Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
J Urol ; 195(2): 493-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26459038

ABSTRACT

PURPOSE: Due to the high recurrence risk of nonmuscle invasive urothelial carcinoma it is crucial to distinguish patients at high risk from those with indolent disease. In this study we used a machine learning algorithm to identify the genes in patients with nonmuscle invasive urothelial carcinoma at initial presentation that were most predictive of recurrence. We used the genes in a molecular signature to predict recurrence risk within 5 years after transurethral resection of bladder tumor. MATERIALS AND METHODS: Whole genome profiling was performed on 112 frozen nonmuscle invasive urothelial carcinoma specimens obtained at first presentation on Human WG-6 BeadChips (Illumina®). A genetic programming algorithm was applied to evolve classifier mathematical models for outcome prediction. Cross-validation based resampling and gene use frequencies were used to identify the most prognostic genes, which were combined into rules used in a voting algorithm to predict the sample target class. Key genes were validated by quantitative polymerase chain reaction. RESULTS: The classifier set included 21 genes that predicted recurrence. Quantitative polymerase chain reaction was done for these genes in a subset of 100 patients. A 5-gene combined rule incorporating a voting algorithm yielded 77% sensitivity and 85% specificity to predict recurrence in the training set, and 69% and 62%, respectively, in the test set. A singular 3-gene rule was constructed that predicted recurrence with 80% sensitivity and 90% specificity in the training set, and 71% and 67%, respectively, in the test set. CONCLUSIONS: Using primary nonmuscle invasive urothelial carcinoma from initial occurrences genetic programming identified transcripts in reproducible fashion, which were predictive of recurrence. These findings could potentially impact nonmuscle invasive urothelial carcinoma management.


Subject(s)
Artificial Intelligence , Carcinoma, Transitional Cell/pathology , Gene Expression Profiling , Neoplasm Invasiveness/pathology , Urinary Bladder Neoplasms/pathology , Aged , Algorithms , Biopsy , Carcinoma, Transitional Cell/surgery , Female , Humans , Machine Learning , Male , Neoplasm Staging , Polymerase Chain Reaction , Predictive Value of Tests , Prognosis , Risk Assessment , Sensitivity and Specificity , Urinary Bladder Neoplasms/surgery
2.
Cancer ; 118(21): 5234-44, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22605513

ABSTRACT

BACKGROUND: One in 4 patients with lymph node-negative, invasive colorectal carcinoma (CRC) develops recurrent disease after undergoing curative surgery, and most die of advanced disease. Predicting which patients will develop a recurrence is a significantly growing, unmet medical need. METHODS: Archival formalin-fixed, paraffin-embedded (FFPE) primary adenocarcinoma tissues obtained at surgery were retrieved from 74 patients with CRC (15 with stage I disease and 59 with stage II disease) for Training/Test Sets. In addition, FFPE tissues were retrieved from 49 patients with stage I CRC and 215 patients with stage II colon cancer for an External Validation (EV) Set (n = 264) from 18 hospitals in 4 countries. No patients had received neoadjuvant/adjuvant therapy. Proprietary genetic programming analysis of expression profiles for 225 prespecified tumor genes was used to create a 36-month recurrence risk signature. RESULTS: Using reverse transcriptase-polymerase chain reaction, a 5-gene rule correctly classified 62 of 92 recurrent patients and 87 of 172 nonrecurrent patients in the EV Set (sensitivity, 0.67; specificity, 0.51). "High-risk" patients had a greater probability of 36-month recurrence (42%) than "low-risk" patients (26%; hazard ratio, 1.80; 95% confidence interval, 1.19-2.71; P = .007; Cox regression) independent of T-classification, the number of lymph nodes examined, histologic grade/subtype, anatomic location, age, sex, or race. The rule outperformed (P = .021) current National Comprehensive Cancer Network Guidelines (hazard ratio, 0.897). The same rule also differentiated the risk of recurrence (hazard ratio, 1.63; P = .031) in a subset of patients from the EV Set who had stage I/II colon cancer only (n = 251). CONCLUSIONS: To the authors' knowledge, the 5-gene rule (OncoDefender-CRC) is the first molecular prognostic that has been validated in both stage I CRC and stage II colon cancer. It outperforms standard clinicopathologic prognostic criteria and obviates the need to retrieve ≥12 lymph nodes for accurate prognostication. It identifies those patients most likely to develop recurrent disease within 3 years after curative surgery and, thus, those most likely to benefit from adjuvant treatment.


Subject(s)
Adenocarcinoma/genetics , Colorectal Neoplasms/genetics , Transcriptome , Adenocarcinoma/pathology , Adenocarcinoma/surgery , Adult , Aged , Aged, 80 and over , Colorectal Neoplasms/pathology , Colorectal Neoplasms/surgery , Female , Humans , Lymphatic Metastasis , Male , Middle Aged , Prognosis , Recurrence , Sensitivity and Specificity
3.
Mol Cancer Ther ; 7(7): 1880-9, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18606718

ABSTRACT

Signaling through the erbB receptor family of tyrosine kinases contributes to the proliferation, differentiation, migration, and survival of a variety of cell types. Abnormalities in members of this receptor family have been shown to play a role in oncogenesis, thus making them attractive targets for anticancer treatments. PF-00299804 is a second-generation irreversible pan-erbB receptor tyrosine kinase inhibitor currently in phase I clinical trials. PF-00299804 is believed to irreversibly inhibit erbB tyrosine kinase activity through binding at the ATP site and covalent modification of nucleophilic cysteine residues in the catalytic domains of erbB family members. Oral administration of PF-00299804 causes significant antitumor activity, including marked tumor regressions in a variety of human tumor xenograft models that express and/or overexpress erbB family members or contain the double mutation (L858R/T790M) in erbB1 (EGFR) associated with resistance to gefitinib and erlotinib. Furthermore, PF-00299804 shows exceptional distribution to human tumor xenografts and excellent pharmacokinetic properties across species.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , Quinazolinones/pharmacology , Quinazolinones/pharmacokinetics , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Xenograft Model Antitumor Assays , Amino Acid Substitution , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , ErbB Receptors/metabolism , Female , Humans , Mice , Mice, SCID , Mutation/genetics , Phosphorylation/drug effects , Species Specificity
4.
BMC Cancer ; 6: 159, 2006 Jun 16.
Article in English | MEDLINE | ID: mdl-16780590

ABSTRACT

BACKGROUND: Previous studies on bladder cancer have shown nodal involvement to be an independent indicator of prognosis and survival. This study aimed at developing an objective method for detection of nodal metastasis from molecular profiles of primary urothelial carcinoma tissues. METHODS: The study included primary bladder tumor tissues from 60 patients across different stages and 5 control tissues of normal urothelium. The entire cohort was divided into training and validation sets comprised of node positive and node negative subjects. Quantitative expression profiling was performed for a panel of 70 genes using standardized competitive RT-PCR and the expression values of the training set samples were run through an iterative machine learning process called genetic programming that employed an N-fold cross validation technique to generate classifier rules of limited complexity. These were then used in a voting algorithm to classify the validation set samples into those associated with or without nodal metastasis. RESULTS: The generated classifier rules using 70 genes demonstrated 81% accuracy on the validation set when compared to the pathological nodal status. The rules showed a strong predilection for ICAM1, MAP2K6 and KDR resulting in gene expression motifs that cumulatively suggested a pattern ICAM1 > MAP2K6 > KDR for node positive cases. Additionally, the motifs showed CDK8 to be lower relative to ICAM1, and ANXA5 to be relatively high by itself in node positive tumors. Rules generated using only ICAM1, MAP2K6 and KDR were comparably robust, with a single representative rule producing an accuracy of 90% when used by itself on the validation set, suggesting a crucial role for these genes in nodal metastasis. CONCLUSION: Our study demonstrates the use of standardized quantitative gene expression values from primary bladder tumor tissues as inputs in a genetic programming system to generate classifier rules for determining the nodal status. Our method also suggests the involvement of ICAM1, MAP2K6, KDR, CDK8 and ANXA5 in unique mathematical combinations in the progression towards nodal positivity. Further studies are needed to identify more class-specific signatures and confirm the role of these genes in the evolution of nodal metastasis in bladder cancer.


Subject(s)
Computer Simulation/statistics & numerical data , Gene Expression Profiling , Genetic Testing/statistics & numerical data , Lymphatic Metastasis/diagnosis , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/secondary , Algorithms , Cohort Studies , Gene Expression , Gene Frequency , Humans , Lymph Nodes/pathology , Lymphatic Metastasis/genetics , Predictive Value of Tests , Reproducibility of Results , Sensitivity and Specificity , Statistics as Topic
5.
Mol Cancer Ther ; 5(5): 1299-308, 2006 May.
Article in English | MEDLINE | ID: mdl-16731763

ABSTRACT

Myoblast cell cycle exit and differentiation are mediated in part by down-regulation of cyclin D1 and associated cyclin-dependent kinase (Cdk) activity. Because rhabdomyosarcoma may represent a malignant tumor composed of myoblast-like cells failing to exit the cell cycle and differentiate, we considered whether excess Cdk activity might contribute to this biology. Cyclin D-dependent Cdk4 and Cdk6 were expressed in most of a panel of six human rhabdomyosarcoma-derived cell lines. Cdk4 was expressed in 73% of alveolar and embryonal rhabdomyosarcoma tumors evaluated using a human tissue microarray. When challenged to differentiate by mitogen deprivation in vitro, mouse C2C12 myoblasts arrested in G(1) phase of the cell cycle, whereas four in the panel of rhabdomyosarcoma cell lines failed to do so. C2C12 myoblasts maintained in mitogen-rich media and exposed to a Cdk4/Cdk6 inhibitor PD 0332991 accumulated in G(1) cell cycle phase. Similar treatment of rhabdomyosarcoma cell lines caused G(1) arrest and prevented cell accumulation in vitro, and it delayed growth of rhabdomyosarcoma xenografts in vivo. Consistent with a role for Cdk4/Cdk6 activity as a regulator of myogenic differentiation, we observed that PD 0332991 exposure promoted morphologic changes and enhanced the expression of muscle-specific proteins in cultured myoblasts and in the Rh30 cell line. Our findings support the concept that pharmacologic inhibition of Cdk4/Cdk6 may represent a useful therapeutic strategy to control cell proliferation and possibly promote myogenic differentiation in rhabdomyosarcoma.


Subject(s)
Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Myoblasts, Skeletal/drug effects , Piperazines/pharmacology , Pyridines/pharmacology , Rhabdomyosarcoma/enzymology , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , G1 Phase/drug effects , Humans , Mice , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/enzymology , Piperazines/metabolism , Pyridines/metabolism , Tumor Cells, Cultured
6.
J Med Chem ; 49(4): 1475-85, 2006 Feb 23.
Article in English | MEDLINE | ID: mdl-16480284

ABSTRACT

Structure-activity relationships for inhibition of erbB1, erbB2, and erbB4 were determined for a series of alkynamide analogues of quinazoline- and pyrido[3,4-d]pyrimidine-based compounds. The compounds were prepared by coupling the appropriate 6-aminoquinazolines or 6-aminopyrido[3,4-d]pyrimidines with alkynoic acids, using EDCI.HCl in pyridine. The compounds showed pan-erbB enzyme inhibition but were on average about 10-fold more potent against erbB1 than against erbB2 and erbB4. For cellular inhibition, the nature of the alkylating side chains was an important determinant, with 5-dialkylamino-2-pentynamide type Michael acceptors providing the highest potency. This is suggested to be due to an improved ability of the amine to participate in an autocatalysis of the Michael reaction with enzyme cysteine residues. Pyrido[3,4-d]pyrimidine analogue 39 was selected for in vivo evaluation and achieved tumor regressions at 10 mg/kg in the A431 human epidermoid carcinoma and at 40 mg/kg for the SF767 human glioblastoma and the SKOV3 human ovarian carcinoma. Complete stasis was observed at 40 mg/kg in the BXPC3 human pancreatic carcinoma as well as in the H125 human non-small-cell lung carcinoma.


Subject(s)
Alkynes/chemical synthesis , Amides/chemical synthesis , Antineoplastic Agents/chemical synthesis , Pyrimidines/chemical synthesis , Quinazolines/chemical synthesis , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Alkynes/chemistry , Alkynes/pharmacology , Amides/chemistry , Amides/pharmacokinetics , Amides/pharmacology , Aniline Compounds/chemical synthesis , Aniline Compounds/chemistry , Aniline Compounds/pharmacokinetics , Aniline Compounds/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line , Dogs , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Haplorhini , Humans , Mice , Mice, Nude , Mice, SCID , Phosphorylation , Pyrimidines/chemistry , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Quinazolines/chemistry , Quinazolines/pharmacokinetics , Quinazolines/pharmacology , Rats , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Receptor, ErbB-4 , Structure-Activity Relationship , Xenograft Model Antitumor Assays
7.
Bioorg Med Chem Lett ; 15(17): 3881-5, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-15993068

ABSTRACT

The inhibition of cyclin-dependent kinase 4 (Cdk4) causes cell cycle arrest and restores a checkpoint that is absent in the majority of tumor cells. Compounds that inhibit Cdk4 selectively are targeted for treating cancer. Appropriate substitution of 2-aminoquinazolines is demonstrated to produce high levels of selectivity for Cdk4 versus closely related serine-threonine kinases.


Subject(s)
Antineoplastic Agents/chemical synthesis , Cyclin-Dependent Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Quinazolines/chemical synthesis , Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinase 4 , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Humans , Inhibitory Concentration 50 , Quinazolines/pharmacology , Structure-Activity Relationship
8.
Mol Cancer Ther ; 4(6): 938-47, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15956251

ABSTRACT

CI-1033 (N-[4-[N-(3-chloro-4-fluorophenyl)amino-7-[3-(4-morpholynyl)propoxy]quinazolin-6-yl]acrylamide, PD 0183805-mesylate salt) was identified as a potent, selective inhibitor of erbB family tyrosine kinases, which are overexpressed in a number of solid tumors and have been shown to be involved in tumor progression. Because objective response of clinical patients to erbB-targeted therapies like CI-1033 has been observed only in a subset of cancer patients that exhibit the intended molecular targets, much emphasis has been placed on the identification of biomarkers of antitumor efficacy. Vascular endothelial growth factor (VEGF) and interleukin-8 (IL-8) were considered as potential biomarkers for CI-1033 due to ease of detection in patient plasma and showed roles in angiogenesis and cancer progression and positive regulation by the erbB receptor family. In the present studies, mice bearing established xenografts (A431 epidermoid carcinoma, H125 non-small cell lung carcinoma, SF767 glioblastoma, and MDA-MB-468 mammary carcinoma) were treated with efficacious and subefficacious doses of CI-1033, and plasma levels and xenograft gene expression of VEGF and IL-8 were evaluated. Oral administration of CI-1033 to tumor-bearing mice at efficacious doses resulted in markedly decreased levels of VEGF and/or IL-8 plasma levels and tumor mRNA levels relative to vehicle-treated control mice in xenograft models that exhibited evaluable levels of these markers. In contrast, subefficacious doses of CI-1033 did not significantly affect VEGF or IL-8 levels in any of the xenograft models. These studies indicate that plasma VEGF and IL-8 may have use as biomarkers of antitumor efficacy for epidermal growth factor receptor/erbB-targeted therapies such as CI-1033 and suggest that further clinical study of these markers in cancer patients are warranted.


Subject(s)
Interleukin-8/blood , Morpholines/pharmacology , Oncogene Proteins v-erbB/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Vascular Endothelial Growth Factor A/blood , Administration, Oral , Animals , Biomarkers, Tumor/blood , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Interleukin-8/genetics , Interleukin-8/metabolism , Mice , Morpholines/administration & dosage , Morpholines/therapeutic use , Oncogene Proteins v-erbB/metabolism , Protein Kinase Inhibitors/therapeutic use , RNA, Messenger/genetics , RNA, Messenger/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
9.
J Med Chem ; 48(7): 2371-87, 2005 Apr 07.
Article in English | MEDLINE | ID: mdl-15801830

ABSTRACT

Inhibition of the cell cycle kinase, cyclin-dependent kinase-4 (Cdk4), is expected to provide an effective method for the treatment of proliferative diseases such as cancer. The pyrido[2,3-d]pyrimidin-7-one template has been identified previously as a privileged structure for the inhibition of ATP-dependent kinases, and good potency against Cdks has been reported for representative examples. Obtaining selectivity for individual Cdk enzymes, particularly Cdk4, has been challenging. Here, we report that the introduction of a methyl substituent at the C-5 position of the pyrido[2,3-d]pyrimidin-7-one template is sufficient to confer excellent selectivity for Cdk4 vs other Cdks and representative tyrosine kinases. Further optimization led to the identification of highly potent and selective inhibitors of Cdk4 that exhibit potent antiproliferative activity against human tumor cells in vitro. The most selective Cdk4 inhibitors were evaluated for antitumor activity against MDA-MB-435 human breast carcinoma xenografts in mice.


Subject(s)
Antineoplastic Agents/chemical synthesis , Cyclin-Dependent Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Pyridines/chemical synthesis , Pyrimidines/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinases/chemistry , Drug Screening Assays, Antitumor , Humans , Mice , Mice, Nude , Models, Molecular , Proto-Oncogene Proteins/chemistry , Pyridines/chemistry , Pyridines/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Stereoisomerism , Transplantation, Heterologous
10.
J Med Chem ; 48(7): 2388-406, 2005 Apr 07.
Article in English | MEDLINE | ID: mdl-15801831

ABSTRACT

A pharmacological approach to inhibition of cyclin-dependent kinases 4 and 6 (Cdk4/6) using highly selective small molecule inhibitors has the potential to provide novel cancer therapies for clinical use. Achieving high levels of selectivity for Cdk4/6, versus other ATP-dependent kinases, presents a significant challenge. The pyrido[2,3-d]pyrimidin-7-one template provides an effective platform for the inhibition of a broad cross-section of kinases, including Cdks. It is now demonstrated that the modification of pyrido[2,3-d]pyrimidin-7-ones to include a 2-aminopyridine side chain at the C2-position provides inhibitors with exquisite selectivity for Cdk4/6 in vitro. This selectivity profile is recapitulated in cells where the most selective inhibitors create a G(1) block at concentrations up to 100-fold the IC(50) for cell proliferation. On the basis of its selectivity profile and pharmacokinetic profile, compound 43 (PD 0332991) was identified as a drug candidate for the treatment of cancer.


Subject(s)
Antineoplastic Agents/chemical synthesis , Cyclin-Dependent Kinases/antagonists & inhibitors , Piperazines/chemical synthesis , Proto-Oncogene Proteins/antagonists & inhibitors , Pyridines/chemical synthesis , Pyrimidines/chemical synthesis , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Biological Availability , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase 6 , G1 Phase/drug effects , Humans , Male , Piperazines/chemistry , Piperazines/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Pyrimidines/chemistry , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Thymidine/metabolism
11.
Exp Cell Res ; 303(1): 56-67, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15572027

ABSTRACT

The erbB receptor family (EGFr, erbB-2, erbB-3, and erbB-4) consists of transmembrane glycoproteins that transduce extracellular signals to the nucleus when activated. erbB family members are widely expressed in epithelial, mesenchymal, and neuronal cells and contribute to the proliferation, differentiation, migration, and survival of these cell types. The present study evaluates the effects of erbB family signaling on cell cycle progression and the role that pRB plays in regulating this process. ErbB family RTK activity was inhibited by PD 158780 in the breast epithelial cell line MCF10A. PD 158780 (0.5 microM) inhibited EGF-stimulated and heregulin-stimulated autophosphorylation and caused a G1 cell cycle arrest within 24 h, which correlated with hypophosporylation of pRB. MCF10A cells lacking functional pRB retained the ability to arrest in G1 when treated with PD 158780. Both cell lines showed induction of p27(KIP1) protein when treated with PD 158780 and increased association of p27(KIP1) with cyclin E-CDK2. Furthermore, CDK2 kinase activity was dramatically inhibited with drug treatment. Changes in other pRB family members were noted with drug treatment, namely a decrease in p107 and an increase in p130. These findings show that the G1 arrest induced through inhibition of erbB family RTK activity does not require functional pRB.


Subject(s)
Cell Cycle/drug effects , Epithelial Cells/metabolism , G1 Phase/drug effects , Oncogenes/physiology , Retinoblastoma Protein/metabolism , Breast , Cell Cycle/physiology , Cell Cycle Proteins/metabolism , Cells, Cultured , Cyclin E/metabolism , Cyclin-Dependent Kinase Inhibitor p27 , Enzyme Activation/drug effects , Epithelial Cells/drug effects , Female , G1 Phase/physiology , Humans , Nuclear Proteins/metabolism , Oncogenes/drug effects , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Proteins/metabolism , Retinoblastoma-Like Protein p107 , Retinoblastoma-Like Protein p130 , Tumor Suppressor Proteins/metabolism
12.
Mol Cancer Ther ; 3(11): 1427-38, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15542782

ABSTRACT

PD 0332991 is a highly specific inhibitor of cyclin-dependent kinase 4 (Cdk4) (IC50, 0.011 micromol/L) and Cdk6 (IC50, 0.016 micromol/L), having no activity against a panel of 36 additional protein kinases. It is a potent antiproliferative agent against retinoblastoma (Rb)-positive tumor cells in vitro, inducing an exclusive G1 arrest, with a concomitant reduction of phospho-Ser780/Ser795 on the Rb protein. Oral administration of PD 0332991 to mice bearing the Colo-205 human colon carcinoma produces marked tumor regression. Therapeutic doses of PD 0332991 cause elimination of phospho-Rb and the proliferative marker Ki-67 in tumor tissue and down-regulation of genes under the transcriptional control of E2F. The results indicate that inhibition of Cdk4/6 alone is sufficient to cause tumor regression and a net reduction in tumor burden in some tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclin-Dependent Kinases/antagonists & inhibitors , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/antagonists & inhibitors , Pyridines/pharmacology , Xenograft Model Antitumor Assays , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 4 , Cyclin-Dependent Kinase 6 , Cyclin-Dependent Kinases/metabolism , Disease Models, Animal , Down-Regulation/drug effects , G1 Phase/drug effects , Humans , Mice , Molecular Structure , Neoplasm Transplantation/pathology , Neoplasms/metabolism , Neoplasms/pathology , Phosphorylation/drug effects , Piperazines/chemistry , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins/metabolism , Pyridines/chemistry , Retinoblastoma Protein/metabolism , Substrate Specificity
13.
Semin Oncol ; 30(5 Suppl 16): 65-78, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14613028

ABSTRACT

The erbB family of cell surface receptor proteins consists of four members, all of which play a role in the development and growth of the normal breast. The activity of this signaling pathway is normally tightly controlled, and dysregulation has been shown to play a significant role in the pathogenesis and progression of breast and other cancers. The potent transforming potential of these receptors is further enhanced by the coexpression of multiple members of this receptor family, which worsens prognosis. Therapeutic blockade of erbB-2 receptor signaling has to date been shown to be effective in only a subset of chemotherapy-resistant breast cancer patients. CI-1033 is a highly potent and selective pan-erbB inhibitor that efficiently blocks signal transduction through all four members of the erbB receptor family. In addition, it covalently binds to these receptors, irreversibility inhibiting them, and thereby provides for prolonged suppression of erbB receptor-mediated signaling. Clinically, it has been shown to have an acceptable side effect profile at potentially therapeutic doses and schedules. Biomarker studies have shown target inhibition in patients, and evidence of antitumor activity has also been observed in phase I studies. Given the broad expression pattern of the erbB family of receptors in solid tumors, and the important proliferative effect of co-expression of multiple erbB receptors, CI-1033, as an irreversible, pan-erbB inhibitor, has the potential to have an important role in the future treatment of breast and other cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Morpholines/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Breast Neoplasms/metabolism , Clinical Trials, Phase I as Topic , Drug Screening Assays, Antitumor , Humans , Ligands , Morpholines/chemistry , Neoplasms/drug therapy , Neoplasms/metabolism , Signal Transduction
14.
Exp Cell Res ; 284(1): 131-9, 2003 Mar 10.
Article in English | MEDLINE | ID: mdl-12648472

ABSTRACT

Over the last decade, drug discovery efforts have generated a myriad of compounds that inhibit the activity of the erbB family of tyrosine kinases with potencies and selectivity that have surpassed original expectations. These characteristics, along with improved pharmaceutical properties, have enabled inhibitors from this class of agents to finally realize their therapeutic potential, and indeed, some are currently producing significant clinical responses. Interestingly, those properties that are essential for a clinically active inhibitor of the erbB family are most readily attained with compounds that bind at the ATP site, and the most successful compounds have shown a distinct convergence to certain common chemical features. The reasons for this trend are beginning to be realized through the generation of an increasing array of crystalline structures for protein kinases as well as advances in molecular modeling. This has allowed a more complete understanding of the precise physical interactions that occur between erbB tyrosine kinase inhibitors and their target(s), which, in turn, has begun to shed light on the mechanism by which these molecules attain their remarkable affinity and specificity.


Subject(s)
Antineoplastic Agents/therapeutic use , Enzyme Inhibitors/therapeutic use , Neoplasms/drug therapy , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Drug Resistance, Neoplasm , Humans , Neoplasms/etiology , Peptide Fragments , Signal Transduction
15.
Semin Oncol ; 29(3 Suppl 11): 11-21, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12138393

ABSTRACT

Transmembrane receptor tyrosine kinases have been shown to play an important role in the modulation of growth factor signaling and regulation of key cellular processes. The erbB receptor family is part of the receptor tyrosine kinase superfamily and consists of four members, erbB-1, erbB-2, erbB-3, and erbB-4. A majority of solid tumors express one or more members of this receptor family, and coexpression of multiple erbB receptors leads to an enhanced transforming potential and worsened prognosis. The erbB receptor family has been shown to play an important role in both the development of the normal breast and in the pathogenesis and progression of breast cancer. Receptor overexpression has also been shown to be a negative prognostic indicator and to correlate with both tumor invasiveness and a lack of responsiveness to standard treatment. Clinically, blockade of the erbB-2 receptor has recently been shown to provide benefit in a subset of chemotherapy-resistant breast cancer patients. CI-1033 is an orally available pan-erbB receptor tyrosine kinase inhibitor that, unlike the majority of receptor inhibitors, effectively blocks signal transduction through all four members of the erbB family. In addition, it blocks the highly tumorigenic, constitutively activated variant of erbB-1, EGFRvIII, and inhibits downstream signaling through both the Ras/MAP kinase, and PI-3 kinase/AKT pathways. CI-1033 is also unique in that it is an irreversible inhibitor, thereby providing prolonged suppression of erbB receptor-mediated signaling. Preclinical data have shown CI-1033 to be efficacious against a variety of human tumors in mouse xenograft models, including breast carcinomas. In a phase I study, CI-1033 has been shown to have an acceptable side effect profile at potentially therapeutic dose levels and demonstrates evidence of target biomarker modulation. Antitumor activity has also been observed in this study, including one partial clinical response and stable disease in over 30% of patients, including one patient with heavily pretreated breast cancer. By virtue of its pan-erbB receptor inhibition and potent interruption of downstream mitogenic signaling pathways, CI-1033 may have clinical activity for solid tumors that overexpress one erbB family member, coexpress multiple members of the erbB family, or express a constitutively activated, mutated form of these receptors. Given the important role of the erbB receptor family in the pathogenesis and progression of breast cancer, an irreversible pan-erbB inhibitor like CI-1033 could have an important role to play in the future treatment of breast cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Morpholines/therapeutic use , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , ErbB Receptors/antagonists & inhibitors , Female , Humans , Mice , Morpholines/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-3/antagonists & inhibitors , Signal Transduction/drug effects
16.
EMBO J ; 21(10): 2407-17, 2002 May 15.
Article in English | MEDLINE | ID: mdl-12006493

ABSTRACT

Overexpression of ErbB-2/HER2 is associated with aggressive human malignancies, and therapeutic strategies targeting the oncoprotein are currently in different stages of clinical application. Tyrosine kinase inhibitors (TKIs) that block the nucleotide-binding site of the kinase are especially effective against tumors. Here we report an unexpected activity of TKIs: along with inhibition of tyrosine phosphorylation, they enhance ubiquitylation and accelerate endocytosis and subsequent intracellular destruction of ErbB-2 molecules. Especially potent is an irreversible TKI (CI-1033) that alkylates a cysteine specific to ErbB receptors. The degradative pathway stimulated by TKIs appears to be chaperone mediated, and is common to the heat shock protein 90 (Hsp90) antagonist geldanamycin and a stress-induced mechanism. In agreement with this conclusion, CI-1033 and geldanamycin additively inhibit tumor cell growth. Based upon a model for drug-induced degradation of ErbB-2, we propose a general strategy for selective destruction of oncoproteins by targeting their interaction with molecular chaperones.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Morpholines/pharmacology , Neoplasms/drug therapy , Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Ubiquitin/metabolism , Benzoquinones , Cell Division/drug effects , Cell Line , Endocytosis , Humans , Lactams, Macrocyclic , Neoplasms/pathology , Quinones/pharmacology , Receptor, ErbB-2/drug effects , Recombinant Proteins/metabolism , Transfection
17.
Oncogene ; 21(1): 119-27, 2002 Jan 03.
Article in English | MEDLINE | ID: mdl-11791182

ABSTRACT

Clinical responses to the HER1 (EGF receptor) inhibitors and HER2/neu/ErbB2 inhibitors correlate with high levels of receptor expression. However, a significant subset of patients with high receptor levels appear to be refractory to treatment. We have observed similar results in the 60 cell lines of the NCI Anti-Cancer Drug Screen using a panel of 11 selective HER1 inhibitors. As expected, low HER1-expressing cell lines were insensitive to HER1 inhibitors. In cell lines with high HER1 expression, low concentrations of HER1 inhibitors potently inhibit both HER1 phosphorylation and the mitogen-activated protein kinase (MAPK) pathway. However, this inhibition did not always correlate with cellular arrest. High HER1-expressing cell lines can be subdivided into two groups based on their sensitivity to HER1 inhibitors. In the sensitive group, receptor and growth inhibition was concordant and occurred at sub-micromolar concentrations of HER1 inhibitors. In the insensitive group, receptor inhibition occurred at a low concentration (< 1 microM) but concentrations that were ten times or higher were required for growth inhibition. Also, neither induction of p21 and cyclin D1 nor p53 status could explain the difference between sensitive and insensitive cells. Although EGF activated the MAPK pathway in all cell lines, only drug-sensitive cell lines responded to EGF (accelerated entry from G1 to S) and to HER1 inhibitors (G1 arrest) by changes in cell cycling. Furthermore, an EGF-dependent immortalized mammary epithelial cell line was extremely sensitive to a panel of HER1 inhibitors. We infer that independence from mitogen-mediated signaling confers insensitivity to HER1 inhibitors in a large subset of cancer cell lines.


Subject(s)
Drug Resistance, Neoplasm , Enzyme Inhibitors/pharmacology , ErbB Receptors/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Quinazolines/pharmacology , Cell Cycle/drug effects , Cell Line, Transformed/drug effects , Cell Line, Transformed/enzymology , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/metabolism , Enzyme Induction/drug effects , ErbB Receptors/physiology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MAP Kinase Signaling System/drug effects , Male , Neoplasm Proteins/physiology , Neoplasms/enzymology , Neoplasms/pathology , Pyrimidines/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/physiology , Transforming Growth Factor alpha/antagonists & inhibitors , Transforming Growth Factor alpha/physiology , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/enzymology , Tumor Suppressor Protein p53/metabolism , Tyrphostins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...