Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters










Publication year range
1.
Cancer Res ; 61(22): 8211-7, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11719452

ABSTRACT

The lack of functional p53 in many cancer cells offers a therapeutic target for treatment. Cells lacking p53 would not be anticipated to demonstrate a G(1) checkpoint and would depend on the G(2) checkpoint to permit DNA repair prior to undergoing mitosis. We hypothesized that the G(2) checkpoint abrogator could preferentially kill p53-inactive cancer cells by removing the only checkpoint that protects these cells from premature mitosis in response to DNA damage. Because Wee1 kinase is crucial in maintaining G(2) arrest through its inhibitory phosphorylation of Cdc2, we developed a high-throughput mass screening assay and used it to screen chemical library for Wee1 inhibitors. A pyridopyrimidine class of molecule, PD0166285 was identified that inhibited Wee1 at a nanomolar concentration. At the cellular level, 0.5 microM PD0166285 dramatically inhibits irradiation-induced Cdc2 phosphorylation at the Tyr-15 and Thr-14 in seven of seven cancer cell lines tested. PD0166285 abrogates irradiation-induced G(2) arrest as shown by both biochemical markers and fluorescence-activated cell sorter analysis and significantly increases mitotic cell populations. Biologically, PD0166285 acts as a radiosensitizer to sensitize cells to radiation-induced cell death with a sensitivity enhancement ratio of 1.23 as shown by standard clonogenic assay. This radiosensitizing activity is p53 dependent with a higher efficacy in p53-inactive cells. Thus, G(2) checkpoint abrogators represent a novel class of anticancer drugs that enhance cell killing of conventional cancer therapy through the induction of premature mitosis.


Subject(s)
Cell Cycle Proteins , Enzyme Inhibitors/pharmacology , G2 Phase/drug effects , Nuclear Proteins , Pyrimidines/pharmacology , Radiation-Sensitizing Agents/pharmacology , Tumor Suppressor Protein p53/physiology , Animals , CDC2 Protein Kinase/antagonists & inhibitors , CDC2 Protein Kinase/metabolism , Cell Death/drug effects , Cell Death/radiation effects , DNA Damage , G2 Phase/physiology , HT29 Cells/drug effects , HT29 Cells/radiation effects , HeLa Cells , Humans , Mice , Mutation , Phosphorylation/drug effects , Protein-Tyrosine Kinases/antagonists & inhibitors , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
2.
Neoplasia ; 3(4): 339-50, 2001.
Article in English | MEDLINE | ID: mdl-11571634

ABSTRACT

Induction of heparin-binding epidermal growth factor-like growth factor (HB-EGF) mRNA in mouse skin organ culture was blocked by two pan-ErbB receptor tyrosine kinase (RTK) inhibitors but not by genetic ablation of ErbB1, suggesting involvement of multiple ErbB species in skin physiology. Human skin, cultured normal keratinocytes, and A431 skin carcinoma cells expressed ErbB1, ErbB2, and ErbB3, but not ErbB4. Skin and A431 cells expressed more ErbB3 than did keratinocytes. Despite strong expression of ErbB2 and ErbB3, heregulin was inactive in stimulating tyrosine phosphorylation in A431 cells. In contrast, it was highly active in MDA-MB-453 breast carcinoma cells. ErbB2 displayed punctate cytoplasmic staining in A431 and keratinocytes, compared to strong cell surface staining in MDA-MB-453. In skin, ErbB2 was cytoplasmic in basal keratinocytes, assuming a cell surface pattern in the upper suprabasal layers. In contrast, ErbB1 retained a cell surface distribution in all epidermal layers. Keratinocyte proliferation in culture was found to be ErbB1-RTK-dependent, using a selective inhibitor. These results suggest that in skin keratinocytes, ErbB2 transduces ligand-dependent differentiation signals, whereas ErbB1 transduces ligand-dependent proliferation/survival signals. Intracellular sequestration of ErbB2 may contribute to the malignant phenotype of A431 cells, by allowing them to respond to ErbB1-dependent growth/survival signals, while evading ErbB2-dependent differentiation signals.


Subject(s)
ErbB Receptors/metabolism , Keratinocytes/metabolism , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Skin Neoplasms/metabolism , Animals , Blotting, Northern , Enzyme Inhibitors/pharmacology , ErbB Receptors/antagonists & inhibitors , Fluorescent Antibody Technique , Heparin/metabolism , Humans , Keratinocytes/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Quinazolines/pharmacology , RNA/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-3/antagonists & inhibitors , Receptor, ErbB-4 , Signal Transduction , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism
3.
J Biol Chem ; 276(20): 16617-23, 2001 May 18.
Article in English | MEDLINE | ID: mdl-11278443

ABSTRACT

Progression through the G1 phase of the cell cycle requires phosphorylation of the retinoblastoma gene product (pRb) by the cyclin D-dependent kinases CDK4 and CDK6, whose activity can specifically be blocked by the CDK inhibitor p16(INK4A). Misregulation of the pRb/cyclin D/p16(INK4A) pathway is one of the most common events in human cancer and has lead to the suggestion that inhibition of cyclin D-dependent kinase activity may have therapeutic value as an anticancer treatment. Through screening of a chemical library, we initially identified the [2,3-d]pyridopyrimidines as inhibitors of CDK4. Chemical modification resulted in the identification of PD 0183812 as a potent and highly selective inhibitor of both CDK4 and CDK6 kinase activity, which is competitive with ATP. Flow cytometry experiments showed that of the cell lines tested, only those expressing pRb demonstrated a G1 arrest when treated with PD 0183812. This arrest correlated in terms of incubation time and potency with a loss of pRb phosphorylation and a block in proliferation, which was reversible. These results suggest a potential use of this chemical class of compounds as therapeutic agents in the treatment of tumors with functional pRb, possessing cell cycle aberrations in other members of the pRb/cyclin D/p16(INK4A) pathway.


Subject(s)
Cell Cycle/drug effects , Cyclin-Dependent Kinases/antagonists & inhibitors , Cyclins/metabolism , Enzyme Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins , Pyridones/pharmacology , Pyrimidines/pharmacology , Animals , Breast Neoplasms , Cell Line , Cyclin D , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase 6 , DNA Replication/drug effects , DNA, Neoplasm/biosynthesis , Female , G1 Phase/drug effects , Humans , Kinetics , Phosphorylation , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Retinoblastoma Protein/metabolism , Spodoptera , Transfection , Tumor Cells, Cultured
4.
J Med Chem ; 43(24): 4606-16, 2000 Nov 30.
Article in English | MEDLINE | ID: mdl-11101352

ABSTRACT

The identification of 8-ethyl-2-phenylamino-8H-pyrido[2, 3-d]pyrimidin-7-one (1) as an inhibitor of Cdk4 led to the initiation of a program to evaluate related pyrido[2, 3-d]pyrimidin-7-ones for inhibition of cyclin-dependent kinases (Cdks). Analysis of more than 60 analogues has identified some clear SAR trends that may be exploited in the design of more potent Cdk inhibitors. The most potent Cdk4 inhibitors reported in this study inhibit Cdk4 with IC(50) = 0.004 microM ([ATP] = 25 microM). X-ray crystallographic analysis of representative compounds bound to the related kinase, Cdk2, reveals that they occupy the ATP binding site. Modest selectivity between Cdks is exhibited by some compounds, and Cdk4-selective inhibitors block pRb(+) cells in the G(1)-phase of the cell division cycle.


Subject(s)
CDC2-CDC28 Kinases , Cyclin-Dependent Kinases/antagonists & inhibitors , Enzyme Inhibitors/chemical synthesis , Proto-Oncogene Proteins , Pyrimidines/chemical synthesis , Adenosine Triphosphate/metabolism , Animals , Cell Division/drug effects , Cell Line , Crystallography, X-Ray , Cyclin-Dependent Kinase 2 , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinases/metabolism , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Insecta/cytology , Kinetics , Models, Molecular , Protein Serine-Threonine Kinases/metabolism , Pyrimidines/chemistry , Pyrimidines/pharmacology , Recombinant Proteins/antagonists & inhibitors , Structure-Activity Relationship
5.
J Med Chem ; 43(22): 4200-11, 2000 Nov 02.
Article in English | MEDLINE | ID: mdl-11063616

ABSTRACT

A series of 3-aryl-1,6-naphthyridine-2,7-diamines and related 2-ureas were prepared and evaluated as inhibitors of the FGF receptor-1 tyrosine kinase. Condensation of 4,6-diaminonicotinaldehyde and substituted phenylacetonitriles gave intermediate naphthyridine-2,7-diamines, and direct reaction of the monoanion of these (NaH/DMF) with alkyl or aryl isocyanates selectively gave the 2-ureas in varying yields (23-93%). For the preparation of more soluble 7-alkylamino-2-ureas, a number of protecting groups for the 2-amine were evaluated (phthaloyl, 4-methoxybenzyl) following selective blocking of the 7-amine (trityl), but these were not superior to the (required) 2-tert-Bu-urea group itself. Direct alkylation of the anion of the (unprotected) 7-amino group with excess 4-(3-chloropropyl)morpholine in DMF gave low (10%) yields of the desired product, but alkylation of the 7-acetamido anion, followed by mild alkaline hydrolysis, raised this to 64%. 3-Phenyl analogues were nonspecific inhibitors of isolated c-Src, FGFR, and PDGFR tyrosine kinases, whereas 3-(2,6-dichlorophenyl) analogues were most effective against c-Src and FGFR, and 3-(3,5-dimethoxyphenyl) derivatives showed high selectivity for FGFR alone. A water-soluble (7-morpholinylpropylamino) analogue retained high FGFR potency (IC(50) 31 nM) and selectivity. Pairwise comparison of the 1, 6-naphthyridines and the corresponding known pyrido[2,3-d]pyrimidine analogues showed little differences in potency or patterns of selectivity, suggesting that the 1-aza atom of the latter is not important for activity. A 7-acetamide derivative inhibited the growth of FGFR-expressing tumor cell lines and was particularly potent against HUVECs (IC(50) 4 nM). This compound was also a very potent inhibitor of HUVEC microcapillary formation (IC(50) 0.01 nM) and Matrigel invasion (IC(50) 7 nM) and showed significant in vivo antitumor effects in a highly vascularized mammary adenocarcinoma 16/c model at nontoxic doses. The compounds are worthy of further evaluation as antiangiogenesis agents.


Subject(s)
Antineoplastic Agents/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Naphthyridines/chemical synthesis , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Urea/analogs & derivatives , Urea/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Mice , Mice, Inbred Strains , Naphthyridines/chemistry , Naphthyridines/pharmacology , Receptor, Fibroblast Growth Factor, Type 1 , Structure-Activity Relationship , Tumor Cells, Cultured , Urea/chemistry , Urea/pharmacology , Xenograft Model Antitumor Assays
6.
Cancer Chemother Pharmacol ; 45(3): 231-8, 2000.
Article in English | MEDLINE | ID: mdl-10663641

ABSTRACT

PURPOSE: The involvement of the EGF receptor (EGFr) family of receptors in cancers suggests that a selective inhibitor of the tyrosine kinase activity of the EGFr family could have a therapeutic effect. PD 0169414, an anilinoquinazoline, is a potent irreversible inhibitor of the EGFr family tyrosine kinase activity with IC(50) values of 0.42 nM against the isolated EGF receptor, and 4.7 nM and 22 nM against EGF- and heregulin-mediated receptor phosphorylation in A431 and MDA-MB-453 cells, respectively. METHODS AND RESULTS: Oral administration of 260 mg/kg per day PD 0169414 for 15 days to animals bearing advanced-stage A431 epidermoid carcinoma produced a 28.2-day delay in tumor growth and resulted in three complete and three partial tumor regressions in six animals. Toxicity at this dose level was limited to <6% loss of initial body weight. Doses of 160 and 100 mg/kg per day produced tumor growth delays of 29.5 and 20.9 days and two and one complete regressions in six animals, respectively. Subcutaneous, intraperitoneal, and oral routes of administration have also shown in vivo antitumor activity of PD 0169414 in a panel of human tumor xenografts. Responsive tumor lines include A431 (human epidermoid carcinoma), H125 (NSCL carcinoma), MCF-7 and UISO-BCA1 (human breast carcinoma), and SK-OV-03 (human ovarian carcinoma). The therapeutic effect ranged from delayed tumor growth (6.4 days delayed tumor growth for 14 days of treatment) to tumor regressions (32.2 days delayed tumor growth and five partial regressions in six animals) in these model systems. CONCLUSION: PD 0169414 is a specific, irreversible inhibitor of EGFr family tyrosine kinases with significant in vivo activity against a variety of relevant human tumor xenografts.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Enzyme Inhibitors/pharmacokinetics , ErbB Receptors/drug effects , Neoplasms, Experimental/drug therapy , Quinazolines/pharmacology , Quinazolines/pharmacokinetics , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , 3T3 Cells , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Area Under Curve , Drug Administration Routes , Drug Administration Schedule , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacology , ErbB Receptors/metabolism , Humans , Infusion Pumps, Implantable , Liver/drug effects , Liver/pathology , Mice , Mice, Inbred ICR , Mice, Nude , Mice, SCID , Neoplasm Transplantation , Neoplasms, Experimental/pathology , Phosphorylation/drug effects , Quinazolines/blood , Quinazolines/therapeutic use , Receptor Protein-Tyrosine Kinases/metabolism , Transplantation, Heterologous , Treatment Outcome , Tumor Cells, Cultured
7.
Nat Med ; 5(7): 810-6, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10395327

ABSTRACT

The mitogen-activated protein kinase pathway is thought to be essential in cellular growth and differentiation. Here we report the discovery of a highly potent and selective inhibitor of the upstream kinase MEK that is orally active. Tumor growth was inhibited as much as 80% in mice with colon carcinomas of both mouse and human origin after treatment with this inhibitor. Efficacy was achieved with a wide range of doses with no signs of toxicity, and correlated with a reduction in the levels of activated mitogen-activated protein kinase in excised tumors. These data indicate that MEK inhibitors represent a promising, noncytotoxic approach to the clinical management of colon cancer.


Subject(s)
Benzamides/pharmacology , Cell Cycle/drug effects , Colonic Neoplasms/pathology , Colonic Neoplasms/physiopathology , Enzyme Inhibitors/pharmacology , Signal Transduction/drug effects , Animals , Benzamides/therapeutic use , Cadherins/analysis , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Division/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/enzymology , Enzyme Activation/drug effects , Enzyme Inhibitors/therapeutic use , Female , Hepatocyte Growth Factor/pharmacology , Humans , Male , Mice , Mice, Inbred Strains , Mice, Nude , Neoplasm Invasiveness/prevention & control , Transplantation, Heterologous , Tumor Cells, Cultured
8.
Anticancer Res ; 19(2A): 919-24, 1999.
Article in English | MEDLINE | ID: mdl-10368634

ABSTRACT

Nasopharyngeal carcinoma (NPC) is a malignancy of epithelial origin occurring with a high incidence in southern China and southeast Asia. Radiotherapy is the main treatment modality for NPC. No effective chemotherapy is available. Since prevention of EGF/EGFR binding by an EGFR specific monoclonal antibody suppressed the growth of NPC xenografts, we examined potential anti-NPC activity by a group of specific inhibitors of the EGFR family of tyrosine kinases. We found that HONE-T1 NPC cells expressed high levels of EGFR tyrosine kinase activity upon stimulation by EGF. The receptor tyrosine kinase activity was specifically inhibited by either reversible (PD158780) or irreversible (PD168393) inhibitors specific for EGFR family tyrosine kinases. This inhibition led to a dose-dependent suppression of anchorage-independent growth as determined by soft agar assays. A structural analog (PD159805) with no inhibitory activity against EGFR tyrosine kinase had no effect on HONE-T1 cell growth in agar. Furthermore, growth of HONE-T1 xenografts in SCID mice was also inhibited by treatment with PD158780 and PD 168393. This data provides an appealing application of EGFR tyrosine kinase inhibitors for the treatment of nasopharyngeal carcinomas.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , ErbB Receptors/antagonists & inhibitors , Nasopharyngeal Neoplasms/drug therapy , Pyrimidines/pharmacology , Animals , Epidermal Growth Factor/pharmacology , Humans , Mice , Mice, SCID , Nasopharyngeal Neoplasms/pathology , Neoplasm Transplantation , Phosphorylation , Transplantation, Heterologous , Tumor Cells, Cultured
9.
J Med Chem ; 42(10): 1803-15, 1999 May 20.
Article in English | MEDLINE | ID: mdl-10346932

ABSTRACT

A series of 6- and 7-acrylamide derivatives of the 4-(phenylamino)quinazoline and -pyridopyrimidine classes of epidermal growth factor receptor (EGFR) inhibitors were prepared from the corresponding amino compounds by reaction with either acryloyl chloride/base or acrylic acid/1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride. All of the 6-acrylamides, but only the parent quinazoline 7-acrylamide, were irreversible inhibitors of the isolated enzyme, confirming that the former are better-positioned, when bound to the enzyme, to react with the critical cysteine-773. Quinazoline, pyrido[3,4-d]pyrimidine, and pyrido[3,2-d]pyrimidine 6-acrylamides were all irreversible inhibitors and showed similar high potencies in the enzyme assay (likely due to titration of the available enzyme). However the pyrido[3,2-d]pyrimidine analogues were 2-6-fold less potent than the others in a cellular autophosphorylation assay for EGFR in A431 cells. The quinazolines were generally less potent overall toward inhibition of heregulin-stimulated autophosphorylation of erbB2 (in MDA-MB-453-cells), whereas the pyridopyrimidines were equipotent. Selected compounds were evaluated in A431 epidermoid and H125 non-small-cell lung cancer human tumor xenografts. The compounds showed better activity when given orally than intraperitoneally. All showed significant tumor growth inhibition (stasis) over a dose range. The poor aqueous solubility of the compounds was a drawback, requiring formulation as fine particulate emulsions.


Subject(s)
Acrylamides/chemical synthesis , Adenosine Triphosphate/metabolism , Antineoplastic Agents/chemical synthesis , Enzyme Inhibitors/chemical synthesis , ErbB Receptors/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrimidines/chemical synthesis , Quinazolines/chemical synthesis , Acrylamides/chemistry , Acrylamides/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Binding Sites , Cell Line , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , ErbB Receptors/metabolism , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Phosphorylation , Pyrimidines/chemistry , Pyrimidines/pharmacology , Quinazolines/chemistry , Quinazolines/pharmacology , Structure-Activity Relationship , Transplantation, Heterologous
10.
Mol Carcinog ; 24(1): 25-8, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10029407

ABSTRACT

Tumor metastasis is the main cause of mortality and treatment failure in cancer patients. It is a complex biological process regulated by alternations in expression of many genes. The p53 tumor suppressor gene has been shown to regulate expression of some metastasis-related genes. p53 transcriptionally activates expression of the genes encoding epidermal growth factor receptor, matrix metalloproteinase (MMP)-2, cathepsin D, and thrombospondin-1 but represses expression of the genes encoding basic fibroblast growth factor and multidrug resistance-1. Decreased expression of E-cadherin is associated with p53 alternations. Because these p53-regulatory genes either promote or inhibit tumor metastasis, the net effect of p53 expression on tumor metastasis depends upon the pattern of expression of these genes in a particular tumor. Because radiotherapy has been shown to increase tumor metastasis in both animal and human studies and because p53 is activated by radiation or DNA-damaging reagents, here we propose the working hypothesis that p53 may promote tumor metastasis upon induction by local radiotherapy or chemotherapy in some tumor types. For patients whose tumors contain wild-type p53, MMP inhibitors might be given with or before radiotherapy or chemotherapy to prevent an increase in tumor metastasis. Special caution should be taken with patients with cancers such as nasopharyngeal carcinoma in which p53 mutation is infrequent and radiotherapy is the main choice of treatment. To test our hypothesis, three studies are proposed and could serve as an initial step in understanding the complex biological process following radiation-induced p53 activation and its roles in regulation of tumor metastasis.


Subject(s)
Gene Expression Regulation, Neoplastic , Genes, p53 , Neoplasm Metastasis/genetics , Tumor Suppressor Protein p53/metabolism , Antineoplastic Agents/adverse effects , Cathepsin D/genetics , DNA Damage , DNA, Neoplasm/drug effects , DNA, Neoplasm/genetics , DNA, Neoplasm/radiation effects , ErbB Receptors/genetics , Gelatinases/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Humans , Matrix Metalloproteinase 2 , Metalloendopeptidases/genetics , Models, Biological , Radiotherapy/adverse effects , Thrombospondin 1/genetics
11.
Proc Natl Acad Sci U S A ; 95(20): 12022-7, 1998 Sep 29.
Article in English | MEDLINE | ID: mdl-9751783

ABSTRACT

A class of high-affinity inhibitors is disclosed that selectively target and irreversibly inactivate the epidermal growth factor receptor tyrosine kinase through specific, covalent modification of a cysteine residue present in the ATP binding pocket. A series of experiments employing MS, molecular modeling, site-directed mutagenesis, and 14C-labeling studies in viable cells unequivocally demonstrate that these compounds selectively bind to the catalytic domain of the epidermal growth factor receptor with a 1:1 stoichiometry and alkylate Cys-773. While the compounds are essentially nonreactive in solution, they are subject to rapid nucleophilic attack by this particular amino acid when bound in the ATP pocket. The molecular orientation and positioning of the acrylamide group in these inhibitors in relation to Cys-773 entirely support these results as determined from docking experiments in a homology-built molecular model of the ATP site. Evidence is also presented to indicate that the compounds interact in an analogous fashion with erbB2 but have no activity against the other receptor tyrosine kinases or intracellular tyrosine kinases that were tested in this study. Finally, a direct comparison between 6-acrylamido-4-anilinoquinazoline and an equally potent but reversible analog shows that the irreversible inhibitor has far superior in vivo antitumor activity in a human epidermoid carcinoma xenograft model with no overt toxicity at therapeutically active doses. The activity profile for this compound is prototypical of a generation of tyrosine kinase inhibitors with great promise for therapeutic significance in the treatment of proliferative disease.


Subject(s)
Enzyme Inhibitors/pharmacology , ErbB Receptors/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Quinazolines/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Binding Sites/genetics , Cell Line , Cysteine/chemistry , Enzyme Inhibitors/chemistry , ErbB Receptors/chemistry , ErbB Receptors/genetics , Humans , Mice , Mice, Nude , Models, Molecular , Mutagenesis, Site-Directed , Neoplasm Transplantation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Protein Conformation , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/genetics , Quinazolines/chemistry , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/genetics , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/genetics , Transplantation, Heterologous , Tumor Cells, Cultured
13.
Oncol Rep ; 4(1): 143-7, 1997.
Article in English | MEDLINE | ID: mdl-21590030

ABSTRACT

Because an enhanced therapeutic gain might be expected with co-administration of a hypoxic cell selective cytotoxin and a compound that induces hemorrhagic necrosis in tumors, the combination of CI-1010 (a potent bioreductive hypoxia selective cyto toxin) and 5,6-dimethylxanthenone-4-acetic acid (DMXAA) has been evaluated against advanced stage (>150 mg) murine colon carcinoma 26 (C26). CI-1010 and DMXAA were administered intraperitoneally over a range of toxic to ineffective doses as single agents and in combination to adult BALB/c x DBA/2 F1 hybrid mice bearing s.c. implants of C26. Both CI-1010 and DMXAA were ineffective as single agents, but regimens combining these two agents were highly active. The administration of DMXAA at 20 mg/kg/inj on days 9, 13, and 17 and CI-1010 at 65 mg/kg/inj on days 9-17 resulted in 60% of the animals tumor free on day 92 of the study. The remaining animals that were not tumor free survivors achieved a delay in tumor growth of 22.4 days. However, this treatment regimen was also considered toxic resulting in 2/10 treatment related deaths. Modification of the CI-1010 treatment schedule to intermittent delivery 24 h after each scheduled dose of DMXAA reduced treatment related toxicity while retaining efficacy. On this schedule the combination of CI-1010 (95 mg/kg/inj) given 24 h after DMXAA (20 mg/kg/inj) on days 9, 13, and 17 resulted in 60% of the treated animals tumor free on day 98 of the study. Treatment failures experienced a tumor growth delay of 11.6 days. Combination chemotherapy with CI-1010 and DMXAA was ineffective when DMXAA was administered 1 h prior to CI-1010, simultaneously with CI-1010, or 1 h after the administration of CI-1010. These results suggest that an enhanced therapeutic interaction between CI-1010 and DMXAA is achievable in vivo and that this interaction requires the development of substantial DMXAA induced tumor hypoxia prior to administration of CI-1010.

14.
FEBS Lett ; 394(2): 121-5, 1996 Sep 30.
Article in English | MEDLINE | ID: mdl-8843147

ABSTRACT

The specificity of protein kinases has been shown to be influenced by residues near the phosphoaccepting amino acid. To examine the determinants for platelet-derived growth factor receptor (PDGFR) tyrosine kinase specificity, a peptide library with three degenerate positions N-terminal to tyrosine was constructed. After reaction with PDGFR, the most abundant phosphopeptides were isolated by immunoaffinity chromatography on a column containing monoclonal anti-phosphotyrosine antibody. Further separation of bound phosphopeptides with reverse-phase HPLC led to the identification of three optimal substrates for PDGFR: Ala-Ala-Asn-Ile-Thr-Tyr-Ala-Ala-Arg-Arg-Gly, Ala-Ala-Asn-Arg-Thr-Tyr-Ala-Ala-Arg-Arg-Gly and Ala-Ala-Leu-Ile-Thr-Tyr-Ala-Ala-Arg-Arg-Gly, where underlined residues are in the degenerate positions of the peptide library. Kinetic analyses of the three individual peptides (synthesized separately) showed these peptides to be among the best reported substrates for PDGFR. Our results expand the range of amino acid residues that have been shown to serve as recognition elements for receptor tyrosine kinases.


Subject(s)
Phosphopeptides/chemistry , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Platelet-Derived Growth Factor , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Chromatography, Affinity , Chromatography, High Pressure Liquid , Kinetics , Peptide Library , Phosphopeptides/chemical synthesis , Phosphopeptides/metabolism , Phosphorylation , Sequence Analysis , Substrate Specificity
15.
Invest New Drugs ; 13(4): 295-302, 1996.
Article in English | MEDLINE | ID: mdl-8824347

ABSTRACT

PD153035 is a potent (Ki = 6 pm) and specific inhibitor of the epidermal growth factor (EGF) receptor tyrosine kinase that suppresses tyrosine phosphorylation of the EGF receptor in A431 cells at nanomolar concentrations in cell culture. We have examined the pharmacokinetics of this compound and its ability to rapidly suppress phosphorylation of the EGF receptor in A431 human epidermoid tumors grown as xenografts in immunodeficient nude mice. Following a single i.p. dose of 80 mg/kg, the drug levels in the plasma and tumor rose to 50 and 22 microM within 15 minutes. While the plasma levels of PD153035 fell below 1 microM by 3 hours, in the tumors it remained at micromolar concentrations for at least 12 hours. The tyrosine phosphorylation of the EGF receptor was rapidly suppressed by 80-90% in the tumors. However receptor phosphorylation returned to control levels after 3 hours despite the continued presence of the drug at concentrations which, based on previous in vitro results, were predicted to maintain inhibition. EGF-stimulated tyrosine kinase activity in tumor extracts was decreased and recovered in parallel with the effects of PD153035 on receptor phosphorylation though the activity had reached only about half of the control activity after three hours. These results demonstrate the potential for using small molecule inhibitors to inhibit the EGF receptor tyrosine kinase in vivo, though a fair evaluation of their potential anti-cancer activity will have to wait for solutions to problems with sustained delivery which may allow us to maintain suppression of EGF receptor phosphorylation.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/enzymology , Enzyme Inhibitors/pharmacology , ErbB Receptors/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/enzymology , Quinazolines/pharmacology , Animals , Cell Division/drug effects , Enzyme Inhibitors/pharmacokinetics , Humans , Mice , Mice, Nude , Neoplasms/metabolism , Quinazolines/pharmacokinetics , Transplantation, Heterologous , Tumor Cells, Cultured
16.
Invest New Drugs ; 14(4): 341-7, 1996.
Article in English | MEDLINE | ID: mdl-9157068

ABSTRACT

CI-980, originally synthesized as a potential folate antagonist, is a tubulin-binding mitotic inhibitor currently in pediatric phase I and adult phase II clinical trials. Because of its extensive tissue distribution in animals and its favorable activity against multidrug resistant (MDR)-cells compared with other mitotic inhibitors, such as vincristine, we examined the membrane transport properties of CI-980. CI-980 accumulated rapidly in L1210 and CHO/K1 cells, reaching intracellular levels 40- and 8-fold higher, respectively, than those in the extracellular medium. Efflux was also quite rapid, but a small fraction of drug remained associated with the cells in drug-free medium. The uptake of CI-980 was not temperature or energy dependent, nor was it saturable up to an extracellular concentration of 100 microM. Inhibitors of nucleoside transport had no effect on CI-980 uptake. A cell line deficient in the transport of reduced folate was not resistant to CI-980, nor did it exhibit reduced CI-980 uptake. A 100-fold excess of the R-enantiomer inhibited CI-980 uptake by only 50%. These results are consistent with a model of CI-980 uptake involving passive diffusion followed by significant but largely reversible binding to intracellular or membrane components.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Carbamates/pharmacokinetics , Folic Acid Antagonists/pharmacokinetics , Pyrazines/pharmacokinetics , Pyridines/pharmacokinetics , Animals , Biological Transport , CHO Cells/metabolism , Carrier Proteins/physiology , Cattle , Cricetinae , Dipyridamole/pharmacology , Drug Screening Assays, Antitumor , Humans , Leukemia L1210/metabolism , Platelet Aggregation Inhibitors/pharmacology , Stereoisomerism
17.
Invest New Drugs ; 14(4): 349-56, 1996.
Article in English | MEDLINE | ID: mdl-9157069

ABSTRACT

CI-994 [aka: acetyldinaline; PD 123654; 4-acetylamino-N-(2'aminophenyl)-benzamide] (Figure 1) is a novel antitumor agent with a unique mechanism of action. It is the acetylated metabolite of dinaline, a compound previously identified as having cytotoxic and cytostatic activity against several murine and human xenograft tumor models. CI-994 had activity against 8/8 solid tumors tested (log cell kills at the highest non-toxic dose): pancreatic ductal adenocarcinoma #02 (4.7); pancreatic adenocarcinoma #03 (3.0; 1/6 cures); colon adenocarcinoma #38 (1.6); colon adenocarcinoma #51/A (1.1); mammary adenocarcinoma #25 (1.7); mammary adenocarcinoma #17/ADR (0.5); Dunning osteogenic sarcoma (4.0); and the human prostate carcinoma LNCaP (1.2). CI-994 had the same spectrum of activity in vivo as dinaline. It also behaved similarly in schedule comparison/toxicity trials. Prolonged administration with lower drug doses was more effective than short-term therapy at higher individual doses. If doses were kept between 40 and 60 mg/kg/injection, prolonged administration (> 50 days) was tolerated with no gross toxicity. Doses > or = 90 mg/kg/injection caused lethality after 4-5 days of administration. The maximum tolerated total dose was also increased with smaller individual doses administered for prolonged intervals. Clinical Phase I trials are ongoing with this agent.


Subject(s)
Antineoplastic Agents/administration & dosage , Phenylenediamines/administration & dosage , Animals , Antineoplastic Agents/toxicity , Benzamides , Drug Administration Schedule , Drug Screening Assays, Antitumor , Female , Humans , In Vitro Techniques , Leukemia L1210/drug therapy , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, SCID , Phenylenediamines/toxicity , Tumor Cells, Cultured/drug effects
18.
Oncol Rep ; 3(6): 1153-9, 1996 Nov.
Article in English | MEDLINE | ID: mdl-21594529

ABSTRACT

CI-921, the 5-methyl-4-carboxamide analog of amsacrine, in combination with cisplatin produced a 6-fold better cell kill in vitro than expected based on additive effects. The combination of CI-921 and cisplatin was subsequently evaluated in three in vivo model systems: intraperitoneally (TP) and intravenously (IV) implanted P388 leukemia, and advanced stage subcutaneously (SC) implanted LC-12 squamous cell carcinoma. All drug treatments were administered IP on an intermittent treatment schedule which was optimal for both agents. Combination therapy was superior to therapy with the best single agent alone, CI-921, in all three model systems. Against IP implanted P388 leukemia, combination therapy produced greater than 8 logs of net tumor cell kill with 60-day survivors (cures). This level of activity was 50-fold greater (1.7 log) than that obtained with CI-921 alone. An IV implant of P388 leukemia was used in a confirmatory study to provide a more rigorous evaluation against disseminated disease. Combination therapy against IV implanted P388 leukemia produced greater than 7.7 logs of net tumor cell kill, which was 630-fold greater (2.8 logs) than that obtained with CI-921 therapy alone. Against advanced stage LC-12 (200-1000 mg tumors at initial treatment), combination therapy improved tumor cell kill by 0.6 log (4-fold) over that obtained with CI-921 therapy alone and also produced greater numbers of 120-day survivors than did single agent therapy with CI-921. The combination of carboplatin and CI-921 was also evaluated against IV implanted P388 leukemia to determine if the enhanced therapeutic effect of CI-921 and cisplatin could be extended to include CI-921 and carboplatin. Combination therapy with CI-921 and carboplatin increased net log tumor cell kill by 0.8 and 1.5 log in two separate tests (6- and 32-fold, respectively) over that obtained with CI-921 therapy alone. The data indicate that combination therapy with CI-921 and platinum containing anticancer agents may have clinical application.

19.
Cancer Res ; 54(16): 4412-8, 1994 Aug 15.
Article in English | MEDLINE | ID: mdl-8044790

ABSTRACT

[SP-4-3-(R)]-[1,1-cyclobutanedicarboxylato(2-)](2-methyl-1,4- butane-diamine-N,N')platinum (CI-973) is a cisplatin analogue that is currently in clinical trial. Preclinically, CI-973 retained activity against L1210, P388, K562, and human ovarian carcinoma sublines resistant to cisplatin in vitro. CI-973 also retained substantial activity [ratio of median life span of treated to control groups x 100% (%T/C) > 190] against cisplatin resistant L1210 and P388 sublines in vivo. Good activity (stasis or tumor burden reduction) was also obtained against five murine solid tumors including breast, colon, and sarcoma. Binary combination therapy with CI-973 and seven clinically utilized anticancer agents was evaluated against murine tumors in vivo for the ability of each combination to produce a superior therapeutic response compared to optimal single agent therapy alone at tolerated doses. The seven agents combined with CI-973 were mitomycin C, cyclophosphamide, doxorubicin, vinblastine, etoposide, ifosfamide, and methotrexate. Of the combination regimens evaluated, only the combination of CI-973 and methotrexate was therapeutically superior to single agent therapy. Against i.v. inoculated P388 leukemia, combination therapy with CI-973 at 32 mg/kg/injection and methotrexate at 43 mg/kg/injection produced 5.3 logs greater cell kill (%T/C = 284, with one cell surviving therapy) than either methotrexate therapy (%T/C = 208, with 2.5 x 10(5) cells surviving therapy) or CI-973 therapy (%T/C = 193 with 2.5 x 10(6) cells surviving therapy) alone. The combination toxicity index of 1.0 indicated additive normal host tissue toxicity with the same target organs for dose limiting toxicity. To better understand the enhanced cell kill in vivo, the combination of CI-973 and methotrexate was evaluated against P388 leukemia in vitro. Clonogenic survival studies showed that the combination of CI-973 and methotrexate was additive rather than synergistic with respect to cell killing in vitro. The lack of greater than additive cell kill in vitro suggested that the mechanism responsible for synergistic kill in vivo was not operative in vitro. The in vivo results suggest that the combination of CI-973 and methotrexate may be useful in the clinic.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carboplatin/analogs & derivatives , Animals , Carboplatin/therapeutic use , Colonic Neoplasms/drug therapy , Cyclophosphamide/administration & dosage , Doxorubicin/administration & dosage , Drug Screening Assays, Antitumor , Etoposide/administration & dosage , Ifosfamide/administration & dosage , Leukemia P388/drug therapy , Mammary Neoplasms, Animal/drug therapy , Melanoma, Experimental/drug therapy , Methotrexate/administration & dosage , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mitomycin/administration & dosage , Sarcoma, Experimental/drug therapy , Vinblastine/administration & dosage
20.
Science ; 265(5175): 1093-5, 1994 Aug 19.
Article in English | MEDLINE | ID: mdl-8066447

ABSTRACT

A small molecule called PD 153035 inhibited the epidermal growth factor (EGF) receptor tyrosine kinase with a 5-pM inhibition constant. The inhibitor was specific for the EGF receptor tyrosine kinase and inhibited other purified tyrosine kinases only at micromolar or higher concentrations. PD 153035 rapidly suppressed autophosphorylation of the EGF receptor at low nanomolar concentrations in fibroblasts or in human epidermoid carcinoma cells and selectively blocked EGF-mediated cellular processes including mitogenesis, early gene expression, and oncogenic transformation. PD 153035 demonstrates an increase in potency over that of other tyrosine kinase inhibitors of four to five orders of magnitude for inhibition of isolated EGF receptor tyrosine kinase and three to four orders of magnitude for inhibition of cellular phosphorylation.


Subject(s)
ErbB Receptors/antagonists & inhibitors , Quinazolines/antagonists & inhibitors , 3T3 Cells , Animals , Cell Transformation, Neoplastic/drug effects , Epidermal Growth Factor/pharmacology , Fibroblast Growth Factor 2/pharmacology , Gene Expression/drug effects , Humans , Kinetics , Mice , Mitosis/drug effects , Phosphorylation/drug effects , Platelet-Derived Growth Factor/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Tumor Cells, Cultured , Tyrosine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...