Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Proc Natl Acad Sci U S A ; 98(22): 12438-43, 2001 Oct 23.
Article in English | MEDLINE | ID: mdl-11606716

ABSTRACT

We have synthesized three peptides from the mdm-2 binding domain of human p53, residues 12-26 (PPLSQETFSDLWKLL), residues 12-20, and 17-26. To enable transport of the peptides across the cell membrane and at the same time to maximize the active mdm-2 binding alpha-helical conformation for these peptides, each was attached at its carboxyl terminus to the penetratin sequence, KKWKMRRNQFWVKVQRG, that contains many positively charged residues that stabilize an alpha-helix when present on its carboxyl terminal end. All three peptides were cytotoxic to human cancer cells in culture, whereas a control, unrelated peptide attached to the same penetratin sequence had no effect on these cell lines. The same three cytotoxic peptides had no effect on the growth of normal cells, including human cord blood-derived stem cells. These peptides were as effective in causing cell death in p53-null cancer cells as in those having mutant or normal p53. Peptide-induced cell death is not accompanied by expression of apoptosis-associated proteins such as Bax and waf(p21). Based on these findings, we conclude that the antiproliferative effects of these p53-derived peptides are not completely dependent on p53 activity and may prove useful as general anticancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Nuclear Proteins , Peptide Fragments/pharmacology , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/pharmacology , Amino Acid Sequence , Animals , Apoptosis/drug effects , Binding Sites , Cell Division/drug effects , Cell Line, Transformed , Female , Genes, ras , Humans , Molecular Sequence Data , Probability , Protein Conformation , Proto-Oncogene Proteins c-mdm2 , Rats , Stem Cells/drug effects , Tumor Suppressor Protein p53/chemistry , Xenopus laevis
2.
Oncology ; 60(4): 316-21, 2001.
Article in English | MEDLINE | ID: mdl-11408799

ABSTRACT

OBJECTIVE: To determine the clinical and laboratory response rate of a gemcitabine and docetaxel combination in human adenocarcinoma of the pancreas in vitro and in vivo. METHODS: Fifteen patients with unresectable pancreatic cancer were treated with gemcitabine, 900 mg/m(2), and docetaxel, 90 mg/m(2), every 3 weeks. Two human pancreatic cancer lines were tested in MTT assays for their response to titrations of gemcitabine and/or docetaxel at different time points and scheduling for biochemical synergy or additional antitumor effects. RESULTS: In vitro testing showed that these two agents were minimally effective alone but when combined, they displayed additional biochemical antiproliferative effects in MTT assays. With intent-to-treat analysis of all 15 patients, 4 patients (27%) achieved an objective response by CT scan, including one complete response. Seven patients (47%) had subjective improvement and decreased serum marker levels of CA 19-9. None of the 12 patients without prior therapy developed nadir white blood cell counts below 1,000/mm(3); 2 of 3 patients with prior radiation therapy developed nadir white blood cell counts below 1,000/mm(3). CONCLUSION: This regimen is well tolerated and appears to have a significant objective response rate. Gemcitabine and docetaxel antitumor effects are additive in vitro, which may help to explain the response rate.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Paclitaxel/analogs & derivatives , Pancreatic Neoplasms/drug therapy , Taxoids , Adenocarcinoma/diagnostic imaging , Adenocarcinoma/mortality , Adult , Aged , Aged, 80 and over , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Docetaxel , Drug Administration Schedule , Humans , Male , Middle Aged , Paclitaxel/administration & dosage , Pancreatic Neoplasms/diagnostic imaging , Pancreatic Neoplasms/mortality , Survival Rate , Tomography, X-Ray Computed , Treatment Outcome , Gemcitabine
4.
Proc (Bayl Univ Med Cent) ; 14(2): 130-3, 2001 Apr.
Article in English | MEDLINE | ID: mdl-16369601
5.
Neurosurgery ; 47(6): 1391-8; discussion 1398-9, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11126910

ABSTRACT

OBJECTIVE: Intracerebral clysis is a drug delivery technique that depends on convection-enhanced microinfusion to achieve therapeutic drug levels within the brain. In this study, brain tumor-bearing rats were treated with topotecan delivered systemically and by the intracerebral clysis method. Our objective was to determine the efficacy and tissue distribution of topotecan delivered by intracerebral clysis. METHODS: The C6/Wistar rat glioma model was used after a thymidine incorporation assay determined topotecan sensitivity of C6 cells in vitro. Long-term survival of animals provided objective measurements of efficacy; records of animal weight during treatment and neurological status served to approximate toxicity. Topotecan tissue penetration was measured in samples of ex vivo tumor and surrounding brain tissue with high-pressure liquid chromatography. RESULTS: Dose escalation demonstrated significant sensitivity of C6 glioma cells to topotecan (median lethal dose, 0.19 micromol/L). Eleven of 12 rats bearing established intracerebral C6 glioma and receiving topotecan by intracerebral clysis survived beyond the end point of 120 days; no untreated control or systemically treated animal survived beyond 26 days (n = 18; P < 0.005). Histopathological assessment of animals demonstrated significant tumor masses in the brains of intraperitoneally treated animals and untreated control animals. In contrast, no residual tumor was found in the brains of intracerebral clysis groups. Animal weights during treatment were markedly reduced by intraperitoneal dosing (n = 6) but not by low-dose intracerebral clysis (32 microg/kg/d for 5 d; n = 6). None of the low-dose intracerebral clysis-treated animals demonstrated neurological toxicity, and one high-dose intracerebral clysis-treated animal (160 microg/kg/d for 2 d; n = 6) died during follow-up. Topotecan was detected well beyond the boundaries of the tumor and even in the contralateral hemisphere in animals treated with intracerebral clysis. CONCLUSION: Topotecan delivered by the intracerebral clysis method is effective for treatment of brain tumors in the rat glioma model. These studies provide compelling justification for further preclinical testing to formally evaluate toxicity and efficacy with variable dosing schedules.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Glioma/drug therapy , Glioma/metabolism , Topotecan/pharmacokinetics , Topotecan/therapeutic use , Animals , Brain Neoplasms/pathology , Drug Delivery Systems , Glioma/pathology , Injections, Intraperitoneal , Neoplasm Transplantation , Rats , Rats, Wistar , Tissue Distribution , Topotecan/administration & dosage , Topotecan/toxicity , Tumor Cells, Cultured
6.
Neurosurgery ; 46(3): 683-91, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10719865

ABSTRACT

OBJECTIVE: Intracerebral clysis (ICC) is a new term we use to describe convection-enhanced microinfusion into the brain. This study establishes baseline parameters for preclinical, in vivo, drug investigations using ICC in a rat glioma model. METHODS: Intracranial pressure was measured, with an intraparenchymal fiber-optic catheter, in male Fischer rats 10, 15, 20, and 25 days after implantation of C6 glioma cells in the right frontal lobe (n = 80) and in control rats without tumor (n = 20), before and during ICC. A 25% albumin solution (100 microl) was infused through an intratumoral catheter at 0.5, 1.0, 2.0, 3.0, and 4.0 microl/min. Infusate distribution was assessed by infusion of fluorescein isothiocyanate-dextran (Mr 20,000), using the aforementioned parameters (n = 36). Brains were sectioned and photographed under ultraviolet light, and distribution was calculated by computer analysis (NIH Image for Macintosh). Safe effective drug distribution was demonstrated by measuring tumor sizes and apoptosis in animals treated with N,N'-bis(2-chloroethyl)-N-nitrosourea via ICC, compared with untreated controls. Magnetic resonance imaging noninvasively confirmed tumor growth before treatment. RESULTS: Intracranial pressure increased with tumor progression, from 5.5 mm Hg at baseline to 12.95 mm Hg on Day 25 after tumor cell implantation. Intracranial pressure during ICC ranged from 5 to 21 mm Hg and was correlated with increasing infusion volumes and increasing rates of infusion. No toxicity was observed, except at the higher ends of the tumor size and volume ranges. Fluorescein isothiocyanate-dextran distribution was greater with larger infusion volumes (30 microl versus 10 microl, n = 8, P < 0.05). No significant differences in distribution were observed when different infusion rates were compared while the volume was kept constant. At tolerated flow rates, the volumes of distribution were sufficient to promote adequate drug delivery to tumors. N,N'-Bis(2-chloroethyl)-N-nitrosourea treatment resulted in significant decreases in tumor size, compared with untreated controls. CONCLUSION: The C6 glioma model can be easily modified to study aspects of interstitial delivery via ICC and the application of ICC to the screening of potential antitumor agents for safety and efficacy.


Subject(s)
Brain Neoplasms/drug therapy , Drug Delivery Systems , Glioma/drug therapy , Animals , Antineoplastic Agents, Alkylating/administration & dosage , Brain/metabolism , Brain/pathology , Brain Neoplasms/physiopathology , Carmustine/administration & dosage , Dextrans/pharmacokinetics , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/pharmacokinetics , Glioma/physiopathology , Image Processing, Computer-Assisted , Injections , Intracranial Pressure , Magnetic Resonance Imaging , Male , Rats , Rats, Inbred F344 , Rats, Wistar , Tissue Distribution
7.
Oncogene ; 19(54): 6216-28, 2000 Dec 14.
Article in English | MEDLINE | ID: mdl-11175336

ABSTRACT

Mechanisms by which chemotherapeutic agents induce apoptosis are not completely understood. Current knowledge of the actual pharmacologic effects of chemotherapy and their biochemical mechanisms are better understood than the downstream events, which initiate the apoptotic cascade. The chemotherapeutic agent cisplatin causes DNA damage and can induce apoptosis in several types of human cancers. We found the formation of previously unreported nuclear complexes between the tumor suppressor protein p53 and the pro-apoptotic protein Bax, in human melanoma cell lines induced into apoptosis following cisplatin exposure. These detergent resistant complexes were detected: after wild type (wt) p53 and Bax increased in the nucleus; at the same time when active cytoplasmic apoptosis related protease, caspase 3/CPP32 appeared; and prior to the detection of apoptotic DNA fragmentation. Three channel fluorescence laser scanning confocal image microscopy revealed that the nuclear Bax/p53 complexes remained in the nucleus and localized proximal to DNA fragmentation sites as assayed by TUNEL after cisplatin exposure. Two human melanoma cell lines, expressing wt p53, were induced into apoptosis after cisplatin exposure, however they differed in the timing of this induction. In both cell lines the formation of nuclear Bax/p53 co-immunoprecipitable complexes correlated with the timing of the induction of apoptosis. The degree of apoptosis induced by different concentrations of cisplatin correlated with the amount of nuclear Bax/p53 complexes. The co-immunoprecipitation of Bax and p53 was found regardless of the antibodies tested and was specific since Bcl-xL/p53 complexes were not detected. Additionally, the human prostate cancer cell line, LNCaP, also formed nuclear Bax/p53 complexes only after apoptosis was induced by paclitaxel.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis , Cisplatin/pharmacology , Melanoma/metabolism , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Blotting, Western , Caspase 3 , Caspases/metabolism , Cell Cycle , Cell Nucleus/ultrastructure , DNA Fragmentation , Electrophoresis, Polyacrylamide Gel , Humans , Kinetics , Macromolecular Substances , Melanoma/pathology , Melanoma/ultrastructure , Microscopy, Confocal , Precipitin Tests , Tumor Cells, Cultured , bcl-2-Associated X Protein
9.
Proc (Bayl Univ Med Cent) ; 13(2): 144-7, 2000 Apr.
Article in English | MEDLINE | ID: mdl-16389368
11.
J Biol Chem ; 274(49): 34924-31, 1999 Dec 03.
Article in English | MEDLINE | ID: mdl-10574967

ABSTRACT

A p53-derived C-terminal peptide induced rapid apoptosis in breast cancer cell lines carrying endogenous p53 mutations or overexpressed wild-type (wt) p53 but was not toxic to nonmalignant human cell lines containing wt p53. Apoptosis occurred through a Fas/APO-1 signaling pathway involving increased extracellular levels of Fas/FasL in the absence of protein synthesis, as well as activation of a Fas/APO-1-specific protease, FLICE. The peptide activity was p53-dependent, and it had no effect in three tumor cell lines with null p53. Furthermore, the C-terminal peptide bound to p53 protein in cell extracts. Thus, p53-dependent, Fas/APO-1 mediated apoptosis can be induced in breast cancer cells with mutant p53 similar to the recently described Fas/APO-1 induced apoptosis by wt p53. However, mutant p53 without p53 peptide does not induce a Fas/APO-1 activation or apoptosis. Docking of the computed low energy conformations for the C-terminal peptide with those for a recently defined proline-rich regulatory region from the N-terminal domain of p53 suggests a unique low energy complex between the two peptide domains. The selective and rapid induction of apoptosis in cancer cells carrying p53 abnormalities may lead to a novel therapeutic modality.


Subject(s)
Apoptosis , Peptides/chemistry , Peptides/pharmacology , Tumor Suppressor Protein p53/metabolism , Amino Acid Sequence , Blotting, Western , Breast/drug effects , Breast/pathology , Breast Neoplasms/pathology , Caspase 8 , Caspase 9 , Caspases/metabolism , Cell Cycle , DNA/metabolism , Fas Ligand Protein , Fibroblasts/drug effects , Fibroblasts/pathology , Flow Cytometry , Genes, Homeobox , Humans , Membrane Glycoproteins/metabolism , Models, Molecular , Molecular Sequence Data , Peptides/genetics , Protein Conformation , Proteins/metabolism , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , fas Receptor/metabolism
12.
Biochem Pharmacol ; 58(10): 1587-92, 1999 Nov 15.
Article in English | MEDLINE | ID: mdl-10535749

ABSTRACT

To determine whether individual protein kinase C (PKC) isozymes differentially phosphorylate sites in the linker region of human P-glycoprotein (P-gp), we used a synthetic peptide substrate, PG-2, exactly corresponding to amino acid residues spanning the region 656-689 of the multidrug resistance gene (MDRI). All tested PKC isozymes phosphorylated PG-2. The maximum phosphate incorporation by calcium-dependent PKC isozymes alpha, betaI, betaII, and gamma was 3, 2, 2, and 3 mol phosphate/mol PG-2, respectively. The maximum phosphate incorporation by calcium-independent isozymes delta, epsilon, eta, and zeta was 1.5, 0.5, 1.5, and 1.5 mol phosphate/mol PG-2, respectively. Two-dimensional tryptic phosphopeptide mapping indicated differential phosphorylation of the PKC consensus sites Ser-661, Ser-667, and Ser-671 by individual isozymes, which may be functionally significant. These data suggest that differential phosphorylation by PKC isoenzymes of PKC sites within the P-gp linker region may play a role in modulating P-gp activity.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Isoenzymes/metabolism , Protein Kinase C/metabolism , Amino Acid Sequence , Humans , In Vitro Techniques , Molecular Sequence Data , Peptide Fragments/metabolism , Peptide Mapping , Phosphopeptides/analysis , Phosphorylation , Recombinant Proteins/metabolism
13.
Cancer Chemother Pharmacol ; 44(1): 74-80, 1999.
Article in English | MEDLINE | ID: mdl-10367752

ABSTRACT

PURPOSE: To determine whether tamoxifen plasma concentrations capable of blocking P-glycoprotein (Pgp) in vitro can be safely achieved in dogs and whether doxorubicin pharmacokinetic alterations occur when tamoxifen is coadministered. METHODS: Tamoxifen dose escalation studies were conducted in 7 normal dogs and in 19 tumor-bearing dogs receiving full-dose chemotherapy. Plasma tamoxifen and serum doxorubicin disposition were analyzed for putative drug interactions. RESULTS: Steady-state plasma concentrations of tamoxifen and N-desmethyl tamoxifen (NDMT) were 5-10 microM following oral tamoxifen administration at 600 mg/m2 every 12 h for 7 days to normal and tumor-bearing dogs. Mild-moderate gastrointestinal toxicity (diarrhea, anorexia) and reversible neurotoxicity were observed in dogs receiving chemotherapy plus high-dose tamoxifen. Myelosuppression was not affected by combined treatment in tumor-bearing dogs. High-dose tamoxifen decreased the clearance and volume of distribution of full-dose doxorubicin. CONCLUSIONS: Concentrations of tamoxifen/ NDMT sufficient to inhibit Pgp may be achieved in dogs receiving full-dose chemotherapy with a moderate but acceptable increase in gastrointestinal toxicity. Tamoxifen affects doxorubicin metabolism in dogs at high doses resulting in increased serum exposure. Pharmacologic manipulation of Pgp expression or function in normal and tumor tissue in dogs may facilitate investigation of novel anticancer treatment strategies in humans.


Subject(s)
Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Agents, Hormonal/pharmacokinetics , Doxorubicin/pharmacokinetics , Drug Resistance, Multiple , Tamoxifen/administration & dosage , Tamoxifen/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , Animals , Antineoplastic Agents, Hormonal/adverse effects , Digestive System/drug effects , Dogs , Dose-Response Relationship, Drug , Doxorubicin/adverse effects , Drug Interactions , Female , Male , Neoplasms, Experimental/metabolism , Tamoxifen/adverse effects , Tissue Distribution
15.
Otolaryngol Head Neck Surg ; 118(4): 464-71, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9560096

ABSTRACT

BACKGROUND: Squamous cell carcinoma of the head and neck (HNSCC) has a high incidence of recurrence and associated second primary malignancy. The retinoid 13-cis-retinoic acid has been shown to be effective as both a chemopreventive and chemotherapeutic agent for HNSCC, but often with treatment-limiting toxicity. The synthetic retinoid fenretinide (N-(4-hydroxyphenyl)retinamide) (HPR) has significant antiproliferative activity against a number of animal and human malignancies and has been used in clinical trials as a chemopreventive agent in patients with breast and prostate cancer and oral leukoplakia. HPR has been shown to have a toxicity profile lower than that for other retinoids used in clinical trials. PURPOSE: The aim of this study was to investigate the effect of HPR on the growth of HNSCC cell lines in vitro. METHODS: Four HNSCC cell lines (JHU-011-SCC, JHU-020-SCC, JHU-022-SCC, and FaDu) were treated with a range of concentrations of HPR for various times. After HPR exposure, cell viability was determined by tetrazolium dye (MTT) colorimetric assay, comparing cell survival with that of untreated control cells. HPR-induced apoptosis was determined by flow-cytometric deoxyribonucleic acid cell-cycle analysis, ultrastructural analysis with electron microscopy, and deoxyribonucleic acid fragmentation detected by gel electrophoresis. RESULTS: HPR caused significant growth inhibition in three of the four HNSCC cell lines in a dose- and time-dependent fashion. In two cell lines (JHU-011-SCC, JHU-020-SCC) a significant antiproliferative effect was achieved between 1 and 2.5 micromol/L HPR after 72 hours of treatment. By deoxyribonucleic acid cell-cycle analysis, electron microscopy, and gel electrophoresis, HPR was shown to induce apoptosis in the JHU-011-SCC and JHU-020-SCC cell lines, but not in the FaDu cell line, which was insensitive to the growth inhibitory effect of HPR. CONCLUSIONS: This study has demonstrated that HPR reduces cell viability in HNSCC cells in vitro at clinically relevant doses, with the growth inhibition occurring through the induction of apoptosis.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Squamous Cell/pathology , Fenretinide/pharmacology , Head and Neck Neoplasms/pathology , Tumor Cells, Cultured/drug effects , Cell Division/drug effects , Electrophoresis, Agar Gel , Flow Cytometry , Humans , In Vitro Techniques , Laryngeal Neoplasms/pathology , Microscopy, Electron , Tumor Cells, Cultured/pathology
16.
Biochem Pharmacol ; 55(4): 523-31, 1998 Feb 15.
Article in English | MEDLINE | ID: mdl-9514088

ABSTRACT

Hepatocellular carcinoma (HCC) is characterized by high drug resistance to currently available chemotherapeutic agents. In a prospective clinical study, we have demonstrated that high-dose tamoxifen significantly enhanced the therapeutic efficacy of doxorubicin in patients with far-advanced HCC. In a search for a possible mechanism, we found that tamoxifen at a clinically achievable concentration (2.5 microM) significantly enhanced doxorubicin-induced cytotoxicity and apoptosis of Hep-3B cells, a multidrug resistance (MDR)-1 expressing HCC cell line. This synergistic cytotoxic effect of tamoxifen, at this concentration, however, was not mediated by MDR inhibition. Instead, as evidenced by both western blot and immunofluorescence studies, tamoxifen inhibited the cytoplasmic-membrane translocation of protein kinase C (PKC)-alpha. 12-O-Tetradecanoylphorbol-13-acetate (TPA) restored the membrane translocation of PKC-alpha and abrogated the synergistic cytotoxicity of tamoxifen. We also showed that tamoxifen, at this concentration, did not directly affect the enzyme activity of PKC. Further, membrane translocation of other membrane-bound proteins, such as Ras protein, was similarly inhibited by tamoxifen, but could not be restored by the addition of TPA. Together, these data suggested that tamoxifen may act on the cytoplasmic membrane, and thereby inhibit PKC-alpha translocation to the membrane where it is activated. We hypothesize that high-dose tamoxifen may be an effective modulator of doxorubicin in the treatment of HCC, and suggest that biochemical modulation of PKC as a measure to improve systemic chemotherapy for HCC deserves further investigation.


Subject(s)
Apoptosis/drug effects , Doxorubicin/pharmacology , Protein Kinase C/metabolism , Tamoxifen/pharmacology , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents, Hormonal/agonists , Antineoplastic Agents, Hormonal/pharmacology , Biological Transport, Active/drug effects , Doxorubicin/administration & dosage , Drug Resistance , Drug Synergism , Enzyme Activation/drug effects , Fluorescent Dyes , Humans , Isoenzymes/metabolism , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/enzymology , Rhodamine 123 , Rhodamines , Tamoxifen/administration & dosage , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured , ras Proteins/metabolism
17.
Hepatogastroenterology ; 45(24): 1955-60, 1998.
Article in English | MEDLINE | ID: mdl-9951847

ABSTRACT

BACKGROUND/AIMS: In vitro data have indicated that tamoxifen (> 2.5 uM) significantly enhances the cytotoxic effect of doxorubicin in hepatocellular carcinoma (HCC) cells. This clinical study was conducted to examine whether tamoxifen, at a dose sufficient to result in a plasma concentration of more than 2.5 uM, may improve the therapeutic efficacy of doxorubicin in patients with advanced HCC. METHODOLOGY: A prospective phase II study was conducted. Eligible patients had unresectable and non-embolizable HCC, objectively measurable tumors, adequate neogram with absolute granulocyte count > 2,000/mm3 and platelet count > 1 x 10/mm3, total serum bilirubin < 3.0 mg/dl, age > or = 75 year, and a Karnofsky performance status < or = 50%. The treatment included oral tamoxifen 40 mg/m2, q.i.d, Day 1 to 7, and intravenous doxorubicin 60 mg/m2, Day 4, repeated every 3 weeks. RESULTS: Between May 1994 and December 1996, a total of 38 patients were enrolled in the study. Thirty-six patients were evaluable for tumor response and treatment-related toxicities. There were 32 men and 4 women, with a median age of 49 years. They received an average of 3.8 (range:1-12) courses of chemotherapy. ECOG (Eastern Cooperative Oncology Group) Grade 3-4 leucopenia and Grade 3-4 thrombocytopenia developed in 27.2% and 12.5% courses given, respectively. Gastrointestinal toxicity was generally mild. Three patients developed symptomatic cardiac toxicity. Twelve patients (33.3%, 95% confidence interval 17-51%) had achieved a partial remission (PR), with a median progression-free survival of 7 months. Median survivals of the responders and non-responders were 10 and 3 months, respectively (p<0.05). The median Karnofsky performance status of the responders improved from 74.0+/-6.3% to a post-chemotherapy value of 93.2+/-4.6% (p<0.05) CONCLUSIONS: High dose tamoxifen appears to be an effective biochemical modulator of doxorubicin in the treatment of HCC. Prospective randomized phase III studies comparing doxorubicin alone versus doxorubicin plus high-dose tamoxifen are needed.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Doxorubicin/administration & dosage , Liver Neoplasms/drug therapy , Tamoxifen/administration & dosage , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/mortality , Disease Progression , Dose-Response Relationship, Drug , Doxorubicin/adverse effects , Doxorubicin/pharmacokinetics , Drug Administration Schedule , Drug Synergism , Female , Follow-Up Studies , Humans , Liver Neoplasms/blood , Liver Neoplasms/mortality , Male , Middle Aged , Prospective Studies , Survival Rate , Tamoxifen/adverse effects , Tamoxifen/pharmacokinetics , Treatment Outcome
18.
Cell Growth Differ ; 8(1): 101-11, 1997 Jan.
Article in English | MEDLINE | ID: mdl-8993839

ABSTRACT

N-(4-Hydroxyphenyl)retinamide (4-HPR, Fenretinide) is a retinoid derivative with antineoplastic activity in various tumor types including prostate carcinoma. The mechanism of action of 4-HPR toxicity is unknown. 4-HPR induces apoptosis in leukemia- and lymphoma-derived cells, neuroblastoma, and small cell lung cancers. The present study was designed to investigate: (a) the mechanism of 4-HPR cytotoxicity in prostate cancer cells; and (b) correlate increased expression of transforming growth factor beta 1 (TGF beta 1) with induction of apoptosis. 4-HPR exposure to PC-3 cells in vitro was associated with apoptosis as evidenced by increased incidence of hypodiploid nuclei in propidium iodide fluorescence histograms and DNA fragmentation. An increase in the percentage of nuclei in the G1 phase of the cell cycle preceded induction of apoptosis. TGF beta 1-increased expression was noted in mRNA levels and in secretion of active TGF beta 1 into culture media. TGF beta 1 and TGF-beta receptor type II detected immunohistochemically were increased in 4-HPR-treated PC-3 cells. Furthermore, 4-HPR-induced cytotoxicity in PC-3 cells was abrogated by the addition of anti-TGF beta 1 antibody. In BT-20 cells, a 4-HPR-resistant breast carcinoma cell line, apoptosis was not observed after exposure to 4-HPR nor was TGF beta 1 expression enhanced in stained cells or in conditioned media. It is concluded that 4-HPR induces the expression of TGF beta 1 in association with the induction of apoptosis.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Fenretinide/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Transforming Growth Factor beta/metabolism , Breast Neoplasms/pathology , Female , Fenretinide/antagonists & inhibitors , Flow Cytometry , Humans , Immunohistochemistry , Male , Protein Serine-Threonine Kinases , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/metabolism , Time Factors , Tumor Cells, Cultured
19.
Biochim Biophys Acta ; 1291(2): 155-62, 1996 Oct 24.
Article in English | MEDLINE | ID: mdl-8898877

ABSTRACT

Since pesticides have been shown to interact with P-glycoprotein (P-gp), the purpose of this study was to examine the possible role of P-gp in pesticide resistance in the tobacco budworm (Heliothis virescens). Using three P-gp antibodies, P-gp expression in various resistant populations of tobacco budworms was found to be 2-6-times that of the susceptible larvae. Tobacco budworm P-gp was glycosylated and localized primarily in the cuticle and fat body with little expression in the mid gut. To determine the role of P-gp in pesticide resistance, resistant tobacco budworm larvae were treated with a P-gp inhibitor, quinidine, and challenged with various doses of thiodicarb. Inhibition of P-gp decreased the LD50 for thiodicarb by a factor of 12.5. Quinidine treatment did not result in a significant inhibition of the P-450 system nor did it alter the feeding of the larvae, suggesting the potential involvement of P-gp in pesticide resistance. An age-dependent increase in P-gp expression was detected in resistant larvae as compared to control, susceptible larvae. This correlates with the reported age-dependent increase in resistance and is further evidence supporting the role of P-gp in the development of pesticide resistance.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Drug Resistance, Multiple/physiology , Insecticides/toxicity , Lepidoptera/drug effects , Thiocarbamates/toxicity , ATP Binding Cassette Transporter, Subfamily B, Member 1/analysis , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/chemistry , Animals , Anisoles/metabolism , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Enzyme Inhibitors/pharmacology , Glycosylation , Larva/chemistry , Larva/drug effects , Lepidoptera/chemistry , Lepidoptera/growth & development , Lethal Dose 50 , Molecular Weight , Piperonyl Butoxide/pharmacology , Quinidine/pharmacology , Quinine/pharmacology , Vinblastine/pharmacology
20.
Biochem Pharmacol ; 52(4): 603-12, 1996 Aug 23.
Article in English | MEDLINE | ID: mdl-8759033

ABSTRACT

To investigate the role of protein kinase C (PKC) in the regulation of multidrug resistance and P-glycoprotein (P-gp) phosphorylation, the natural isomer of sphingosine (SPH), D-erythro sphingosine (De SPH), and its three unnatural stereoisomers were synthesized. The SPH isomers showed similar potencies as inhibitors of in vitro PKC activity and phorbol binding, with IC50 values of approximately 50 microM in both assays. Treatment of multidrug-resistant MCF-7ADR cells with SPH stereoisomers increased vinblastine (VLB) accumulation up to 6-fold at 50 microM but did not alter VLB accumulation in drug-sensitive MCF-7 wild-type (WT) cells or accumulation of 5-fluorouracil in either cell line. Phorbol dibutyrate treatment of MCF-7ADR cells increased phosphorylation of P-gp, and this increase was inhibited by prior treatment with SPH stereoisomers. Treatment of MCF-7ADR cells with SPH stereoisomers decreased basal phosphorylation of the P-gp, suggesting inhibition of PKC-mediated phosphorylation of P-gp. Most drugs that are known to reverse multidrug resistance, including several PKC inhibitors, have been shown to directly interact with P-gp and inhibit drug binding. SPH stereoisomers did not inhibit specific binding of [3H] VLB to MCF-7ADR cell membranes or [3H]azidopine photoaffinity labeling of P-gp or alter P-gp ATPase activity. These results suggest that SPH isomers are not substrates of P-gp and suggest that modulation of VLB accumulation by SPH stereoisomers is associated with inhibition of PKC-mediated phosphorylation of P-gp.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/drug effects , Breast Neoplasms/metabolism , Sphingosine/pharmacology , Vinblastine/metabolism , Animals , Drug Resistance, Multiple , Female , Humans , Phosphorylation , Rats , Stereoisomerism , Tumor Cells, Cultured/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...