126 results on '"Hait, W N"'
Search Results
2. Development of New Treatments for Prostate Cancer
- Author
-
DiPaola, R. S., primary, Abate-Shen, C., additional, and Hait, W. N., additional
- Published
- 2005
- Full Text
- View/download PDF
3. Silencing of stathmin induces tumor-suppressor function in breast cancer cell lines harboring mutant p53
- Author
-
Alli, E, primary, Yang, J-M, additional, and Hait, W N, additional
- Published
- 2006
- Full Text
- View/download PDF
4. Phase I Clinical and Pharmacologic Study of 13-cis-Retinoic Acid, Interferon Alfa, and Paclitaxel in Patients With Prostate Cancer and Other Advanced Malignancies
- Author
-
DiPaola, R. S., primary, Rafi, M. M., additional, Vyas, V., additional, Toppmeyer, D., additional, Rubin, E., additional, Patel, J., additional, Goodin, S., additional, Medina, M., additional, Medina, P., additional, Zamek, R., additional, Zhang, C., additional, White, E., additional, Gupta, E., additional, and Hait, W. N., additional
- Published
- 1999
- Full Text
- View/download PDF
5. Clinical and Biologic Activity of an Estrogenic Herbal Combination (PC-SPES) in Prostate Cancer
- Author
-
DiPaola, R. S., primary, Zhang, H., additional, Lambert, G. H., additional, Meeker, R., additional, Licitra, E., additional, Rafi, M. M., additional, Zhu, B. T., additional, Spaulding, H., additional, Goodin, S., additional, Toledano, M. B., additional, Hait, W. N., additional, and Gallo, M. A., additional
- Published
- 1999
- Full Text
- View/download PDF
6. Activity and regulation by growth factors of calmodulin-dependent protein kinase III (elongation factor 2-kinase) in human breast cancer
- Author
-
Parmer, T G, primary, Ward, M D, additional, Yurkow, E J, additional, Vyas, V H, additional, Kearney, T J, additional, and Hait, W N, additional
- Published
- 1998
- Full Text
- View/download PDF
7. Sensitivity of K562 human chronic myelogenous leukemia blast cells transfected with a human multidrug resistance cDNA to cytotoxic drugs and differentiating agents.
- Author
-
Hait, W N, primary, Choudhury, S, additional, Srimatkandada, S, additional, and Murren, J R, additional
- Published
- 1993
- Full Text
- View/download PDF
8. Selective Cyclic Nucleotide Phosphodiesterase Inhibitors as Potential Therapeutic Agents.
- Author
-
Weiss, B and Hait, W N
- Published
- 1977
- Full Text
- View/download PDF
9. Cyclosporine treatment of refractory T-cell lymphomas.
- Author
-
Cooper, Dennis L., Braverman, Irwin M., Sarris, Andreas H., Durivage, Henry J., Saidman, Bruce H., Davis, Carol A., Hait, William N., Cooper, D L, Braverman, I M, Sarris, A H, Durivage, H J, Saidman, B H, Davis, C A, and Hait, W N
- Published
- 1993
- Full Text
- View/download PDF
10. Biochemical modulation of 5-fluorouracil with brequinar: results of a phase I study.
- Author
-
Buzaid, Antonio, Pizzorno, Giuseppe, Marsh, John, Ravikumar, Thanjavur, Murren, John, Todd, Mary, Strair, Roger, Poo, Wen-Jen, Hait, William, Buzaid, A C, Pizzorno, G, Marsh, J C, Ravikumar, T S, Murren, J R, Todd, M, Strair, R K, Poo, W J, and Hait, W N
- Abstract
Biochemical modulation can increase the efficacy of 5-fluorouracil (5-FU). Pizzorno et al. have previously shown that brequinar, a de novo pyrimidine synthesis inhibitor, enhances the antitumor effect of 5-FU in vivo [Cancer Res 52: 1660-1665, 1992]. On the basis of their data, we conducted a phase I study of brequinar in combination with 5-FU in patients with refractory solid tumors. The initial dose (100 mg/m2) of brequinar was raised in 100-mg/m2 increments in cohorts of three assessable patients. The initial dose of 5-FU was 500 mg/m2, but escalation was allowed in patients who showed no significant toxic reaction. Brequinar was administered over 1 h and 5-FU over 2 h starting 18-20 h after the initiation of infusion of brequinar. Treatments were repeated weekly. Responses were evaluated after 4 weeks (one course) and then every 8 weeks thereafter. Pharmacokinetics of brequinar and determination of plasma uridine levels were performed in at least three patients at each dose level. Of the 25 patients registered in the study, 21 were assessable for toxicity studies. The dose of brequinar was escalated up to 600 mg/m2. In addition, the dose of 5-FU was increased to 600 mg/m2 as a result of a lack of a significant toxic reaction in the first nine patients. No objective responses were observed. One patient developed grade 3 stomatitis, and one developed grade 3 esophagitis at the 400 and 600 mg/m2 dose of brequinar, respectively. Brequinar produced a dose-dependent decrease in plasma uridine levels at doses up to 500 mg/m2. No additional decrease in plasma uridine occurred with higher doses of brequinar, thus suggesting a plateau effect. This observation prompted us to terminate the study before reaching the maximum tolerated dose of brequinar. Our data indicate that brequinar in doses > or = 400 mg/m2 results in significant biochemical modulation. The lack of toxicity seen at these doses of brequinar suggests that the initial dose of the effector agent 5-FU should be increased in future studies. [ABSTRACT FROM AUTHOR]
- Published
- 1995
- Full Text
- View/download PDF
11. Phase I trial of combined therapy with bleomycin and the calmodulin antagonist, trifluoperazine.
- Author
-
Hait, William, Morris, Steve, Lazo, John, Figlin, Robert, Durivage, Henry, White, Kathleen, Schwartz, Peter, Hait, W N, Morris, S, Lazo, J S, Figlin, R J, Durivage, H J, White, K, and Schwartz, P E
- Subjects
ANTINEOPLASTIC agents ,ANTIPSYCHOTIC agents ,BLEOMYCIN ,CLINICAL trials ,COMPARATIVE studies ,DRUG administration ,CLINICAL drug trials ,RESEARCH methodology ,MEDICAL cooperation ,PHENOTHIAZINE ,RESEARCH ,RESEARCH funding ,TUMORS ,EVALUATION research - Abstract
Calmodulin antagonists, such as trifluoperazine, can enhance the cytotoxic effects of bleomycin both in tissue culture and in vivo. Therefore, we evaluated the effects of combination treatment with these drugs in a phase I clinical trial. Patients with objectively measurable or evaluable cancer refractory to conventional treatment who had an acceptable performance status (ECOG 0-2) and acceptable laboratory studies were eligible. All patients gave written informed consent. A cycle of therapy consisted of three weekly treatments with trifluoperazine (days 1-4) and 30 IU bleomycin (day 3). After three patients completed a cycle of therapy without experiencing dose-limiting toxicity, new patients were entered in the study and received a higher dose of trifluoperazine. The dose of bleomycin remained constant. Evaluable patients received at least 2 weeks of treatment and survived for 6 weeks; of 19 patients, 2 were unevaluable. The major toxicities were neurological and pulmonary and included one case of fatal pneumonia with interstitial pulmonary fibrosis. There was no hematologic toxicity. Two patients underwent partial responses (PRs) and two had complete responses (CRs). We conclude that trifluoperazine can safely be given with bleomycin and that further study of the potential efficacy of this treatment is indicated. [ABSTRACT FROM AUTHOR]
- Published
- 1989
- Full Text
- View/download PDF
12. Inhibition of growth of leukemic cells by inhibitors of calmodulin: phenothiazines and melittin.
- Author
-
Hait, William, Grais, Linda, Benz, Constance, Cadman, Ed., Hait, W N, Grais, L, Benz, C, and Cadman, E C
- Abstract
Calmodulin, a ubiquitous calcium-binding protein, has recently been shown to play an important role in cellular proliferation. The calmodulin inhibitors melittin, trifluoperazine, and chlorpromazine inhibited the growth and clonogenicity of human and murine leukemic cells, and their potency reflected their activity as inhibitors of calmodulin. Melittin, which is a far more potent inhibitor of calmodulin activity, was also a more potent inhibitor of cell growth and clonogenicity. The less active phenothiazine metabolite, chlorpromazine sulfoxide, had much less potent cytotoxic activity. [ABSTRACT FROM AUTHOR]
- Published
- 1985
- Full Text
- View/download PDF
13. Activity and regulation by growth factors of calmodulin-dependent protein kinase III (elongation factor 2-kinase) in human breast cancer.
- Author
-
Parmer, T G, Ward, M D, Yurkow, E J, Vyas, V H, Kearney, T J, and Hait, W N
- Subjects
PROTEIN kinases ,BREAST cancer ,MITOGENS - Abstract
Calmodulin-dependent protein kinase III (CaM kinase III, elongation factor-2 kinase) is a unique member of the Ca2+/CaM-dependent protein kinase family. Activation of CaM kinase III leads to the selective phosphorylation of elongation factor 2 (eEF-2) and transient inhibition of protein synthesis. Recent cloning and sequencing of CaM kinase III revealed that this enzyme represents a new superfamily of protein kinases. The activity of CaM kinase III is selectively activated in proliferating cells; inhibition of the kinase blocked cells in G0/G1-S and decreased viability. To determine the significance of CaM kinase III in breast cancer, we measured the activity of the kinase in human breast cancer cell lines as well as in fresh surgical specimens. The specific activity of CaM kinase III in human breast cancer cell lines was equal to or greater than that seen in a variety of cell lines with similar rates of proliferation. The specific activity of CaM kinase III was markedly increased in human breast tumour specimens compared with that of normal adjacent breast tissue. The activity of this enzyme was regulated by breast cancer mitogens. In serum-deprived MDA-MB-231 cells, the combination of insulin-like growth factor I (IGF-I) and epidermal growth factor (EGF) stimulated cell proliferation and activated CaM kinase III to activities observed in the presence of 10% serum. Inhibition of enzyme activity blocked cell proliferation induced by growth factors. In MCF-7 cells separated by fluorescence-activated cell sorting. CaM kinase III was increased in S-phase over that of other phases of the cell cycle. In summary, the activity of Ca2+/CaM-dependent protein kinase III is controlled by breast cancer mitogens and appears to be constitutively activated in human breast cancer. These results suggest that CaM kinase III may contribute an important link between growth factor/receptor interactions, protein synthesis and the induction of cellular proliferation in human breast cancer. [ABSTRACT FROM AUTHOR]
- Published
- 1999
- Full Text
- View/download PDF
14. Rapid Stem Cell Differentiation Induced by 19-Nortestosterone Decanoate.
- Author
-
Gorshein, D., Hait, W. N., Besa, E. C., Jepson, Joanne H., and Gardner, F. H.
- Published
- 1974
- Full Text
- View/download PDF
15. Potentiation of DNA damage and cytotoxicity by calmodulin antagonists
- Author
-
Rosenthal, S. A. and Hait, W. N.
- Subjects
Bleomycin ,Sulfonamides ,Hot Temperature ,Calmodulin ,DNA Repair ,Cell Survival ,Tumor Cells, Cultured ,Animals ,Humans ,Antineoplastic Agents ,Research Article ,DNA Damage - Abstract
Many physical and chemical agents used in anti-neoplastic therapy are known to act by effecting DNA damage. The possibility that calmodulin may be an important modulator of the response to DNA damage in eukaryotic cells has been explored in a number of studies which use calmodulin antagonists in combination with agents known to damage DNA. We review these studies and discuss the therapeutic potential of calmodulin antagonists in combination therapy.
- Published
- 1988
16. Interactions of 1-methyl-4-phenylpyridinium and other compounds with P-glycoprotein: relevance to toxicity of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine
- Author
-
Staal, R. G., Yang, J. M., Hait, W. N., and Sonsalla, P. K.
- Published
- 2001
- Full Text
- View/download PDF
17. Pharmacology of drugs that alter multidrug resistance in cancer.
- Author
-
Ford, J M and Hait, W N
- Published
- 1990
- Full Text
- View/download PDF
18. Cyclic nucleotide phosphodiesterase of normal and leukemic lymphocytes. Kinetic properties and selective alteration of the activity of the multiple molecular forms.
- Author
-
Hait, W N and Weiss, B
- Published
- 1979
19. Biochemical correlates of the antitumor and antimitochondrial properties of gossypol enantiomers.
- Author
-
Benz, C C, Keniry, M A, Ford, J M, Townsend, A J, Cox, F W, Palayoor, S, Matlin, S A, Hait, W N, and Cowan, K H
- Abstract
Racemic gossypol has been shown to have antitumor properties that may be due to its ability to uncouple tumor mitochondria or to its inhibitory effects on a variety of nonmitochondrial enzymes. We have studied the antimitochondrial and enzyme-inhibiting properties of gossypol in human carcinoma cell lines of breast (MCF-7, T47-D), ovarian (OVCAR-3) colon (HCT-8), and pancreatic (MiaPaCa) origin by comparing the effects of its purified (+)- and (-)-enantiomers. (-)-Gossypol shows up to 10-fold greater antiproliferative activity than (+)-gossypol in the cancer cell lines and in normal hematopoietic stem cells grown in vitro, with IC50 values ranging from 1.5 to 4.0 microM for the cancer cells and from 10 to 20 microM for the human marrow stem cells. As well, multidrug-resistant MCF/Adr cells appear more resistant to (-)-gossypol than their parental cell line. Electron microscopy indicates that the earliest ultrastructural change in tumor cells exposed to a cytotoxic (10 microM) concentration of (-)-gossypol is the selective destruction of their mitochondria. Consistent with this observation, 31P magnetic resonance spectroscopy detects pronounced changes in tumor cell high energy phosphate metabolism within 24 hr of (-)-gossypol treatment, manifest by 1.6- to greater than 50-fold differential reductions in the intracellular ratios of ATP/Pi, relative to (+)-gossypol-treated cell lines; the magnitude of these antimitochondrial effects correlates with the antiproliferative activity of (-)-gossypol. Northern blot RNA analyses suggest that treatment with a 5-10 microM dose of (-)-gossypol induces a transient increase in the expression of heat shock gene products, particularly hsp-70 transcripts. The mean 5-fold increase in (-)-gossypol-induced hsp-70 mRNA appears coincident with a comparable heat-stimulated increase in transcript levels, as compared with control or (+)-gossypol-treated cells. The enzyme-inhibiting properties of gossypol enantiomers were compared in cell-free assays measuring glutathione-S-transferase-alpha, -mu, and pi activities, calmodulin stimulation of cyclic nucleotide phosphodiesterase, and protein kinase C activity. Both enantiomers are near equivalent antagonists of calmodulin stimulation and protein kinase C activity, exceeding the potency of known inhibitors such as phenothiazines by as much as 50-fold. In contrast, (-)-gossypol is a 3-fold more potent inhibitor of glutathione-S-transferase-alpha and -pi isozyme activity, resulting in IC50 values of 1.6 and 7.0 microM, respectively, for these two isozymes.(ABSTRACT TRUNCATED AT 400 WORDS)
- Published
- 1990
20. Structural features determining activity of phenothiazines and related drugs for inhibition of cell growth and reversal of multidrug resistance.
- Author
-
Ford, J M, Prozialeck, W C, and Hait, W N
- Abstract
Phenothiazines and structurally related compounds inhibit cellular proliferation and sensitize multidrug-resistant (MDR) cells to chemotherapeutic agents. To identify more potent pharmaceuticals, we studied the structure-activity relationships of 30 phenothiazines and related compounds on cellular proliferation and MDR in sensitive MCF-7 and resistant MCF-7/DOX human breast cancer cells. Substitutions on the phenothiazine ring that increased hydrophobicity increased antiproliferative and anti-MDR activities. For example, -Cl and -CF3 groups increased whereas -OH groups decreased potency. Modifying the length of the alkyl bridge and the type of amino side chain also influenced potency. Compounds with increased activity against cellular proliferation and MDR possessed a four-carbon bridge rather than a three- or two-carbon bridge and a piperazinyl amine rather than a noncyclic amino group. Compounds with tertiary amines were better anti-MDR agents than those with secondary or primary amines but were equipotent antiproliferative agents. The effects of these substituents were unrelated to hydrophobicity. The structure-activity relationships suggest that an ideal phenothiazine structure for reversing MDR has a hydrophobic nucleus with a -CF3 ring substitution at position 2, connected by a four-carbon alkyl bridge to a para-methyl-substituted piperazinyl amine. We subsequently studied related compounds having certain of these properties. Substitution of a carbon for a nitrogen at position 10 of the tricyclic ring, with a double bond to the side chain (thioxanthene), further increased activity against MDR. For example, (trans)-flupenthixol, the most potent of these compounds, increased the potency of doxorubicin against MDR cells by 15-fold, as compared with its stereoisomer (cis)-flupenthixol (5-fold) or its phenothiazine homolog fluphenazine (3-fold). (cis)- and (trans)-flupenthixol were equipotent antiproliferative agents. (trans)-flupenthixol was not accumulated more than (cis)-flupenthixol in MDR cells, implying that their stereospecific anti-MDR effects were not the result of selective differences in the access of the drugs to intracellular targets. Both drugs increased the accumulation of doxorubicin in MDR cells, but not in sensitive cells, suggesting that they modulate MDR by interacting with a uniquely overexpressed cellular target in these resistant cells. The apparent lack of clinical toxicity of (trans)-flupenthixol makes it an attractive drug for further investigation.
- Published
- 1989
21. Enhanced bleomycin-induced DNA damage and cytotoxicity with calmodulin antagonists.
- Author
-
Lazo, J S, Hait, W N, Kennedy, K A, Braun, I D, and Meandzija, B
- Abstract
A wide variety of structurally different calmodulin antagonists enhanced the cytotoxicity of bleomycin A2 to leukemic L1210 cells. This potentiation occurred with nontoxic concentrations of calmodulin antagonists. The most potent blockers of L1210 calmodulin activity, melittin and mastoparan, were the most potent potentiators of bleomycin A2 cytotoxicity. Less potent agents such as pimozide, a diphenylbutylpiperidine, trifluoperazine and chlorpromazine, phenothiazines, and W-7, a naphthalene sulfonamide, required higher concentrations for potentiation of bleomycin A2-induced cytotoxicity, while homologs that lack anticalmodulin activity failed to increase the cytotoxicity seen with bleomycin A2. The potentiation of bleomycin A2 cytotoxicity was not due to an elevated cellular content of bleomycin A2 or to inhibition of bleomycin A2 inactivation. Using alkaline elution techniques, we found that pimozide increased bleomycin A2-induced DNA damage in intact L1210 cells. Pimozide did not, however, directly increase the formation of reactive species by bleomycin as measured by single or double strand breakage of covalently closed circular DNA. Thus, the potentiation of bleomycin cytotoxicity by these agents appears to be mediated by an increased damage to cellular DNA; this may be due to inhibition of DNA repair. The hypothesized calmodulin-dependent mechanism was not shared by all agents that caused breaks in DNA because no potentiation in cytotoxicity was observed when calmodulin antagonists were combined with either etoposide or X-irradiation.
- Published
- 1985
22. Elongation factor-2 kinase: immunological evidence for the existence of tissue-specific isoforms
- Author
-
Hait, W. N., Ward, M. D., Trakht, I. N., and Ryazanov, A. G.
- Published
- 1996
- Full Text
- View/download PDF
23. Structure-activity relationships of phenothiazines and related drugs for inhibition of protein kinase C.
- Author
-
Aftab, D T, Ballas, L M, Loomis, C R, and Hait, W N
- Abstract
Phenothiazines are known to inhibit the activity of protein kinase C. To identify structural features that determine inhibitory activity against the enzyme, we utilized a semiautomated assay [Anal. Biochem. 187:84-88 (1990)] to compare the potency of greater than 50 phenothiazines and related compounds. Potency was decreased by trifluoro substitution at position 2 on the phenothiazine nucleus and increased by quinoid structures on the nucleus. An alkyl bridge of at least three carbons connecting the terminal amine to the nucleus was required for activity. Primary amines and unsubstituted piperazines were the most potent amino side chains. We selected 7,8-dihydroxychlorpromazine (DHCP) (IC50 = 8.3 microM) and 2-chloro-9-(3-[1-piperazinyl]propylidene)thioxanthene (N751) (IC50 = 14 microM) for further study because of their potency and distinct structural features. Under standard (vesicle) assay conditions, DHCP was noncompetitive with respect to phosphatidylserine and a mixed-type inhibitor with respect to ATP. N751 was competitive with respect to phosphatidylserine and noncompetitive with respect to ATP. Using the mixed micelle assay, DHCP was a competitive inhibitor with respect to both phosphatidylserine and ATP. DHCP was selective for protein kinase C compared with cAMP-dependent protein kinase, calmodulin-dependent protein kinase type II, and casein kinase. N751 was more potent against protein kinase C compared with cAMP-dependent protein kinase and casein kinase but less potent against protein kinase C compared with calmodulin-dependent protein kinase type II. DHCP was analyzed for its ability to inhibit different isoenzymes of protein kinase C, and no significant isozyme selectivity was detected. These data provide important information for the rational design of more potent and selective inhibitors of protein kinase C.
- Published
- 1991
24. Pharmacological properties of fluphenazine-mustard, an irreversible calmodulin antagonist.
- Author
-
Hait, W N, Glazer, L, Kaiser, C, Cross, J, and Kennedy, K A
- Abstract
We describe an improved synthesis and properties of fluphenazine-mustard, a potent phenothiazine having an alkylating chlorethylamine chain in its structure. The drug possesses anticalmodulin activity equivalent to the parent compound, but unlike fluphenazine dihydrochloride, the mustard derivative irreversibly antagonizes the ability of calmodulin to activate cyclic nucleotide phosphodiesterase. This property is partially calcium-dependent and can be overcome by coincubation with excess fluphenazine dihydrochloride. The compound irreversibly inactivated calmodulin when incubated with intact cells and caused single-stranded breakage of DNA. Fluphenazine-mustard possesses potent antiproliferative and cytotoxic properties against malignant cell lines that are likely to be mediated through both of these actions.
- Published
- 1987
25. The effect of steroid metabolites on the hematopoietic stem cell pool.
- Author
-
Hait, W N, Gorshein, D, Besa, E C, Jepson, J H, and Gardner, F H
- Published
- 1973
26. Detection of Mutations in the Mitogen-Activated Protein Kinase Pathway in Human Melanoma
- Author
-
Alsina, J., Gorski, D. H., Germino, F. J., Shih, W., Lu, S. -E, Zhang, Z. -G, Yang, J. -M, Hait, W. N., and James Goydos
27. Non-Hodgkin's lymphoma for the nononcologist
- Author
-
Hait, W. N., primary
- Published
- 1985
- Full Text
- View/download PDF
28. Antitumor and Toxic Effects of Combination Chemotherapy With Bleomycin and a Phenothiazine Anticalmodulin Agent1
- Author
-
Hait, W. N., primary, Lazo, J. S., additional, Chen, D.-L., additional, Gallichio, V. S., additional, and Filderman, A. E., additional
- Published
- 1988
- Full Text
- View/download PDF
29. Calmodulin: a potential target for cancer chemotherapeutic agents.
- Author
-
Hait, W N, primary and Lazo, J S, additional
- Published
- 1986
- Full Text
- View/download PDF
30. Terfenadine (Seldane[R]): a new drug for restoring sensitivity to multidrug resistant cancer cells
- Author
-
Hait, W. N., Gesmonde, J. F., Murren, J. R., and Yang, J.-M.
- Published
- 1993
- Full Text
- View/download PDF
31. Effects of KS-501, KS-502 and their enantiomers on calmodulin-sensitive enzyme activity and cellular proliferation
- Author
-
Hait, W. N., Gesmonde, J., and Cheng, E.
- Published
- 1995
- Full Text
- View/download PDF
32. Activation of phospholipase C induces the expression of the multidrug resistance (MDR1) gene through the Raf-MAPK pathway.
- Author
-
Yang JM, Vassil AD, and Hait WN
- Subjects
- 3T3 Cells, Animals, Enzyme Activation, Enzyme Inhibitors pharmacology, Isoenzymes antagonists & inhibitors, Mice, Mitogen-Activated Protein Kinases antagonists & inhibitors, Phospholipase C gamma, Promoter Regions, Genetic physiology, Transfection, Type C Phospholipases antagonists & inhibitors, ATP Binding Cassette Transporter, Subfamily B, Member 1 genetics, Gene Expression physiology, Isoenzymes metabolism, Mitogen-Activated Protein Kinases metabolism, Proto-Oncogene Proteins c-raf metabolism, Type C Phospholipases metabolism
- Abstract
Resistance to multiple, unrelated cancer chemotherapeutic drugs can be mediated by P-glycoprotein, the MDR1 gene product. Numerous substances, including chemotherapeutic drugs, heavy metals, growth factors, activated oncogenes, or changes in temperature increase MDR1 gene expression. Because several of these factors regulate cellular function through the activation of phospholipase C (PLC), we postulated that PLC-mediated signaling could be central to regulating the expression of MDR1. Transfection of NIH 3T3 cells with a pMJ30-PLC-gamma 1 expression vector increased the activity of the MDR1 promoter by 2- to 10-fold. PLC-mediated activation required a region between -106 and -99 of the MDR1 promoter. Treatment of cotransfected cells with platelet-derived growth factor further enhanced the activity of the MDR1 promoter. The stimulatory effect of PLC on the MDR1 promoter was increased by cotransfection with constitutively active v-raf and was blocked by the dominant-negative mutant, c-Raf-C4. The activity of mitogen-activated protein kinase (MAPK) was also increased in PLC-gamma 1-transfected cells. Furthermore, PD-98059 and U0126, two MAPK inhibitors, blocked PLC-gamma 1-induced expression of MDR1. The results of Northern blot analysis showed that activation of PLC by heat shock and growth factors increased expression of endogenous MDR1 mRNA in human renal carcinoma cells. These effects were blocked by inhibitors of the PLC-MAPK pathway. In summary, our results indicate for the first time that activation of PLC by a variety of cellular stimuli can regulate the expression of MDR1 and that the transcriptional modulation of MDR1 expression by PLC is mediated by the Raf-MAPK pathway.
- Published
- 2001
33. The prognostic and predictive values of ECD-HER-2.
- Author
-
Hait WN
- Subjects
- Breast Neoplasms drug therapy, Breast Neoplasms pathology, Female, Humans, Neoplasm Metastasis, Predictive Value of Tests, Prognosis, Survival Analysis, Breast Neoplasms blood, Receptor, ErbB-2 blood
- Abstract
The search for a simple, sensitive test to reliably determine prognosis and predict response to therapy in patients with cancer is an important area of translational research. In this issue of Clinical Cancer Research, Hayes et al. (Clin. Cancer Res., 7: 601-604, 2001) report the results of an ancillary Cancer and Leukemia Group B protocol designed to determine whether the circulating extracellular domain of HER-2/neu (ECD-HER-2) was indicative of prognosis or predictive of response to therapy in women with metastatic breast cancer. Results were drawn from a sample of 242 patients of whom 89 had elevated values of the protein. These women had been enrolled in a variety of Cancer and Leukemia Group B protocols evaluating either the efficacy of dose in the use of megestrol acetate as second-line hormonal treatment or in patients enrolled into several chemotherapeutic protocols, many containing doxorubicin. They report that patients with pretreatment elevation of ECD-HER-2 had a worse prognosis than those who did not, but that there was no convincing correlation of elevated ECD-HER-2 with response to either endocrine or chemotherapy. Although the small number of patients and the retrospective study design allows one to draw only tentative conclusions, this report raises several important issues for the conduct of translational research and points to several new hypotheses for future testing.
- Published
- 2001
34. Disruption of the EF-2 kinase/Hsp90 protein complex: a possible mechanism to inhibit glioblastoma by geldanamycin.
- Author
-
Yang J, Yang JM, Iannone M, Shih WJ, Lin Y, and Hait WN
- Subjects
- Animals, Benzoquinones, Cell Division drug effects, Cell Survival drug effects, Elongation Factor 2 Kinase, Glioblastoma drug therapy, Glioblastoma pathology, Humans, Lactams, Macrocyclic, Mice, Mice, Nude, Rats, Rifabutin analogs & derivatives, Rifabutin pharmacology, Tumor Cells, Cultured, Antibiotics, Antineoplastic pharmacology, Calcium-Calmodulin-Dependent Protein Kinases metabolism, Glioblastoma metabolism, HSP90 Heat-Shock Proteins metabolism, Quinones pharmacology
- Abstract
Glioblastoma multiforme is the most treatment-resistant brain tumor. Elongation factor-2 (EF-2) kinase (calmodulin kinase III) is a unique protein kinase that is overexpressed in glioma cell lines and in human surgical specimens. Several mitogens activate this kinase and inhibitors block mitogen activation and produce cell death. Geldanamycin (GA) is a benzoquinone ansamycin antibiotic that disrupts Hsp90-protein interactions. Because EF-2 kinase is chaperoned by Hsp90, we investigated the effects of GA on the viability of glioma cells, the expression of EF-2 kinase protein, and the interaction between Hsp90 and EF-2 kinase. GA was a potent inhibitor of the clonogenicity of four glioma cells lines with IC(50)s ranging from 1 to 3 nM. 17-allylamino-17-demethoxygeldanamycin (17-AAG), a less toxic and less potent derivative of GA, inhibited the clonogenicity of glioma cells with IC(50) values of 13 nM in C6 cells and 35 nM in T98G cells. Treatment of cell lines for 24-48 h of GA or 17-AAG disrupted EF-2-kinase/Hsp90 interactions as measured by coimmunoprecipitation, resulting in a decreased amount of recoverable kinase in cell lysates. The ability of GA to inhibit the growth of glioma cells was abrogated by overexpressing EF-2 kinase. In addition, 17-AAG significantly inhibited the growth of a glioma xenograft in nude mice. These studies demonstrate for the first time the activity of GAs against human gliomas in vitro and in vivo and suggest that destruction of EF-2 kinase may be an important cytotoxic mechanism of this unique class of drug.
- Published
- 2001
35. State-of-the-art prostate cancer treatment and research. A report from the Cancer Institute of New Jersey.
- Author
-
DiPaola RS, Kumar P, Hait WN, and Weiss RE
- Subjects
- Adult, Aged, Antineoplastic Agents administration & dosage, Antineoplastic Agents therapeutic use, Antineoplastic Agents, Phytogenic administration & dosage, Antineoplastic Agents, Phytogenic therapeutic use, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Clinical Trials as Topic, Forecasting, Genes, bcl-2 genetics, Genes, p53 genetics, Humans, Immunotherapy, Active, Interferons administration & dosage, Interferons therapeutic use, Male, Mass Screening, Middle Aged, Mutation, Orchiectomy, Phytotherapy, Plant Extracts administration & dosage, Plant Extracts therapeutic use, Prostate-Specific Antigen blood, Prostatic Neoplasms drug therapy, Prostatic Neoplasms genetics, Prostatic Neoplasms mortality, Prostatic Neoplasms radiotherapy, Quality of Life, Research, Retinoids administration & dosage, Retinoids therapeutic use, Testosterone blood, Time Factors, Drugs, Chinese Herbal, Prostatic Neoplasms therapy
- Abstract
Prostate cancer is a devastating disease that will be diagnosed in approximately 200,000 men in 2001. New methods for screening, prevention, and treatment are being developed. In addition, novel agents for the treatment of resistant prostate cancer are being developed in clinical trials. This review summarizes the recent efforts in diet, screening, novel systemic therapies, and alternative medicine for prostate cancer.
- Published
- 2001
36. Tubulin targeting agents.
- Author
-
Hait WN, Rubin E, and Goodin S
- Subjects
- Animals, Binding Sites, Humans, Neoplasms metabolism, Antineoplastic Agents pharmacology, Neoplasms drug therapy, Tubulin drug effects
- Published
- 2001
37. Regulation of expression of the multidrug resistance protein MRP1 by p53 in human prostate cancer cells.
- Author
-
Sullivan GF, Yang JM, Vassil A, Yang J, Bash-Babula J, and Hait WN
- Subjects
- Carcinoma genetics, Cell Cycle, Clone Cells, Colonic Neoplasms genetics, Dose-Response Relationship, Drug, Doxorubicin pharmacology, Flow Cytometry, Humans, Leukotriene C4 metabolism, Male, Melanoma genetics, MutS Homolog 3 Protein, Recombinant Proteins metabolism, Transfection, Tumor Suppressor Protein p53 genetics, Vincristine pharmacology, Antineoplastic Agents pharmacology, DNA-Binding Proteins biosynthesis, Multidrug Resistance-Associated Proteins, Prostatic Neoplasms metabolism, Tumor Suppressor Protein p53 metabolism
- Abstract
The expression of several drug-resistance genes, including MRP and p53, increases with advancing stage of human prostate cancer. Altered transcription could account for the genotypic alterations associated with prostate cancer progression, and it was recently reported that the promoter of MRP1 is activated in the presence of mutant p53. To determine whether there is a relationship between p53 status and the expression of MRP1, a human, temperature-sensitive p53 mutant (tsp Val(138)) was transfected into LNCaP human prostate cancer cells. In the transfected cell line (LVCaP), the wild-type p53 produced growth arrest at the G1/S interface of the cell cycle, inhibited colony formation, and induced p21(waf1/cip1). Temperature shifting to 38 degrees C (p53 mutant) produced a time-dependent increase in expression of MRP1. This change in MRP1 expression was also seen in isogenic cell lines in which p53 was inactivated by human papilloma virus (HPV)16E6 protein or by a dominant-negative mutant. Functional assays revealed a decrease in drug accumulation and drug sensitivity associated with mutant p53 and increased MRP1 expression. These results provide the first mechanistic link between expression of MRP1 and mutation of p53 in human prostate cancer and support recent clinical associations. Furthermore, these data suggest a mechanism tying accumulation of p53 mutations to the multidrug resistance phenotype seen in this disease.
- Published
- 2000
- Full Text
- View/download PDF
38. Treatment of multidrug resistant (MDR1) murine leukemia with P-glycoprotein substrates accelerates the course of the disease.
- Author
-
Yang JM, Yang GY, Medina DJ, Vassil AD, Liao J, and Hait WN
- Subjects
- ATP Binding Cassette Transporter, Subfamily B, Member 1 genetics, Animals, Cell Membrane drug effects, Cell Membrane metabolism, Cell Movement drug effects, Disease Progression, Female, Flupenthixol pharmacology, Humans, Hydrogen-Ion Concentration, Leukemia P388 metabolism, Liver Neoplasms metabolism, Liver Neoplasms pathology, Liver Neoplasms secondary, Mechlorethamine pharmacology, Mice, Neoplasm Invasiveness, Neoplasm Transplantation, Paclitaxel pharmacokinetics, Paclitaxel pharmacology, Survival Rate, Tumor Cells, Cultured, Vincristine pharmacokinetics, Vincristine pharmacology, ATP Binding Cassette Transporter, Subfamily B, Member 1 metabolism, Drug Resistance, Neoplasm, Leukemia P388 pathology
- Abstract
The prognosis of patients with tumors expressing P-glycoprotein (P-gp), the MDR1 gene product, is generally poor. It is assumed that this is due to decreased tumor responsiveness that results from decreased drug accumulation. We observed that treatment of animals bearing MDR1-transfected leukemic cells with P-gp substrates (i.e., drugs that are transported by P-gp) significantly worsened host survival compared to treatment with vehicle or non-P-gp substrates. This effect was seen with cancer chemotherapeutic agents (paclitaxel and vincristine) and with the MDR modulator, trans-flupenthixol. To determine the mechanism(s) underlying this observation, we studied alterations in pharmacokinetics, pharmacodynamics, and metastasis. We found that the drug-induced acceleration of disease was associated with increased metastases. P-gp(+) cells treated with P-gp substrates demonstrated several pro-metastatic features, including membrane ruffling and invasion through a hepatocyte monolayer. These results suggest that the treatment of MDR tumors with P-gp substrates may produce changes in malignant behavior that could adversely affect therapeutic outcomes., (Copyright 1999 Academic Press.)
- Published
- 1999
- Full Text
- View/download PDF
39. DNA damage increases sensitivity to vinca alkaloids and decreases sensitivity to taxanes through p53-dependent repression of microtubule-associated protein 4.
- Author
-
Zhang CC, Yang JM, Bash-Babula J, White E, Murphy M, Levine AJ, and Hait WN
- Subjects
- Animals, Biopolymers, Bleomycin pharmacology, Cytarabine pharmacology, DNA, Neoplasm radiation effects, Doxorubicin pharmacology, Gene Expression Regulation, Neoplastic radiation effects, Genes, p53, Humans, Mice, Microtubule-Associated Proteins genetics, Microtubules drug effects, Neoplasm Proteins genetics, Neoplasm Proteins physiology, Rats, Transcription, Genetic drug effects, Tumor Cells, Cultured, Ultraviolet Rays, Vinblastine pharmacology, Antineoplastic Agents, Phytogenic pharmacology, Bridged-Ring Compounds pharmacology, DNA Damage, DNA, Neoplasm drug effects, Drug Resistance, Neoplasm, Gene Expression Regulation, Neoplastic drug effects, Microtubule-Associated Proteins biosynthesis, Neoplasm Proteins biosynthesis, Paclitaxel pharmacology, Taxoids, Tumor Suppressor Protein p53 physiology, Vinca Alkaloids pharmacology
- Abstract
Taxanes and Vinca alkaloids are among the most active classes of drugs in the treatment of cancer. Yet, fewer than 50% of previously untreated patients respond, and clinicians have few ways of predicting who will benefit from treatment and who will not. Mutations in p53 occur in more than half of human malignancies and may alter the sensitivity to a variety of anticancer therapies. We have shown that the transcriptional status of p53 determines the sensitivity to antimicrotubule drugs and that this is mediated through the regulation of microtubule-associated protein 4 (MAP4). Expression of MAP4 is transcriptionally repressed by wild-type p53. Increased expression of MAP4, which occurs when p53 is transcriptionally inactive, increases microtubule polymerization, paclitaxel binding, and sensitivity to paclitaxel, a drug that stabilizes polymerized microtubules. In contrast, overexpression of MAP4 decreases microtubule binding and sensitivity to Vinca alkaloids, which promotes microtubule depolymerization. To determine whether induction of endogenous wild-type p53 by DNA-damaging agents alters the expression of MAP4 and changes the sensitivity to antimicrotubule drugs, we assayed cell lines with wild-type or mutant p53 for the expression of MAP4 and drug sensitivity before and after DNA damage. UV irradiation, bleomycin, and doxorubicin increased wild-type p53 expression and decreased MAP4 expression. These changes were associated with decreased sensitivity to paclitaxel and increased sensitivity to vinblastine. These changes in drug sensitivity were no longer observed when p53 and MAP4 returned to baseline levels. Changes in drug sensitivity following DNA-damaging agents were associated with decreased binding of paclitaxel and increased binding of Vinca alkaloids. In contrast, DNA damage did not alter the sensitivity to non-microtubule-active drugs, such as 1-beta-D-arabinofuranosylcytosine and doxorubicin. Changes in drug sensitivity following DNA-damaging drugs were not observed in cells with mutant p53. These studies demonstrate that induction of wild-type p53 by DNA-damaging agents can affect the sensitivity to antimicrotubule drugs through the regulation of MAP4 expression and may have implications for the design of clinical anticancer therapies.
- Published
- 1999
40. People of the year. William N. Hait, MD, PhD. Interview by Bill Berlin.
- Author
-
Hait WN
- Subjects
- Awards and Prizes, New Jersey, Research, United States, Medical Oncology
- Published
- 1998
41. Clinical and biologic activity of an estrogenic herbal combination (PC-SPES) in prostate cancer.
- Author
-
DiPaola RS, Zhang H, Lambert GH, Meeker R, Licitra E, Rafi MM, Zhu BT, Spaulding H, Goodin S, Toledano MB, Hait WN, and Gallo MA
- Subjects
- Animals, Antineoplastic Agents, Phytogenic adverse effects, Antineoplastic Agents, Phytogenic chemistry, Breast drug effects, Drugs, Chinese Herbal adverse effects, Drugs, Chinese Herbal chemistry, Female, Humans, Libido drug effects, Male, Mice, Plant Extracts adverse effects, Plant Extracts chemistry, Prostate-Specific Antigen blood, Prostatic Neoplasms physiopathology, Saccharomyces cerevisiae drug effects, Testosterone blood, Thrombophlebitis chemically induced, Uterus drug effects, Yeasts drug effects, Antineoplastic Agents, Phytogenic pharmacology, Drugs, Chinese Herbal pharmacology, Plant Extracts pharmacology, Prostatic Neoplasms drug therapy, Receptors, Estrogen drug effects
- Abstract
Background: Herbal mixtures are popular alternatives to demonstrated therapies. PC-SPES, a commercially available combination of eight herbs, is used as a nonestrogenic treatment for cancer of the prostate. Since other herbal medicines have estrogenic effects in vitro, we tested the estrogenic activity of PC-SPES in yeast and mice and in men with prostate cancer., Methods: We measured the estrogenic activity of PC-SPES with transcriptional-activation assays in yeast and a biologic assay in mice. We assessed the clinical activity of PC-SPES in eight patients with hormone-sensitive prostate cancer by measuring serum prostate-specific antigen and testosterone concentrations during and after treatment., Results: In complementary yeast assays, a 1:200 dilution of an ethanol extract of PC-SPES had estrogenic activity similar to that of 1 nM estradiol, and in ovariectomized CD-1 mice, the herbal mixture increased uterine weights substantially. In six of six men with prostate cancer, PC-SPES decreased serum testosterone concentrations (P<0.05), and in eight of eight patients it decreased serum concentrations of prostate-specific antigen. All eight patients had breast tenderness and loss of libido, and one had venous thrombosis. High-performance liquid chromatography, gas chromatography, and mass spectrometry showed that PC-SPES contains estrogenic organic compounds that are distinct from diethylstilbestrol, estrone, and estradiol., Conclusions: PC-SPES has potent estrogenic activity. The use of this unregulated mixture of herbs may confound the results of standard or experimental therapies and may produce clinically significant adverse effects.
- Published
- 1998
- Full Text
- View/download PDF
42. The expression of drug resistance gene products during the progression of human prostate cancer.
- Author
-
Sullivan GF, Amenta PS, Villanueva JD, Alvarez CJ, Yang JM, and Hait WN
- Subjects
- ATP Binding Cassette Transporter, Subfamily B, Member 1 analysis, ATP Binding Cassette Transporter, Subfamily B, Member 1 biosynthesis, ATP-Binding Cassette Transporters biosynthesis, DNA Topoisomerases, Type II analysis, DNA Topoisomerases, Type II biosynthesis, Disease Progression, Glutathione Transferase analysis, Glutathione Transferase biosynthesis, Humans, Immunohistochemistry, Isoenzymes analysis, Isoenzymes biosynthesis, Male, Multidrug Resistance-Associated Proteins, Neoplasm Staging, Odds Ratio, Prostatic Neoplasms genetics, Prostatic Neoplasms surgery, Proto-Oncogene Proteins c-bcl-2 analysis, Proto-Oncogene Proteins c-bcl-2 biosynthesis, Tumor Suppressor Protein p53 analysis, Tumor Suppressor Protein p53 biosynthesis, ATP-Binding Cassette Transporters analysis, Drug Resistance, Multiple, Prostatic Neoplasms pathology
- Abstract
Prostate cancer progresses from a localized disease to a widely disseminated malignancy. Each step along this progression pathway involves multiple genetic alterations that impart a survival advantage to the tumor cell over its normal counterparts and may confer resistance to therapy. Because metastatic prostate cancer is one of the most therapy-resistant human neoplasms, we studied the expression of certain molecular determinants of drug resistance in the context of tumor progression. Paraffin-embedded formalin-fixed resected prostates were chosen based on Gleason grade and surgical stage. Immunohistochemistry was used to detect the expression of multidrug resistance protein (MRP), topoisomerase II alpha, p53, glutathione S-transferase pi, Bcl-2, and P-glycoprotein in these specimens. We found that all of the proteins were expressed in resected prostate except for P-glycoprotein. The expression of MRP, topoisomerase II alpha, p53, and Bcl-2 increased with the Gleason grade. In addition, the expression of MRP, topoisomerase II alpha, and p53 increased with the surgical stage. In contrast, the glutathione S-transferase pi and Bcl-2 expression decreased with the increasing surgical stage. Stage was the strongest indicator of protein expression. These results suggest that drug resistance gene products are expressed in prostate cancer at the time of surgical resection. Thus, although the emergence of the "pan-resistance" phenotype in prostate cancer may partly be a function of the selection pressure exerted by therapeutic interventions, certain determinants of chemoresistance may be caused by genetic changes accompanying tumorigenesis.
- Published
- 1998
43. The role of MAP4 expression in the sensitivity to paclitaxel and resistance to vinca alkaloids in p53 mutant cells.
- Author
-
Zhang CC, Yang JM, White E, Murphy M, Levine A, and Hait WN
- Subjects
- 3T3 Cells, Animals, Antineoplastic Agents metabolism, Cell Cycle drug effects, Cell Line, Kidney, Mice, Mice, Inbred BALB C, Microtubule-Associated Proteins drug effects, Microtubules drug effects, Microtubules metabolism, Paclitaxel metabolism, Paclitaxel pharmacology, Polymers metabolism, Rats, Receptors, Drug analysis, Tumor Suppressor Protein p53 drug effects, Vinblastine pharmacology, Vincristine pharmacology, Antineoplastic Agents pharmacology, Drug Resistance, Multiple, Microtubule-Associated Proteins biosynthesis, Mutation drug effects, Tumor Suppressor Protein p53 genetics
- Abstract
Mutations in p53 change the sensitivity to cancer chemotherapeutic drugs. Whereas many drugs, including the vinca alkaloids, often become less effective when p53 is transcriptionally inactivated, several, most notably paclitaxel, may become more effective. In studying the underlying mechanism(s), we found that increased MAP4 expression, which occurs with transcriptionally silent p53, is associated with increased sensitivity to paclitaxel and decreased sensitivity to vinca alkaloids. Using murine fibroblasts transfected with MAP4, we directly demonstrated that the changes in drug sensitivity were associated with parallel alterations in drug-induced apoptosis and cell-cycle arrest. Immunofluorescent staining of the microtubule network revealed that cells with increased MAP4 expression displayed an increase in polymerized microtubules and an increased binding of fluorsceinated paclitaxel. Since MAP4 stabilizes polymerized microtubules, overexpression of this gene provides a plausible mechanism to explain the altered sensitivity to microtubule-active drugs in the presence of mutant p53.
- Published
- 1998
- Full Text
- View/download PDF
44. Regulation of the function of P-glycoprotein by epidermal growth factor through phospholipase C.
- Author
-
Yang JM, Sullivan GF, and Hait WN
- Subjects
- Drug Resistance, Multiple, Enzyme Activation drug effects, Epidermal Growth Factor biosynthesis, ErbB Receptors drug effects, Humans, Inositol Phosphates analysis, Phosphorylation, Tumor Cells, Cultured, ATP Binding Cassette Transporter, Subfamily B, Member 1 physiology, Epidermal Growth Factor pharmacology, Type C Phospholipases metabolism
- Abstract
Many multidrug-resistant (MDR) cell lines overexpress the epidermal growth factor receptor (EGFR) as well as P-glycoprotein (P-gp). However, the role of the increased EGFR in P-gp-mediated drug resistance remains unclear. Since recent studies suggest that activation of phospholipase C (PLC) could increase the phosphorylation of P-gp, and activation of the EGFR would also activate PLC, we investigated whether the effect of epidermal growth factor (EGF) on the phosphorylation of P-gp was mediated through PLC. Treatment of the human MDR breast cancer cell line, MCF-7/AdrR, with EGF increased the phosphorylation of P-gp by 20-50%. The increased phosphorylation of P-gp was accompanied by stimulation of PLC activity, as measured by the production of inositol, 1,4,5-trisphosphate and diacylglycerol, products of phosphatidylinositol-4,5-bisphosphate hydrolysis. Treatment of MDR cells with EGF also had detectable effects on P-gp function. For example, following incubation of MCF-7/AdrR cells with ECF, we observed a consistent decrease in total vinblastine (VBL) accumulation. Kinetic analysis revealed this change to be due to an increase in membrane efflux. The latter was measured by the initial uptake velocity, which was inhibited by EGF. VBL uptake measured at 0-320 sec was inhibited by 20-40%, which was associated with a similar increase in VBL efflux. EGF had no effect on drug accumulation, uptake, or efflux in sensitive MCF-7 cells. These data indicate that EGF can modulate the phosphorylation and function of P-gp, and suggest that this effect may be initiated by the activation of PLC.
- Published
- 1997
- Full Text
- View/download PDF
45. Identification of a new class of protein kinases represented by eukaryotic elongation factor-2 kinase.
- Author
-
Ryazanov AG, Ward MD, Mendola CE, Pavur KS, Dorovkov MV, Wiedmann M, Erdjument-Bromage H, Tempst P, Parmer TG, Prostko CR, Germino FJ, and Hait WN
- Subjects
- Adenosine Triphosphate metabolism, Amino Acid Sequence, Animals, Base Sequence, Binding Sites, Caenorhabditis elegans enzymology, Calcium-Calmodulin-Dependent Protein Kinases biosynthesis, Cloning, Molecular, Conserved Sequence, DNA Primers, Dictyostelium enzymology, Elongation Factor 2 Kinase, Humans, Mice, Molecular Sequence Data, Polymerase Chain Reaction, Protein Biosynthesis, Protozoan Proteins, Rabbits, Rats, Recombinant Proteins biosynthesis, Recombinant Proteins chemistry, Recombinant Proteins metabolism, Reticulocytes enzymology, Sequence Homology, Amino Acid, Transcription, Genetic, Calcium-Calmodulin-Dependent Protein Kinases chemistry, Calcium-Calmodulin-Dependent Protein Kinases metabolism
- Abstract
The several hundred members of the eukaryotic protein kinase superfamily characterized to date share a similar catalytic domain structure, consisting of 12 conserved subdomains. Here we report the existence and wide occurrence in eukaryotes of a protein kinase with a completely different structure. We cloned and sequenced the human, mouse, rat, and Caenorhabditis elegans eukaryotic elongation factor-2 kinase (eEF-2 kinase) and found that with the exception of the ATP-binding site, they do not contain any sequence motifs characteristic of the eukaryotic protein kinase superfamily. Comparison of different eEF-2 kinase sequences reveals a highly conserved region of approximately 200 amino acids which was found to be homologous to the catalytic domain of the recently described myosin heavy chain kinase A (MHCK A) from Dictyostelium. This suggests that eEF-2 kinase and MHCK A are members of a new class of protein kinases with a novel catalytic domain structure.
- Published
- 1997
- Full Text
- View/download PDF
46. Effects of rottlerin, an inhibitor of calmodulin-dependent protein kinase III, on cellular proliferation, viability, and cell cycle distribution in malignant glioma cells.
- Author
-
Parmer TG, Ward MD, and Hait WN
- Subjects
- Animals, Cell Cycle drug effects, Cell Division drug effects, Cell Size drug effects, Cell Survival drug effects, Dose-Response Relationship, Drug, Glioblastoma ultrastructure, Humans, Microscopy, Electron, Rats, Tumor Cells, Cultured drug effects, Acetophenones pharmacology, Benzopyrans pharmacology, Calcium-Calmodulin-Dependent Protein Kinases antagonists & inhibitors, Enzyme Inhibitors pharmacology, Glioblastoma enzymology
- Abstract
Calmodulin-dependent protein kinases phosphorylate certain substrates that have been implicated in regulating cellular proliferation. For example, upon mitogenic stimulation, there is a rapid activation of calmodulin-dependent protein kinase III (CaM kinase III), which leads to the phosphorylation of elongation factor 2. Recently, our laboratory demonstrated that the activity of CaM kinase III is increased in glioma cells following exposure to mitogens and is diminished or absent in nonproliferating glial tissue. Rottlerin, a 5,7-dihydroxy-2,2-dimethyl-6-(2,4,6-trihydroxy-3-methyl-5-acetylbenzy l)-8-cinnamoyl-1,2-chromene isolated from the pericarps of Mallotus phillippinensis, has been shown to be an effective CaM kinase III inhibitor. Therefore, we evaluated the effects of rottlerin on the growth and viability of glioblastoma cell lines. Rottlerin decreased growth and induced cytotoxicity in rat (C6) and two human gliomas (T98G and U138MG) at concentrations that inhibited the activity of CaM kinase III in vitro and in vivo. Far less demonstrable effects were observed on other Ca2++/CaM-sensitive kinases. Incubation of glial cells with rottlerin produced a block at the G1-S interface and the appearance of a population of cells with a <2N complement of DNA. In addition, rottlerin induced changes in cellular morphology such as cell shrinkage, accumulation of cytoplasmic vacuoles, and packaging of cellular components within membranes. These data suggest that CaM kinase III may be an important link between the activation of CaM-dependent signaling, proliferation, and viability in malignant cells, and that inhibition of CaM kinase III may represent an interesting pharmacological target in malignant gliomas.
- Published
- 1997
47. The effects of altered cellular calmodulin expression on the growth and viability of C6 glioblastoma cells.
- Author
-
Prostko CR, Zhang C, and Hait WN
- Subjects
- Animals, Cell Division, Cell Survival, Gene Expression, Glioma metabolism, Rats, Recombinant Proteins, Transfection, Tumor Cells, Cultured, Calmodulin metabolism, Glioma pathology
- Abstract
Calmodulin (CaM) is involved in cellular processes that are vital to cell proliferation and viability. Elevated CaM content is seen in transformed cells. Anti-CaM compounds alone are cytotoxic to tumor cells and are synergistic with certain cancer chemotherapeutic agents. However, all known CaM antagonists are nonselective, complicating interpretation of these studies. To more rigorously analyze the relationship between CaM protein expression and the behavior of cancer cells, tumor-derived cell lines were engineered such that CaM concentration could be manipulated by overexpressing CaM RNA. A full-length rat CaM I cDNA was inserted into the mammalian expression vector pMTCB6+ so that either CaM mRNA (sense) or antisense RNA was expressed under the control of an inducible metallothionein promoter. Constructs were introduced into C6 glioblastoma cells by liposome-mediated transfection and colonies were selected in G418. Significantly fewer clones were recovered from transfections with antisense vectors compared to CaM sense RNA or control (empty) vector alone. This difference was attributed to the cytotoxic effects of antisense CaM RNA as opposed to differences in transfection efficiencies. CaM expression was analyzed at the RNA level by Northern blotting and CaM protein concentrations were quantitated by immunofluorescence. Clones were identified in which CaM protein could be increased or decreased following exposure to zinc ions. Changes in CaM mRNA preceded changes in CaM protein by several hours. Overexpression of CaM had no significant effects on the growth of C6 cells. However, reductions in CaM lead to decreased growth rates of C6 cells and lowered cell viability.
- Published
- 1997
48. Inhibitory effect of alkylating modulators on the function of P-glycoprotein.
- Author
-
Yang JM, Sullivan GF, Makhey DB, LaVoie EJ, and Hait WN
- Subjects
- Azides metabolism, Breast Neoplasms metabolism, Dihydropyridines metabolism, Dose-Response Relationship, Drug, Doxorubicin pharmacology, Drug Resistance, Neoplasm, Female, Flupenthixol analogs & derivatives, Flupenthixol chemistry, Fluphenazine analogs & derivatives, Fluphenazine chemistry, Humans, Tumor Cells, Cultured drug effects, ATP Binding Cassette Transporter, Subfamily B, Member 1 physiology, Breast Neoplasms drug therapy, Drug Resistance, Multiple, Flupenthixol pharmacology, Fluphenazine pharmacology, Vinblastine metabolism
- Abstract
Modulators of P-glycoprotein (P-gp) are often themselves transported out of cells, thereby limiting their effectiveness. It may be possible to develop more effective modulators of multidrug resistance by designing drugs that irreversibly block the function of P-gp. Therefore, we studied the effect of the mustard derivatives of fluphenazine (FPN) and trans-flupenthixol (FPT) on P-gp function. Both fluphenazine-mustard (FPN-M) and trans-flupenthixol-mustard (FPT-M) possessed alkylating activity, as assayed using 4-(p-nitrobenzyl) pyridine. Multidrug-resistant MCF-7/AdrR cells were incubated with FPN or FPN-M, or FPT or FPT-M for 1 h, washed for varying number of times in phosphate-buffered saline (PBS), then resuspended in medium containing [3H]vinblastine (VBL), and assayed for steady-state accumulation of the drug. Washing had far less of an effect on the ability of FPN-M and FPT-M to increase VBL accumulation compared to their parent compounds. After eight washes in excess PBS, the cells initially exposed to FPN or FPT accumulated only 30% and 50% of the initially accumulated drug, whereas the FPN-M- or FPT-M-treated cells accumulated approximately 75% and 90% of the control, respectively. FPN-M and FPT-M also increased the uptake and decreased the efflux of VBL from MDR cells despite repeated washing. We also examined the effects of these modulators on sensitivity of MDR cells to cytotoxic agents. FPN-M and FPT-M sensitized MCF-7/AdrR cells to VBL and doxorubicin to a greater extent than their parent compounds. These studies point out the potential of "irreversible" P-gp modulators to produce prolonged chemosensitization.
- Published
- 1997
49. Interaction of P-glycoprotein with protein kinase C in human multidrug resistant carcinoma cells.
- Author
-
Yang JM, Chin KV, and Hait WN
- Subjects
- Breast Neoplasms drug therapy, Carcinoma drug therapy, Humans, Phosphorylation, Precipitin Tests, Sensitivity and Specificity, Tumor Cells, Cultured, ATP Binding Cassette Transporter, Subfamily B, Member 1 metabolism, Breast Neoplasms metabolism, Carcinoma metabolism, Drug Resistance, Multiple, Isoenzymes metabolism, Protein Kinase C metabolism
- Abstract
Indirect evidence has suggested that P-glycoprotein (P-gp), the multidrug transporter, is phosphorylated by protein kinase C (PKC) and that phosphorylation modulates its transport function. To address the first premise more directly, ie., that P-gp is phosphorylated by PKC, we investigated the interaction between P-gp and PKC in sensitive and multidrug resistant MCF-7 and KB human carcinoma cell lines. We found that P-gp and PKC were coimmunoprecipitated from the multidrug-resistant cell lines MCF-7/AdrR and KB-V-1, using antibodies to either protein. The association between the two proteins was enhanced by phorbol 12-myristate 13-acetate, an analogue of diacylglycerol that induces translocation of PKC to the plasma membrane. The anti-P-gp immunoprecipitates contained PKC activity as measured by direct phosphorylation reactions. The interaction of PKC with P-gp displayed isozyme specificity: PKC-alpha, -beta, gamma, -epsilon, and -phi, but not -delta, -mu, -zeta, -lambda, were found to coimmunoprecipitate with P-gp. These studies indicate that P-gp closely interacts with PKC and serves as a substrate, and that specific isozymes of this kinase may be involved in the phosphorylation of the multidrug transporter.
- Published
- 1996
50. Trifluoperazine as a modulator of multidrug resistance in refractory breast cancer.
- Author
-
Murren JR, Durivage HJ, Buzaid AC, Reiss M, Flynn SD, Carter D, and Hait WN
- Subjects
- ATP Binding Cassette Transporter, Subfamily B, Member 1 biosynthesis, Adult, Aged, Antineoplastic Agents, Phytogenic adverse effects, Breast Neoplasms pathology, Dopamine Antagonists administration & dosage, Dopamine Antagonists therapeutic use, Drug Interactions, Drug Therapy, Combination, Female, Gene Expression Regulation, Neoplastic drug effects, Humans, Middle Aged, Trifluoperazine administration & dosage, Trifluoperazine therapeutic use, Vinblastine adverse effects, ATP Binding Cassette Transporter, Subfamily B, Member 1 antagonists & inhibitors, Antineoplastic Agents, Phytogenic therapeutic use, Breast Neoplasms drug therapy, Dopamine Antagonists pharmacology, Drug Resistance, Multiple genetics, Trifluoperazine pharmacology, Vinblastine therapeutic use
- Abstract
Overexpression of P-glycoprotein (P-gp) has been implicated as the mechanism of multidrug resistance (MDR) in a number of human cancers, including carcinoma of the breast. We conducted a clinical trial to determine whether the P-gp inhibitor, trifluoperazine, could sensitize patients with refractory breast cancer to vinblastine chemotherapy. Adult patients with histologically confirmed, refractory, advanced breast cancer were treated with vinblastine at a dose of 1.7 mg/m2 per day by continuous infusion for five consecutive days. Patients who did not respond after two cycles were subsequently treated with vinblastine plus trifluoperazine at a dose of 8 mg twice daily during the five days of chemotherapy. In patients from whom tumor samples were available, the expression of P-gp was determined by immunocytochemistry. Of 35 patients enrolled, 30 were evaluable, 2 of whom (7%) achieved a partial response to vinblastine alone. Among the 16 patients treated with vinblastine plus trifluoperazine there was one response (6%) which lasted 16 weeks. Tumor samples were available from 16 patients, and 14 (87%) were immunoreactive for P-pg. P_pg expression was detected both in the patient who responded to vinblastine plus trifluoperazine and in one of the two patients who responded to vinblastine alone. Continuous-infusion vinblastine demonstrated limited activity in this study. Furthermore, trifluoperazine did not effectively reverse established resistance to vinblastine. This failure may be related the presence of multiple mechanisms of drug resistance in the heavily pretreated population, or because ineffective concentrations of the modulator were achieved in vivo. Future studies should evaluate more effective modulators, and attempt to reverse MDR earlier in the course of treatment, before other forms of resistance can develop.
- Published
- 1996
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.