers located in both physiologic compartments (intraperitoneal and subcutaneous) of SCID mice. Treatment of the host mice with antiviral agents can suppress virus replication in these hollow fiber cultures. The potential use of this system for early in vivo screening of anti-HIV compounds is discussed.


Subject(s)
Antiviral Agents/pharmacology , Drug Evaluation, Preclinical/methods , HIV/drug effects , Animals , Cells, Cultured , HIV Core Protein p24/analysis , Humans , Mice , Mice, SCID , RNA-Directed DNA Polymerase/metabolism , Virus Cultivation , Virus Replication/drug effects , Zalcitabine/pharmacology , Zidovudine/pharmacology
12.
Cancer Res ; 55(13): 2794-9, 1995 Jul 01.
Article in English | MEDLINE | ID: mdl-7796405

ABSTRACT

[(3,5-Di-tert-butyl-4-hydroxybenzylidene)-malononitrile] (AG17), a "tyrphostin" tyrosine kinase antagonist, was found to inhibit tumor cell growth with 50% growth inhibition ranging from 0.7 to 4.0 microM in a panel of 13 human tumor cell lines, as evaluated by tetrazolium dye reduction and inhibition of precursor incorporation into macromolecules. The promyelocytic leukemia cell line HL-60(TB), was the most sensitive with irreversible total growth inhibition after 12 h of exposure to 1.5 microM drug. Antiproliferative effects of AG17 in HL-60(TB) cells were temporally related to disruption of mitochondrial function, which occurred within 1 h after drug exposure as demonstrated by a significantly decreased mass of ATP in drug-treated cells, loss of the fluorescent mitochondrial membrane potential probe rhodamine 123, and ultrastructural examination of mitochondria using fluorescence and electron microscopy. Specific decreases of total or tyrosine-phosphorylated substrate at concentrations of the drug not affecting ATP levels were not detected. These data raise the possibility that AG17 may act in part by altering mitochondrial function and/or structure, and that impairment of mitochondrial function may be exploitable as a potentially useful mechanism to modulate tumor cell proliferation. This study also emphasizes the importance of evaluating carefully the effects of potential protein kinase antagonists, since these structures have effects in intact cells in addition to what might be expected from in vitro enzyme assays.


Subject(s)
Growth Inhibitors , Mitochondria/drug effects , Nitriles/pharmacology , Phenols/pharmacology , Tyrphostins , Adenosine Triphosphate/metabolism , Cell Division/drug effects , Humans , In Vitro Techniques , Intracellular Membranes/drug effects , Permeability , Protein-Tyrosine Kinases/antagonists & inhibitors , Rhodamine 123 , Rhodamines , Time Factors , Tumor Cells, Cultured
13.
Drug Metab Dispos ; 23(7): 676-80, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7587953

ABSTRACT

The monocyclic terpene d-limonene, a major component in many citrus essential oils (1-3), has been used for many years as a flavoring agent, food additive, and fragrance (1, 2). It was recently demonstrated that limonene exhibits both chemopreventive and therapeutic effects against chemically induced mammary tumors in rats (4-10). Mechanistic studies revealed that limonene inhibits the posttranslational isoprenylation of 21-26 kDa cellular proteins implicated in cell growth and proliferation (11-13). Limonene is extensively metabolized by a variety of mammalian species (14-17). Its principal circulating metabolites identified in the rat, perillic acid and dihydroperillic acid, are also effective inhibitors of isoprenylation and cellular proliferation in vitro (17, 18). Furthermore, one of the metabolic precursors of these compounds, perillyl alcohol (16), is considerably more potent than limonene against the in vivo rat mammary tumor models (19). A preliminary report of an ongoing phase I clinical trial with limonene indicated that a single oral dose of 100 mg/kg is well tolerated (20). However, an extrapolation based upon the rat mammary tumor regression studies suggests that the minimum human dose requirement would be 1000 mg/kg/ day (6). The administration of such a large dose, which amounts to more than 80 ml of an oily volatile liquid, on a continuing basis may cause problems. Thus, perillyl alcohol is currently being developed as a clinical candidate at the National Cancer Institute because of its greater potency than limonene, which may enable potentially effective systemic concentrations of the active principals to be achieved at considerably lower doses.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Antineoplastic Agents/pharmacokinetics , Monoterpenes , Terpenes/pharmacokinetics , Administration, Oral , Animals , Antineoplastic Agents/blood , Dogs , Female , Gas Chromatography-Mass Spectrometry , Male , Terpenes/blood
14.
Antimicrob Agents Chemother ; 39(1): 9-14, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7695336

ABSTRACT

Michellamine B (MB) is a naturally occurring naphthylisoquinoline alkaloid of novel chemical structure with activity against human immunodeficiency virus (HIV) types 1 and 2 in vitro. In conjunction with its preclinical evaluation, the plasma pharmacokinetics of MB was characterized in mice and dogs treated by intravenous infusions of 1- and 15-min durations, respectively. At doses ranging from 1 to 9 mg/kg of body weight, the drug exhibited apparent first-order kinetics in both species, affording triexponential plasma concentration-time profiles. Treatment with doses of 5 to 9 mg/kg provided peak plasma levels within the range that completely inhibits the cytopathic effects of HIV upon cultured human lymphoblastoid cells (50 to 100 micrograms/ml) without evidence of toxicity. MB had a biological half-life of 2.8 +/- 0.8 h in mice, with a mean residence time of 2.1 +/- 0.3 h, and a total plasma clearance of 2.4 +/- 0.5 ml/min/kg (mean +/- standard deviation; n = 3); however, the terminal-phase contribution to the area under the plasma profile from time zero to infinity was 44.6% +/- 12.9%. In contrast, the terminal phase was the primary determinant of drug disposition in dogs, accounting for 74.1% +/- 2.8% (n = 3) of the area under the curve. Furthermore, the systemic duration of MB was significantly longer in the dogs than in mice, as indicated by mean values of the apparent half-life (11.6 +/- 1.2 h), mean residence time (12.3 +/- 1.8 h), and clearance (0.50 +/- 0.08 ml/min/kg). However, there were no statistical difference between its apparent volume of distribution in the mice (0.60 +/- 0.08 liters/kg) and dogs (0.50 +/- 0.07 liters/kg). A single dog was also treated with a total dose of 97 mg/kg given as a 72-h constant-rate intravenous infusion, since prolonged systemic exposure to potentially therapeutic drug concentrations will very likely be required for clinical anti-HIV effects. Within 4 h after starting the infusion, the plasma MB concentration exceeded 18 micrograms/ml, it reported 50% effective concentration against HIV in vitro, and subsequently increased to 41 micrograms/ml at the end of the infusion. There were no clinical or pathological indications of toxicity. Whereas the total plasma clearance (0.48 ml/min/kg) was within the range observed for dogs treated by 15-min infusion, extension of the postinfusion sampling period from 24 h to 4 days facilitated better definition of the terminal exponential phase, yielding a value of 25.6 h for the biological half-life of MB. The amount of drug excreted by dogs unchanged in the urine ranged from 3.7 to 11.1% of the administered dose. Thus, the major pathways by which the drug is eliminated from the body remain to be identified. On the basis of these findings, continued development of MB as a novel lead compound for the treatment of HIV infection is warranted.


Subject(s)
Antiviral Agents/pharmacokinetics , HIV-1/drug effects , HIV-2/drug effects , Isoquinolines/pharmacokinetics , Naphthalenes/pharmacokinetics , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/toxicity , Dogs , Dose-Response Relationship, Drug , Female , Infusions, Intravenous , Isoquinolines/administration & dosage , Isoquinolines/toxicity , Male , Metabolic Clearance Rate , Mice , Naphthalenes/administration & dosage , Naphthalenes/toxicity
15.
Life Sci ; 57(2): 131-41, 1995.
Article in English | MEDLINE | ID: mdl-7603295

ABSTRACT

Advancement of potential anti-cancer agents from "discovery" in an in vitro screen to pre-clinical development requires a demonstration of in vivo efficacy in one or more animal models of neoplastic disease. Most such models require considerable materials in terms of laboratory animals and test compound as well as substantial amounts of time (and cost) to determine whether a given experimental agent or series of agents have even minimal anti-tumor activity. The present study was initiated to assess the feasibility of employing an alternate methodology for preliminary in vivo evaluations of therapeutic efficacy. Results of experimentation to date demonstrate that a hollow fiber encapsulation/implantation methodology provides quantitative indices of drug efficacy with minimum expenditures of time and materials. Following further pharmacologic calibrations, the hollow fiber technique is anticipated (a) to identify compounds having moderate to prominent anti-cancer activity and (b) to facilitate the identification of sensitive tumor cell line "targets" and optimal or near-optimal treatment regimens for subsequent testing using standard in vivo solid tumor models. The potential suitability of this methodology is demonstrated with several standard anti-neoplastic agents.


Subject(s)
Neoplasms/pathology , Polymers , Polyvinyls , Sulfones , Tumor Cells, Cultured , Animals , Antineoplastic Agents/pharmacology , Cell Communication/physiology , Cell Division/physiology , Cytological Techniques , Drug Screening Assays, Antitumor , Female , Humans , Male , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms/drug therapy , Transplantation, Heterologous , Tumor Cells, Cultured/drug effects
16.
Cancer Chemother Pharmacol ; 36(4): 305-15, 1995.
Article in English | MEDLINE | ID: mdl-7628050

ABSTRACT

The plasma pharmacokinetics of the anti-tumor antibiotic geldanamycin (GM: NSC 122750), a naturally occurring benzoquinoid ansamycin, was characterized in mice and a beagle dog. Concentrations of GM well above 0.1 microgram/ml, which was typically effective against neoplastic cell lines responsive to the drug in vitro, were achieved in the plasma of the mice and the dog treated by i.v. injection. However, the systemic duration of the drug was relatively short. Plasma levels decayed below 0.1 microgram/ml within 3-4 h after administration of the apparent maximum tolerated doses, which were approximately 20 mg/kg for the mice and 4 mg/kg for the dog. The drug exhibited linear pharmacokinetic behavior within the dose ranges studied. However, there were significant interspecies differences in its disposition. Whereas the mean biological half-life of GM was slightly longer in the mice (77.7 min) than in the dog (57.9 min), its mean residence time in the dog (46.6 min) was more than twofold greater than that observed in the mice (20.7 min). Nevertheless, the drug was cleared from plasma much faster by the dog (49.4 ml/min per kg) than by the mice (30.5 ml/min per kg). These apparent anomalies were principally associated with differences in the relative significance of the terminal phase upon overall drug disposition. The liver appeared to be the principal target organ of acute drug toxicity in the dog. Doses of 2.0 and 4.2 mg/kg both produced elevations in serum levels of the transaminases and other indicators of liver function characteristic of acute hepatic necrosis. Additional effects included symptoms of minor gastrointestinal toxicity and alterations in serum chemistry parameters consistent with less severe nephrotoxicity. Drug-related toxicity appeared to be reversible. In consideration of the potential for acute hepatotoxic reactions to GM, as well as to the other benzoquinoid ansamycins based upon structural analogy, additional pharmacological and therapeutic information is required to ascertain whether these compounds are viable candidates for clinical development.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , Quinones/pharmacology , Animals , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/pharmacokinetics , Benzoquinones , Chromatography, High Pressure Liquid , Dogs , Drug Screening Assays, Antitumor , Female , Half-Life , Humans , Lactams, Macrocyclic , Liver/drug effects , Liver/pathology , Male , Metabolic Clearance Rate , Mice , Mice, Inbred Strains , Necrosis , Quinones/adverse effects , Quinones/pharmacokinetics , Tumor Cells, Cultured/drug effects
17.
J Natl Cancer Inst ; 87(1): 46-51, 1995 Jan 04.
Article in English | MEDLINE | ID: mdl-7666463

ABSTRACT

BACKGROUND: Jasplakinolide, a cyclodepsipeptide produced by an Indo-Pacific sponge, Jaspis johnstoni, has been reported to inhibit the growth of breast cancer cells. PURPOSE: The effects of jasplakinolide on the proliferation of three human immortalized prostate carcinoma cell lines (PC-3, LNCaP, and TSU-Pr1) were studied. The growth-inhibitory effect of jasplakinolide on the PC-3 cell line was studied in detail to elucidate its mechanism of action. METHODS: Cell counts were used to study growth inhibition. A protein-based microplate assay was used to assess the time of exposure needed to cause persistent growth inhibition and to study the effects of jasplakinolide analogues. Metabolic changes were assessed by following the incorporation of radiolabeled precursors. The effects of jasplakinolide on the cytoskeleton were studied by fluorescent microscopy, using rhodamine phalloidin (RP) and antibodies to cytoskeletal components. Changes in RP binding were quantified by extracting bound fluorescent material from fixed cells and measuring the amount of fluorescence in a spectrofluorometer. RESULTS: The growth of PC-3, LNCaP, and TSU-Pr1 cells was potently inhibited by exposure to jasplakinolide for 48 hours; doses of jasplakinolide that led to 50% growth inhibition were 65 nM for PC-3 cells, 41 nM for LNCaP cells, and 170 nM for TSU-Pr1 cells. In PC-3 cells, exposure to 160 nM for 48 hours led to total growth inhibition, which persisted for several days even after drug removal. Several jasplakinolide analogues also inhibited the growth of PC-3 cells, although analogues in which the rigidity of the macrolide ring was altered were ineffective. No early changes in the synthesis of DNA, RNA, or protein or in intracellular adenosine triphosphate levels were seen in the PC-3 cells after exposure to jasplakinolide. Growth inhibition by jasplakinolide was accompanied by striking morphologic changes. Exposure for several doublings led to multinucleated cells. Further investigation of these changes in the PC-3 cells revealed a dramatic and early disruption of the actin cytoskeleton and a statistically significant decrease in RP binding. The doses of jasplakinolide, the time of exposure, and the pattern of growth inhibition by structural analogues corresponded with the changes seen in actin distribution. CONCLUSIONS: Jasplakinolide represents a novel marine natural product with potent in vitro antiproliferative activity against human prostate carcinoma cell lines, and it appears to target the actin cytoskeleton. IMPLICATIONS: Jasplakinolide is a potential candidate for further preclinical development and a lead structure for a novel class of therapeutic agents that can disrupt the actin cytoskeleton in mammalian cells.


Subject(s)
Actins/drug effects , Antineoplastic Agents/pharmacology , Depsipeptides , Prostatic Neoplasms/drug therapy , Humans , Male , Peptides, Cyclic/pharmacology , Tumor Cells, Cultured
18.
Int J Oncol ; 7(4): 847-54, 1995 Oct.
Article in English | MEDLINE | ID: mdl-21552913

ABSTRACT

Genistein (GEN) has recently generated considerable interest as a potential agent for the prevention and treatment of cancer. The present investigation was undertaken to determine if the concentrations of drug shown to inhibit the growth of human tumor cell lines by 50% in vitro (IC50=2-27 mu g/ml) can be achieved and sustained systemically in mice. We found that GEN plasma levels decreased biexponetially from 64 mu g/ml to 0.55 mu g/ml during the initial 40 min after i.v. injection of a 52 mg/kg dose. Mean half-lives of the two initial disposition phases were 2.5+/-0.4 min and 7.1+/-1.1 min in mice treated with doses of 9-52 mg/kg. Plasma profiles of i.v. GEN exhibited a prominent secondary peak near 78 min followed by a terminal decay phase with a 39.5+/-16.8 min half-life. Although these features are suggestive of enterohepatic cycling, the mean apparent total plasma clearance of GEN (66.5+/-7.3 ml/min/kg) was nevertheless similar to hepatic blood flow. The systemic availability of GEN from a 180 mg/kg p.o. dose, which afforded 1.1 mu g/ml peak plasma concentration, was only 12%. Thus, bolus i.v. and p.o. administration of GEN failed to either achieve or adequately sustain plasma levels of the drug within the target range established by in vitro antitumor studies. Plasma levels resulting from i.p. injection of a 185 mg/kg dose were 5-times greater on average than achieved by the p.o. route. While the plasma concentration exceeded the IC50 values for the majority of human cancer cell lines responsive to GEN for only a short period of time, drug levels remained above 2 mu g/ml, the IC50 of the most sensitive cell lines, for 4 h. Extrapolation from the single dose study suggests that repetitive i.p. injection of at least 200 mg/kg GEN every 8 h will afford continuous systemic exposure to potentially cytostatic concentrations of the drug against these cell lines. This information should facilitate efforts to assess the effectiveness of GEN in appropriate in vivo tumor models.

19.
J Natl Cancer Inst ; 86(24): 1846-52, 1994 Dec 21.
Article in English | MEDLINE | ID: mdl-7990159

ABSTRACT

BACKGROUND: Cancer treatments that appear promising in tissue culture are often less effective in solid tumors, in part because of the proliferative and microenvironmental heterogeneity that develops in these tumors as they grow. Heterogeneous tumor models are thus needed for drug screening. PURPOSE: Our goal was to develop and test for drug evaluation a solid tumor model based on cell growth inside biocompatible hollow fibers. METHODS: Building on the experience of Hollingshead and co-workers with a sparse-cell, hollow-fiber tumor model, we tested six human tumor cell lines for in vitro growth inside 450-microns internal-diameter polyvinylidine fluoride fibers and examined them histologically. Human SW620 colon carcinoma cells grown in hollow fibers were also examined using electron microscopy, and their doxorubicin sensitivity was assessed. A colorimetric assay based on sulforhodamine B was adopted to replace the more cumbersome clonogenic cell survival assay. RESULTS: Five of the human tumor cell lines tested grew to confluence, forming heterogeneous in vitro tumors with subpopulations of viable and necrotic cells. For SW620 hollow-fiber tumors, maximum viable cell populations in excess of 10(8) cells/mL were obtained after 8 days of growth. This viable cell density remained roughly constant for 3-4 days, permitting dose-response experiments over this time interval. Tumor cells in hollow fibers were much more resistant to a 4-hour doxorubicin exposure than were tumor cells in monolayers: LC50 values (i.e., the drug concentrations at which the plating efficiency equals one-half the plating efficiency of untreated cells) of 3.5 microM and 0.16 microM were obtained for hollow-fiber tumors and monolayers, respectively. LC50 values decreased when drug exposure time was increased. Results from the colorimetric assay were in agreement with those from the clonogenic assay. CONCLUSION: The successful growth of tumor cells to confluence in hollow fibers and the feasibility of performing in vitro drug dose-response experiments with a relatively easy colorimetric assay demonstrate the potential of the hollow-fiber solid tumor model as a tool for experimental therapeutic research. IMPLICATION: Hollow-fiber solid tumors may prove useful for experimental drug evaluation.


Subject(s)
Tumor Cells, Cultured/cytology , Tumor Stem Cell Assay/methods , Antineoplastic Agents/pharmacology , Biocompatible Materials , Cell Division/drug effects , Colorimetry , Dose-Response Relationship, Drug , Doxorubicin/pharmacology , Humans , Tumor Cells, Cultured/drug effects
20.
Cancer Chemother Pharmacol ; 33(4): 325-30, 1994.
Article in English | MEDLINE | ID: mdl-8281627

ABSTRACT

Rapamycin-28-N,N-dimethylglycinate methanesulfonate salt (RG), synthesized as a potential water-soluble prodrug to facilitate parenteral administration of the antineoplastic macrolide rapamycin (RA), is active against intracranially implanted human glioma in mice. Preclinical pharmacokinetic studies to evaluate the prodrug were conducted in male CD2F1 mice treated with 10, 25, 50 and 100 mg/kg doses of RG by rapid i.v. injection. The plasma concentration of RG decayed in a distinctly triphasic manner following treatment with the 100 mg/kg dose; however, prodrug disposition was apparent biexponential at each of the lower doses. RG exhibited dose-dependent pharmacokinetics, characterized by an increase in the total plasma clearance from 12.5 to 39.3 ml.min-1.kg-1 for dosage escalations in the range 10-50 mg/kg, while clearance values at doses of 50 and 100 mg/kg were similar. The terminal rate constants decreased linearly as the dose was increased from 10 to 100 mg/kg, eliciting an apparent prolongation of the biological half-life from 2.1 to 4.8 h. There was also a sequential increase in the steady state apparent volume of distribution from 1.73 to 8.75 l/kg. These observations are consistent with saturable binding of RG to plasma proteins while binding to tissue remains linear. Nevertheless, conversion to RA appeared to represent a prominent route of RG elimination. The molar plasma concentration of RA exceeded that of the prodrug within 30-90 min after i.v. treatment and declined very slowly thereafter, with plasma levels sustained between 0.1 and 10 microM for 48 h at each of the doses evaluated. Thus, RG effectively served as a slow release delivery system for RA, implying the possibility of maintaining therapeutic plasma levels of the drug from a more convenient dosing regimen than a continuous infusion schedule. The present findings, coupled with the demonstrated in vivo activity of RG against human brain tumor models, warrant its continued development as a much needed chemotherapeutic agent for the treatment of brain neoplasms.


Subject(s)
Antifungal Agents/pharmacokinetics , Polyenes/pharmacokinetics , Prodrugs/pharmacokinetics , Sirolimus/analogs & derivatives , Animals , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Male , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...