Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Eur J Cancer ; 40(6): 890-8, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15120044

ABSTRACT

The introduction of imaging methods suitable for rodents offers opportunities for new anticancer efficacy models. Traditional models do not provide the level of sensitivity afforded by these precise and quantitative techniques. Bioluminescent endpoints, now feasible because of sensitive charge-coupled device cameras, can be non-invasively detected in live animals. Currently, the most common luminescence endpoint is firefly luciferase, which, in the presence of O(2) and ATP, catalyses the cleavage of the substrate luciferin and results in the emission of a photon of light. In vivo implantation of tumour cells transfected with the luciferase gene allows sequential monitoring of tumour growth within the viscera by measuring these photon signals. Furthermore, tumour cell lines containing the luciferase gene transcribed from an inducible promoter offer opportunities to study molecular-target modulation without the need for ex vivo evaluations of serial tumour samples. In conjunction with this, transgenic mice bearing a luciferase reporter mechanism can be used to monitor the tumour microenvironment as well as to signal when transforming events occur. This technology has the potential to reshape the efficacy evaluations and drug-testing algorithms of the future.


Subject(s)
Antineoplastic Agents/therapeutic use , Luminescent Measurements , Neoplasms/drug therapy , Animals , Cell Line, Tumor , Drug Evaluation , Humans , Luciferases , Mice , Mice, Transgenic , Models, Animal , Neoplasms/pathology , Photography
2.
Genome Biol ; 2(10): RESEARCH0041, 2001.
Article in English | MEDLINE | ID: mdl-11597333

ABSTRACT

BACKGROUND: Flavopiridol, a flavonoid currently in cancer clinical trials, inhibits cyclin-dependent kinases (CDKs) by competitively blocking their ATP-binding pocket. However, the mechanism of action of flavopiridol as an anti-cancer agent has not been fully elucidated. RESULTS: Using DNA microarrays, we found that flavopiridol inhibited gene expression broadly, in contrast to two other CDK inhibitors, roscovitine and 9-nitropaullone. The gene expression profile of flavopiridol closely resembled the profiles of two transcription inhibitors, actinomycin D and 5,6-dichloro-1-beta-D-ribofuranosyl-benzimidazole (DRB), suggesting that flavopiridol inhibits transcription globally. We were therefore able to use flavopiridol to measure mRNA turnover rates comprehensively and we found that different functional classes of genes had distinct distributions of mRNA turnover rates. In particular, genes encoding apoptosis regulators frequently had very short half-lives, as did several genes encoding key cell-cycle regulators. Strikingly, genes that were transcriptionally inducible were disproportionately represented in the class of genes with rapid mRNA turnover. CONCLUSIONS: The present genomic-scale measurement of mRNA turnover uncovered a regulatory logic that links gene function with mRNA half-life. The observation that transcriptionally inducible genes often have short mRNA half-lives demonstrates that cells have a coordinated strategy to rapidly modulate the mRNA levels of these genes. In addition, the present results suggest that flavopiridol may be more effective against types of cancer that are highly dependent on genes with unstable mRNAs.


Subject(s)
Antineoplastic Agents/pharmacology , Flavonoids/pharmacology , Gene Expression Regulation, Neoplastic , Lymphoma, B-Cell/genetics , Lymphoma, Large B-Cell, Diffuse/genetics , Piperidines/pharmacology , RNA Stability , Dactinomycin/pharmacology , Dichlororibofuranosylbenzimidazole/pharmacology , Gene Expression Profiling , Humans , Kinetics , Lymphoma, B-Cell/metabolism , Lymphoma, Large B-Cell, Diffuse/metabolism , Nucleic Acid Synthesis Inhibitors/pharmacology , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , RNA, Neoplasm/metabolism , Transcription, Genetic/drug effects , Tumor Cells, Cultured
3.
Cancer Res ; 61(10): 4030-7, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11358822

ABSTRACT

P-Glycoprotein-mediated drug efflux can yield a multidrug-resistance (MDR) phenotype that is associated with a poor response to cancer chemotherapy. Pervilleine A, a novel tropane alkaloid obtained from a chloroform extract of Erythroxylum pervillei as the result of bioactivity-guided fractionation, was found to restore the vinblastine sensitivity of cultured multidrug-resistant KB-V1 and CEM/VLB(100) cells, with IC(50) values of 0.36 and 0.02 microM, respectively. Similarly, the chemosensitivity of KB-8-5 cells to colchicine was restored with an IC(50) value of 0.61 microM. The mechanism of this response was evaluated with a number of model systems. First, incubation of multidrug-resistant KB-V1 and CEM/VLB(100) cells with up to 45 microM pervilleine A for 72 h did not significantly affect either the transcription of MDR1, as revealed by reverse transcriptional-PCR-based analysis of MDR1 mRNA, or levels of P-glycoprotein, as shown by Western blots. ATP-dependent binding of [(3)H]vinblastine observed with isolated multidrug-resistant KB-V1 cell membrane vesicles was inhibited by pervilleine A in a dose-dependent manner, and kinetic analysis indicted competitive inhibition with respect to vinblastine binding with a K(i) of 7.3 microM. Consistent with this effect, intracellular accumulation of [(3)H]vinblastine was increased from 0.18 pmol [(3)H]vinblastine/50 x 10(4) cells to approximately 5 pmol [(3)H]vinblastine/50 x 10(4) cells in the presence of 40 microM pervilleine A. To explore the potential relevance of these responses, KB-V1 or KB-8-5 cells were placed in hollow fibers and implanted into NCr nu/nu mice. Cell growth was not significantly inhibited when vinblastine or pervilleine A were administered as single agents, but when used in combination, inhibition of up to 75% was observed. Equimolar doses of verapamil were less effective. These data suggest that pervilleine A is an effective inhibitor of P-glycoprotein and should be further evaluated for clinical utility.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Drug Resistance, Multiple , Tropanes/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/pharmacology , Blotting, Western , Cell Division/drug effects , Colchicine/pharmacology , Drug Screening Assays, Antitumor , Genes, MDR/drug effects , Humans , Inhibitory Concentration 50 , KB Cells/drug effects , Phenotype , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured/drug effects , Verapamil/pharmacology , Vinblastine/pharmacokinetics , Vinblastine/pharmacology
4.
Anticancer Res ; 20(2A): 903-11, 2000.
Article in English | MEDLINE | ID: mdl-10810375

ABSTRACT

The hollow fiber assay, a unique in vivo model, permits the simultaneous evaluation of compound efficacy against multiple cell lines in two physiological compartments. This assay has been used to characterize in vivo activity of cytotoxic compounds. The purpose of the present study was to characterize and optimize this assay for compounds with a defined mechanism of action, specifically cell cycle inhibition. Two human tumor cell lines and one normal human cell line were loaded into polyvinylidene fluoride hollow fibers at two or more cell concentrations and grown in mice for 3-10 days. The data demonstrate the importance of characterizing the initial loading density of various cell lines in the evaluation of compounds. All studies were performed with cells in the linear part of the cell growth curves. Initial loading densities of 1-2 x 10(4) cells/fiber gave the greatest opportunity for growth in the three human cell lines tested (HCT116 colon carcinoma, NCI-H460 non-small cell carcinoma, and AG 1523 normal fibroblast). Utilizing the MTT assay, standard curves were constructed to correlate the final number of cells with optical density (OD) readings at 540 nm in order to calculate cell numbers in the fibers. Insights into the mechanism of action of cisplatin have been gained using Western blot analysis of the cell cycle markers PCNA (a protein present throughout the cell cycle) and Rb (a protein that acts as a tumor suppressor gene product) from the hollow fiber cells. In cisplatin-treated NCI-H460 cells both PCNA and Rb phosphorylation decreased, suggesting the arrest of the cells prior to the S phase. Standard therapeutic agents, cisplatin, racemic flavopiridol, cyclophosphamide and mitomycin C, were evaluated independently in the hollow fiber assay and the xenograft model. The data demonstrate that compounds active in the hollow fiber assay are also active in the xenograft.


Subject(s)
Antineoplastic Agents/toxicity , Cell Cycle/drug effects , Cell Survival/drug effects , Colonic Neoplasms/pathology , Drug Screening Assays, Antitumor/methods , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/pathology , Cell Division/drug effects , Cisplatin/toxicity , Cyclophosphamide/toxicity , Drug Screening Assays, Antitumor/instrumentation , Fibroblasts/cytology , Fibroblasts/drug effects , Flavonoids/toxicity , Humans , Lung Neoplasms/pathology , Male , Membranes, Artificial , Mice , Mice, Nude , Mitomycin/toxicity , Piperidines/toxicity , Polyvinyls , Proliferating Cell Nuclear Antigen/analysis , Retinoblastoma Protein/analysis , Transplantation, Heterologous , Tumor Cells, Cultured
5.
Biochem Pharmacol ; 58(6): 1057-66, 1999 Sep 15.
Article in English | MEDLINE | ID: mdl-10509758

ABSTRACT

The pyridine nucleotide 6-aminonicotinamide (6AN) was shown recently to sensitize a number of human tumor cell lines to cisplatin in vitro. The present studies were undertaken to compare the drug concentrations and length of exposure required for this sensitization in vitro with the drug exposure that could be achieved in mice in vivo. Human K562 leukemia cells and A549 lung cancer cells were incubated with 6AN for various lengths of time, exposed to cisplatin for 1-2 hr, and assayed for Pt-DNA adducts as well as the ability to form colonies. K562 cells displayed progressive increases in Pt-DNA adducts and cisplatin sensitivity during the first 10 hr of 6AN exposure. An 18-hr 6AN exposure was likewise more effective than a 6-hr 6AN exposure in sensitizing A549 cells to cisplatin. HPLC analysis of 6AN and its metabolite, 6-amino-NAD+, permitted assessment of exposures achieved in vivo after i.v. administration of 10 mg/kg of 6AN to CD2F1 mice. 6AN reached peak serum concentrations of 80-90 microM and was cleared rapidly, with T1/2alpha and T1/2beta values of 7.4 and 31.3 min, respectively. Bioavailability was 80-100% with identical plasma pharmacokinetics after i.p. administration. At least 25% of the 6AN was excreted unchanged in the urine. The metabolite 6-amino-NAD+ was detected in perchloric acid extracts of brain, liver, kidney, and spleen, but not in serum. Efforts to prolong systemic 6AN exposure by administering multiple i.p. doses or using osmotic pumps resulted in lethal toxicity. These results demonstrated that 6AN exposures required to sensitize tumor cells to cisplatin in vitro are difficult to achieve in vivo.


Subject(s)
6-Aminonicotinamide/pharmacokinetics , 6-Aminonicotinamide/administration & dosage , 6-Aminonicotinamide/metabolism , 6-Aminonicotinamide/pharmacology , Animals , Antineoplastic Agents/pharmacology , Cell Division/drug effects , Cisplatin/pharmacology , Drug Interactions , Humans , K562 Cells , Mice , NAD/analogs & derivatives , NAD/pharmacokinetics , Tissue Distribution , Tumor Cells, Cultured
6.
J Nat Prod ; 62(4): 633-5, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10217729

ABSTRACT

Bioassay-guided fractionation of organic extracts of the gorgonian Alertigorgia sp. has yielded the previously known suberosenone (1), a cytotoxic tricyclic sesquiterpene of the quadrone class, and alertenone (2), a dimer of suberosenone. The structure of 2 was determined by spectral analysis; the 1D TOCSY experiment was particularly useful in the structure elucidation. Comparison of the in vitro cytotoxicity of alertenone and suberosenone revealed that the dimeric alertenone was devoid of cytotoxicity below 35 microg/mL. In a hollow-fiber assay model of in vivo activity, suberosenone exhibited some growth inhibition of two of six tumor cell lines tested.


Subject(s)
Antineoplastic Agents/pharmacology , Cnidaria/chemistry , Sesquiterpenes/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Drug Screening Assays, Antitumor , Humans , Magnetic Resonance Spectroscopy , Mass Spectrometry , Models, Molecular , Sesquiterpenes/chemistry , Sesquiterpenes/isolation & purification , Spectrophotometry, Infrared , Spectrophotometry, Ultraviolet , Tumor Cells, Cultured
7.
Bioorg Med Chem Lett ; 9(2): 133-8, 1999 Jan 18.
Article in English | MEDLINE | ID: mdl-10021914

ABSTRACT

In vivo anti-HIV efficacy of (+)-calanolide A has been evaluated in a hollow fiber mouse model. It was demonstrated that the compound was capable of suppressing virus replication in two distinct and separate physiologic compartments (i.p. and s.c.) following oral or parenteral administration on a once- or twice-daily treatment schedule. A synergistic effect was observed for the combination of (+)-calanolide A and AZT.


Subject(s)
Anti-HIV Agents/pharmacology , Coumarins/pharmacology , Animals , Cell Line , Cell Survival , Cells, Cultured , Coumarins/administration & dosage , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Therapy, Combination , HIV Core Protein p24/blood , HIV Core Protein p24/drug effects , HIV Reverse Transcriptase/drug effects , HIV Reverse Transcriptase/metabolism , Humans , Macrophages/drug effects , Mice , Pyranocoumarins , Time Factors , Zidovudine/pharmacology
8.
Nucleosides Nucleotides ; 17(8): 1409-43, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9672703

ABSTRACT

The activity of a series of compounds related to adenosine-N1-oxide (1) and 1-(benzyloxy)adenosine (42) against vaccinia virus has been determined both in vitro and in a vaccinia mouse tailpox model. Significant activities have been found both in vitro and in vivo for a number of the synthetic compounds.


Subject(s)
Adenosine/analogs & derivatives , Adenosine/chemical synthesis , Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Cyclic N-Oxides/chemistry , Vaccinia virus/drug effects , Vaccinia/drug therapy , Adenosine/chemistry , Adenosine/pharmacology , Animals , Antiviral Agents/chemistry , Cyclic N-Oxides/pharmacology , Mice
9.
J Med Chem ; 41(13): 2184-93, 1998 Jun 18.
Article in English | MEDLINE | ID: mdl-9632350

ABSTRACT

Select chemokine receptors act as coreceptors for HIV-1 entry into human cells and represent targets for antiviral therapy. In this report we describe a distamycin analogue, 2,2'-[4, 4'-[[aminocarbonyl]amino]bis[N,4'-di[pryrrole-2-carboxamide- 1, 1'-dimethyl]]-6,8-naphthalenedisulfonic acid]hexasodium salt (NSC 651016), that selectively inhibited chemokine binding to CCR5, CCR3, CCR1, and CXCR4, but not to CXCR2 or CCR2b, and blocked chemokine-induced calcium flux. Inhibition was not due to nonspecific charge interactions at the cell surface, but was based on a specific competition for the ligand receptor interaction sites since the inhibitory effect was specific for some but not all chemoattractant receptors. NSC 651016 inhibited in vitro replication of a wide range of HIV-1 isolates, as well as HIV-2 and SIV, and exhibited in vivo anti-HIV-1 activity in a murine model. In contrast, a distamycin analogue with similar structure and charge and the monomeric form of NSC 651016 demonstrated no inhibitory effects. These data demonstrate that molecules which interfere with HIV-1 entry into cells by targeting specific chemokine coreceptors can provide a viable approach to anti-HIV-1 therapy. NSC 651016 represents an attractive candidate for the chemotherapeutic treatment of HIV-1 infection and as a microbicide to prevent the sexual transmisssion of HIV-1. Moreover, NSC 651016 can serve as a template for medicinal chemical modifications leading to more effective antivirals.


Subject(s)
Anti-HIV Agents/pharmacology , HIV-1/drug effects , Naphthalenesulfonates/pharmacology , Receptors, Chemokine/antagonists & inhibitors , Virus Replication/drug effects , Administration, Cutaneous , Animals , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacokinetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/metabolism , Binding, Competitive , Calcium/metabolism , Cell Line , Dimerization , Drug Resistance, Microbial , HIV-1/isolation & purification , HIV-1/metabolism , HIV-1/physiology , HIV-2/drug effects , HIV-2/isolation & purification , Humans , Injections, Intraperitoneal , Injections, Intravenous , Ligands , Membrane Fusion/drug effects , Mice , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Naphthalenesulfonates/administration & dosage , Naphthalenesulfonates/chemistry , Naphthalenesulfonates/pharmacokinetics , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/immunology , Reverse Transcriptase Inhibitors/pharmacology , Signal Transduction/drug effects , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/isolation & purification
10.
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
11.
Nat Med ; 3(4): 395-401, 1997 Apr.
Article in English | MEDLINE | ID: mdl-9095172

ABSTRACT

We have used the COMPARE computer algorithm and Nm23 expression as a marker of tumor metastatic potential to examine the in vitro antiproliferative activity of chemotherapeutic drugs on human breast carcinoma and melanoma cell lines. None of 171 compounds in clinical use or under development and only 40 of 30,000 repository compounds exhibited preferential growth inhibition of low-Nm23-expressing, metastatically aggressive cell lines with a Pearson correlation coefficient of < or = -0.64. Characterization of one compound, NSC 645306, is presented including in vivo activity in a hollow fiber assay. The data demonstrate a novel approach to drug identification for aggressive human tumors.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/analysis , Breast Neoplasms/drug therapy , Melanoma/drug therapy , Monomeric GTP-Binding Proteins , Nucleoside-Diphosphate Kinase , Transcription Factors/analysis , Algorithms , Female , Humans , NM23 Nucleoside Diphosphate Kinases , Neoplasm Metastasis , Tumor Cells, Cultured
12.
J Virol Methods ; 62(1): 63-70, 1996 Oct.
Article in English | MEDLINE | ID: mdl-8910649

ABSTRACT

Rauscher murine leukemia virus induces an erythroleukemia in susceptible strains of mice that is associated with splenomegaly and viremia. This animal model has been used for evaluating the in vivo efficacy of potential anti-HIV agents. The in vivo antiviral activity of therapeutic agents has usually been determined by measuring a reduction in the spleen weights of compound-treated mice or by quantitating viremia with the UV-XC plaque assay. The UV-XC assay, however, is time-consuming and labor-intensive. Virions of Rauscher murine leukemia virus, like other retroviruses, contain the enzyme reverse transcriptase. Quantitating the level of this enzyme in infected mouse sera provides a more rapid measure of viremia in the animal. We have examined the effects of several reagents, including detergent, KCl, EGTA, dGMP, spermine, as well as protease and RNase inhibitors, on the reverse transcriptase assay. The optimized assay method was effective in evaluating the antiviral activity of AZT in the Rauscher murine leukemia virus in vivo model. The assay is also amenable to automation if large numbers of assays are required.


Subject(s)
Leukemia, Experimental , RNA-Directed DNA Polymerase/analysis , Rauscher Virus/isolation & purification , Retroviridae Infections , Tumor Virus Infections , Animals , HIV Infections/drug therapy , Mice , RNA-Directed DNA Polymerase/drug effects , Viral Load , Zidovudine/pharmacology
13.
Antiviral Res ; 27(3): 317-23, 1995 Jun.
Article in English | MEDLINE | ID: mdl-8540752

ABSTRACT

Ribavirin has been reported to enhance the activity of ddI against HIV. We explored this enhancement of antiviral activity in Rauscher murine leukemia virus (RMuLV) models in vitro and in vivo. The significant finding in these studies was that combinations of the drugs exhibited virus titer reductions that were greater than would be expected if the drug interactions were simply additive. These effects were designated synergistic by the method of Prichard and Shipman (Prichard, M.N. and Shipman, C., Jr. (1990). A three-dimensional model to analyze drug-drug interaction, Antiviral Res. 14, 181-206). In addition to the antiviral synergy, we also observed some synergistic toxicity in the animal model.


Subject(s)
Didanosine/pharmacology , Leukemia, Experimental/drug therapy , Rauscher Virus/drug effects , Retroviridae Infections/drug therapy , Ribavirin/pharmacology , Tumor Virus Infections/drug therapy , Animals , Cell Line , Drug Synergism , Leukemia, Experimental/virology , Male , Mice , Rauscher Virus/isolation & purification , Weight Gain
14.
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
15.
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
16.
AIDS Res Hum Retroviruses ; 9(11): 1097-106, 1993 Nov.
Article in English | MEDLINE | ID: mdl-7508723

ABSTRACT

R82913 and R86183, two derivatives of tetrahydroimidazo[4,5,1-jk][1,4]-benzodiazepin-2(1H)-thione (TIBO), were found to potently and selectively inhibit the replication and cell killing effects of a panel of biologically diverse laboratory and clinical strains of HIV-1. The two compounds exhibited significant activity in all human cell lines tested, as well as in fresh human peripheral blood lymphocytes and macrophages. One of these two compounds (R82913) was found to significantly inhibit the replication of a murine retrovirus (Rauscher murine leukemia virus) in both UV-XC plaque formation and virus yield reduction assays. R86183, despite differing from R82913 only in the positioning of a single chlorine molecule, was not active against the murine retrovirus but was 10-fold more potent in inhibiting HIV-1 replication. Combination antiviral assays with other reverse transcriptase inhibitors, including AZT, ddC, and carbovir, yielded synergistic anti-HIV activity with both TIBO derivatives. Additive to slightly synergistic results were obtained in combinations with ddI and phosphonoformic acid whereas additive to antagonistic activity was detected in combination with dextran sulfate.


Subject(s)
Antiviral Agents/pharmacology , Benzodiazepines/pharmacology , HIV-1/drug effects , Imidazoles/pharmacology , Rauscher Virus/drug effects , Animals , Antiviral Agents/administration & dosage , Benzodiazepines/administration & dosage , Cell Line , Didanosine/administration & dosage , Drug Synergism , HIV Reverse Transcriptase , HIV-1/physiology , HIV-2/drug effects , HIV-2/physiology , Humans , Imidazoles/administration & dosage , Mice , Rauscher Virus/physiology , Reverse Transcriptase Inhibitors , Virus Replication/drug effects , Zidovudine/administration & dosage
17.
Antiviral Res ; 21(3): 247-65, 1993 Jul.
Article in English | MEDLINE | ID: mdl-7692815

ABSTRACT

Thiazolobenzimidazole (NSC 625487) was a highly potent inhibitor of human immunodeficiency virus-induced cell killing and viral replication in a variety of human cell lines, as well as fresh human peripheral blood lymphocytes and macrophages. The compound was active against a panel of biologically diverse laboratory and clinical strains of HIV-1, including the AZT-resistant strain G910-6. However, the agent was inactive against HIV-2 and a pyridinone-resistant strain (A17) of HIV-1, a strain which is cross-resistant to several structurally diverse members of a common pharmacologic class of nonnucleoside reverse transcriptase inhibitors. The compound selectively inhibited HIV-1 reverse transcriptase but not HIV-2 reverse transcriptase. Combinations of thiazolobenzimidazole with either AZT or ddI synergistically inhibited HIV-1 induced cell killing in vitro. Thiazolobenzimidazole also inhibited the replication of the Rauscher murine leukemia retrovirus. Thus, thiazolobenzimidazole is a new active anti-HIV-1 chemotype and may represent a subclass of nonnucleoside reverse transcriptase inhibitors with an enhanced range of anti-retroviral activity.


Subject(s)
Antiviral Agents/pharmacology , Benzimidazoles/pharmacology , HIV-1/drug effects , Reverse Transcriptase Inhibitors , Thiazoles/pharmacology , Base Sequence , Cell Line , DNA, Single-Stranded , Didanosine/pharmacology , Drug Synergism , HIV Reverse Transcriptase , HIV-1/enzymology , Humans , Leukemia Virus, Murine/drug effects , Molecular Sequence Data , Molecular Structure , Zidovudine/pharmacology
18.
Antiviral Res ; 18(3-4): 267-74, 1992 Jun.
Article in English | MEDLINE | ID: mdl-1416908

ABSTRACT

A system for evaluating the activity of antiviral agents against Rauscher murine leukemia virus (R-MuLV) has been developed using an enzyme linked immunosorbent assay technique. The activity of various antiviral compounds demonstrated in this assay system has been compared to their activity in the UV-XC plaque reduction assay, which has been used historically for evaluating anti-R-MuLV compounds. The assay is based upon detection of R-MuLV encoded p30 protein production in virus infected murine cells. The assay reagents are readily available and the assay system is amenable to automated data collection systems. Cytotoxicity evaluations are conducted in parallel to the Rauscher MuLV ELISA assay in order to assess drug-induced reductions in cell viability. Cytotoxicity evaluations are important to interpretation of the ELISA results since reductions in cell viability reduce viral protein production which would indicate an antiviral drug effect. This system is less sensitive than the classical UV-XC plaque reduction assay; however, it does offer an alternative to the time-consuming and labor-intensive plaque assay.


Subject(s)
Antiviral Agents/therapeutic use , Enzyme-Linked Immunosorbent Assay/methods , Leukemia, Experimental/drug therapy , Rauscher Virus , Animals , Antiviral Agents/metabolism , Mice , Microbial Sensitivity Tests/methods , Viral Plaque Assay
19.
Article in English | MEDLINE | ID: mdl-1517963

ABSTRACT

Over 50 different commercially available sulfonic acid-containing dyes were analyzed for their ability to prevent HIV-1-induced cell killing and in inhibiting HIV-1 replication. Compounds of remarkably similar structure, but with differing patterns of sulfonic acid group substitutions, had a wide range of potency in inhibiting HIV-1. Chicago sky blue (CSB) was highly effective in the inhibition of HIV-1 with less toxicity to CEM-SS cells than most of the other sulfonated dyes tested. Synthesis of CSB was undertaken to produce a product greater than 98% pure and this compound was used to elucidate the possible mechanisms by which this class of structurally related compounds inhibits HIV-1. Addition of CSB to cells infected at high multiplicity at any time up to 24 h after infection, unlike dideoxycytidine (ddC) or oxathiin carboxanilide (OC), inhibited HIV-1-induced cell killing. Other postinfection time course studies revealed that CSB had to be present for 24 h or longer immediately after infection to be protective. Virus binding to cells occurred in the presence of CSB, but the requirement for virion envelope-cell membrane fusion was delayed. CSB was a potent inhibitor of the reverse transcriptase (RT) of both HIV-1 and HIV-2, although it was less active against HIV-2 in a cell killing-based assay. CSB also inhibited Rauscher and LP-BM5 murine leukemia viruses. CSB appears to disrupt the interaction between viral proteins and cell membranes, both in the fusion step early in the infection cycle and in the development of syncytia in the late stages of virus infection.


Subject(s)
Antiviral Agents/pharmacology , Coloring Agents/pharmacology , HIV-1/drug effects , Sulfonic Acids/pharmacology , Antiviral Agents/chemistry , Azo Compounds/pharmacology , Cell Line , Coloring Agents/chemistry , Giant Cells/microbiology , HIV-1/physiology , Humans , Molecular Structure , Sulfonic Acids/chemistry , Virus Replication/drug effects
20.
Antimicrob Agents Chemother ; 36(1): 206-8, 1992 Jan.
Article in English | MEDLINE | ID: mdl-1317146

ABSTRACT

A virus-host cell system in which human cytomegalovirus-infected human cells are entrapped in agarose plugs has been developed. This model provides an inexpensive method for the in vivo evaluation (with outbred, immunocompetent mice) of antiviral drugs against human viruses such as cytomegalovirus that replicate primarily or only in human cells.


Subject(s)
Antiviral Agents/therapeutic use , Cytomegalovirus Infections/drug therapy , Animals , Cells, Cultured , Cytomegalovirus/drug effects , Drug Evaluation, Preclinical/methods , Ganciclovir/therapeutic use , Humans , Mice , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...