Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Nat Cancer ; 5(8): 1250-1266, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38992135

ABSTRACT

Despite tremendous progress in precision oncology, adaptive resistance mechanisms limit the long-term effectiveness of molecularly targeted agents. Here we evaluated the pharmacological profile of MTX-531 that was computationally designed to selectively target two key resistance drivers, epidermal growth factor receptor and phosphatidylinositol 3-OH kinase (PI3K). MTX-531 exhibits low-nanomolar potency against both targets with a high degree of specificity predicted by cocrystal structural analyses. MTX-531 monotherapy uniformly resulted in tumor regressions of squamous head and neck patient-derived xenograft (PDX) models. The combination of MTX-531 with mitogen-activated protein kinase kinase or KRAS-G12C inhibitors led to durable regressions of BRAF-mutant or KRAS-mutant colorectal cancer PDX models, resulting in striking increases in median survival. MTX-531 is exceptionally well tolerated in mice and uniquely does not lead to the hyperglycemia commonly seen with PI3K inhibitors. Here, we show that MTX-531 acts as a weak agonist of peroxisome proliferator-activated receptor-γ, an attribute that likely mitigates hyperglycemia induced by PI3K inhibition. This unique feature of MTX-531 confers a favorable therapeutic index not typically seen with PI3K inhibitors.


Subject(s)
Drug Resistance, Neoplasm , ErbB Receptors , Protein Kinase Inhibitors , Xenograft Model Antitumor Assays , Humans , Animals , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Mice , Drug Resistance, Neoplasm/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Cell Line, Tumor , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Colorectal Neoplasms/drug therapy , Female , Phosphatidylinositol 3-Kinases/metabolism , Head and Neck Neoplasms/drug therapy
2.
Mol Cancer Ther ; 23(1): 24-34, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-37723046

ABSTRACT

Therapeutic resistance remains a major obstacle to successful clinical management of diffuse intrinsic pontine glioma (DIPG), a high-grade pediatric tumor of the brain stem. In nearly all patients, available therapies fail to prevent progression. Innovative combinatorial therapies that penetrate the blood-brain barrier and lead to long-term control of tumor growth are desperately needed. We identified mechanisms of resistance to radiotherapy, the standard of care for DIPG. On the basis of these findings, we rationally designed a brain-penetrant small molecule, MTX-241F, that is a highly selective inhibitor of EGFR and PI3 kinase family members, including the DNA repair protein DNA-PK. Preliminary studies demonstrated that micromolar levels of this inhibitor can be achieved in murine brain tissue and that MTX-241F exhibits promising single-agent efficacy and radiosensitizing activity in patient-derived DIPG neurospheres. Its physiochemical properties include high exposure in the brain, indicating excellent brain penetrance. Because radiotherapy results in double-strand breaks that are repaired by homologous recombination (HR) and non-homologous DNA end joining (NHEJ), we have tested the combination of MTX-241F with an inhibitor of Ataxia Telangiectasia Mutated to achieve blockade of HR and NHEJ, respectively, with or without radiotherapy. When HR blockers were combined with MTX-241F and radiotherapy, synthetic lethality was observed, providing impetus to explore this combination in clinically relevant models of DIPG. Our data provide proof-of-concept evidence to support advanced development of MTX-241F for the treatment of DIPG. Future studies will be designed to inform rapid clinical translation to ultimately impact patients diagnosed with this devastating disease.


Subject(s)
Brain Stem Neoplasms , Diffuse Intrinsic Pontine Glioma , Humans , Child , Mice , Animals , Diffuse Intrinsic Pontine Glioma/drug therapy , Diffuse Intrinsic Pontine Glioma/genetics , Diffuse Intrinsic Pontine Glioma/metabolism , Neoplasm Recurrence, Local , DNA Repair , Signal Transduction , DNA/therapeutic use , Brain Stem Neoplasms/drug therapy , Brain Stem Neoplasms/genetics , Brain Stem Neoplasms/pathology
3.
Cancer Res ; 83(2): 316-331, 2023 01 18.
Article in English | MEDLINE | ID: mdl-36409827

ABSTRACT

Neurofibromin 1 (NF1) loss of function (LoF) mutations are frequent in melanoma and drive hyperactivated RAS and tumor growth. NF1LoF melanoma cells, however, do not show consistent sensitivity to individual MEK, ERK, or PI3K/mTOR inhibitors. To identify more effective therapeutic strategies for treating NF1LoF melanoma, we performed a targeted kinase inhibitor screen. A tool compound named MTX-216 was highly effective in blocking NF1LoF melanoma growth in vitro and in vivo. Single-cell analysis indicated that drug-induced cytotoxicity was linked to effective cosuppression of proliferation marker Ki-67 and ribosomal protein S6 phosphorylation. The antitumor efficacy of MTX-216 was dependent on its ability to inhibit not only PI3K, its nominal target, but also SYK. MTX-216 suppressed expression of a group of genes that regulate mitochondrial electron transport chain and are associated with poor survival in patients with NF1LoF melanoma. Furthermore, combinations of inhibitors targeting either MEK or PI3K/mTOR with an independent SYK kinase inhibitor or SYK knockdown reduced the growth of NF1LoF melanoma cells. These studies provide a path to exploit SYK dependency to selectively target NF1LoF melanoma cells. SIGNIFICANCE: A kinase inhibitor screen identifies SYK as a targetable vulnerability in melanoma cells with NF1 loss of function.


Subject(s)
Antineoplastic Agents , Melanoma , Humans , Neurofibromin 1/genetics , Syk Kinase/genetics , Syk Kinase/therapeutic use , Cell Line, Tumor , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Antineoplastic Agents/pharmacology , Mitogen-Activated Protein Kinase Kinases , Phosphatidylinositol 3-Kinases/metabolism
4.
Cancer Res ; 80(19): 4042-4043, 2020 10 01.
Article in English | MEDLINE | ID: mdl-33008803

ABSTRACT

Significant advances in tumor sequencing have led to an explosion in our knowledge of the genetic complexity of cancer. For many cancers, the selection of a targetable alteration is not readily apparent, especially when confronted with mutational variants of unknown significance. The complex clinical landscape of MEK mutations illustrates the need for improved methods to identify those patients, independent of tumor histology, who would benefit from treatment with a MAP kinase pathway inhibitor. In this issue of Cancer Research, Hanrahan and colleagues adopt an in silico platform to attempt to distinguish benign MEK mutations from those that are functional and, therefore, most likely to be therapeutically actionable.See related article by Hanrahan et al., p. 4233.


Subject(s)
Benchmarking , Neoplasms , Computer Simulation , Humans , Mitogen-Activated Protein Kinase Kinases , Mutation , Neoplasms/genetics
6.
Mol Cancer Ther ; 17(12): 2495-2506, 2018 12.
Article in English | MEDLINE | ID: mdl-30254182

ABSTRACT

The ineffectiveness of chemotherapy in patients with pancreatic cancer highlights a critical unmet need in pancreatic cancer therapy. Two commonly mutated genes in pancreatic cancer, KRAS and CDKN2A, have an incidence exceeding 90%, supporting investigation of dual targeting of MEK and CDK4/6 as a potential therapeutic strategy for this patient population. An in vitro proliferation synergy screen was conducted to evaluate response of a panel of high passage and patient-derived pancreatic cancer models to the combination of trametinib and palbociclib to inhibit MEK and CDK4/6, respectively. Two adenosquamous carcinoma models, L3.6pl and UM59, stood out for their high synergy response. In vivo studies confirmed that this combination treatment approach was highly effective in subcutaneously implanted L3.6pl and UM59 tumor-bearing animals. Both models were refractory to single-agent treatment. Reverse-phase protein array analysis of L3.6pl tumors excised from treated animals revealed strong downregulation of COX-2 expression in response to combination treatment. Expression of COX-2 under a CMV-driven promoter and shRNA knockdown of COX-2 both led to resistance to combination treatment. Our findings suggest that COX-2 may be involved in the improved therapeutic outcome seen in some pancreatic tumors that fail to respond to MEK or CDK4/6 inhibitors alone but respond favorably to their combination.


Subject(s)
Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclooxygenase 2/metabolism , Mitogen-Activated Protein Kinase Kinases/metabolism , Pancreatic Neoplasms/enzymology , Protein Kinase Inhibitors/pharmacology , Animals , Apoptosis Regulatory Proteins/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Down-Regulation/drug effects , Female , G1 Phase/drug effects , Humans , Mice, Nude , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Pancreatic Neoplasms/pathology , Piperazines/pharmacology , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyridines/pharmacology , Pyridines/therapeutic use , Pyridones/pharmacology , Pyridones/therapeutic use , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , RNA-Binding Proteins/metabolism , Retinoblastoma Protein/metabolism , Up-Regulation/drug effects
7.
Cancer Discov ; 8(5): 534-536, 2018 05.
Article in English | MEDLINE | ID: mdl-29716939

ABSTRACT

It has generally been assumed that MEK mutants function similarly to one another and respond in the same manner to targeted drugs. Gao and colleagues challenge this assumption and report that MEK1 mutants fall into three unique phenotypic classes with respect to RAF dependency. A new class of MEK1 mutants is shown here to be RAF-independent, resistant to allosteric MEK inhibitors, and yet sensitive to treatment with a new ATP-competitive MEK inhibitor. Cancer Discov; 8(5); 534-6. ©2018 AACRSee related article by Gao et al., p. 648.


Subject(s)
Protein Kinase Inhibitors , Alleles , MAP Kinase Kinase 1/genetics
8.
Clin Colon Rectal Surg ; 31(3): 199-204, 2018 May.
Article in English | MEDLINE | ID: mdl-29720906

ABSTRACT

The establishment and validation of preclinical models that faithfully recapitulate the pathogenesis and treatment response of human colorectal cancer (CRC) is critical to expedient therapeutic advances in the clinical management of this disease. Integral to the application of precision medicine for patients diagnosed with metastatic CRC is the need to understand the molecular determinants of response for a given therapy. Preclinical models of CRC have proven invaluable in answering many of our basic questions relating to the molecular aberrations that drive colorectal tumor progression. This review will address the comparative merits and limitations of the broad spectrum of in vitro and in vivo models available for study of colorectal tumors and their response to experimental therapies.

9.
Nat Commun ; 7: 13084, 2016 10 05.
Article in English | MEDLINE | ID: mdl-27703239

ABSTRACT

Glutathione S-transferase omega 1 (GSTO1) is an atypical GST isoform that is overexpressed in several cancers and has been implicated in drug resistance. Currently, no small-molecule drug targeting GSTO1 is under clinical development. Here we show that silencing of GSTO1 with siRNA significantly impairs cancer cell viability, validating GSTO1 as a potential new target in oncology. We report on the development and characterization of a series of chloroacetamide-containing potent GSTO1 inhibitors. Co-crystal structures of GSTO1 with our inhibitors demonstrate covalent binding to the active site cysteine. These potent GSTO1 inhibitors suppress cancer cell growth, enhance the cytotoxic effects of cisplatin and inhibit tumour growth in colon cancer models as single agent. Bru-seq-based transcription profiling unravelled novel roles for GSTO1 in cholesterol metabolism, oxidative and endoplasmic stress responses, cytoskeleton and cell migration. Our findings demonstrate the therapeutic utility of GSTO1 inhibitors as anticancer agents and identify the novel cellular pathways under GSTO1 regulation in colorectal cancer.


Subject(s)
Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Drug Resistance, Neoplasm , Gene Silencing , Glutathione Transferase/antagonists & inhibitors , Glutathione Transferase/genetics , Acetamides/chemistry , Antineoplastic Agents/pharmacology , Catalytic Domain , Cell Line, Tumor , Cell Movement , Cell Survival , Crystallography, X-Ray , Cysteine/chemistry , Drug Design , Endoplasmic Reticulum/metabolism , Enzyme Inhibitors/pharmacology , HCT116 Cells , High-Throughput Nucleotide Sequencing , Humans , Neoplasm Transplantation , Oxidative Stress , RNA, Small Interfering/metabolism
10.
Clin Cancer Res ; 22(2): 405-14, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26369631

ABSTRACT

PURPOSE: The emerging need for rational combination treatment approaches led us to test the concept that cotargeting MEK and CDK4/6 would prove efficacious in KRAS-mutant (KRAS(mt)) colorectal cancers, where upregulated CDK4 and hyperphosphorylated retinoblastoma (RB) typify the vast majority of tumors. EXPERIMENTAL DESIGN: Initial testing was carried out in the HCT-116 tumor model, which is known to harbor a KRAS mutation. Efficacy studies were then performed with five RB(+) patient-derived colorectal xenograft models, genomically diverse with respect to KRAS, BRAF, and PIK3CA mutational status. Tolerance, efficacy, and pharmacodynamic evaluation of target modulation were evaluated in response to daily dosing with either agent alone or concurrent coadministration. RESULTS: Synergy was observed in vitro when HCT-116 cells were treated over a broad range of doses of trametinib and palbociclib. Subsequent in vivo evaluation of this model showed a higher degree of antitumor activity resulting from the combination compared to that achievable with single-agent treatment. Testing of colorectal patient-derived xenograft (PDX) models further showed that combination of trametinib and palbociclib was well tolerated and resulted in objective responses in all KRAS(mt) models tested. Stasis was observed in a KRAS/BRAF wild-type and a BRAF(mt) model. CONCLUSIONS: Combination of trametinib and palbociclib was well tolerated and highly efficacious in all three KRAS-mutant colorectal cancer PDX models tested. Promising preclinical activity seen here supports clinical evaluation of this treatment approach to improve therapeutic outcome for patients with metastatic colorectal cancer.


Subject(s)
Colorectal Neoplasms/genetics , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 6/genetics , MAP Kinase Signaling System/genetics , Mutation/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Drug Therapy, Combination/methods , Female , HCT116 Cells , Humans , Mice , Mice, Nude , Mutation/drug effects , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins B-raf/genetics
11.
Sci Signal ; 8(397): ra98, 2015 Oct 06.
Article in English | MEDLINE | ID: mdl-26443705

ABSTRACT

Yes-associated protein 1 (YAP1) is a transcriptional coactivator in the Hippo signaling pathway. Increased YAP1 activity promotes the growth of tumors, including that of colorectal cancer (CRC). Verteporfin, a drug that enhances phototherapy to treat neovascular macular degeneration, is an inhibitor of YAP1. We found that verteporfin inhibited tumor growth independently of its effects on YAP1 or the related protein TAZ in genetically or chemically induced mouse models of CRC, in patient-derived xenografts, and in enteroid models of CRC. Instead, verteporfin exhibited in vivo selectivity for killing tumor cells in part by impairing the global clearance of high-molecular weight oligomerized proteins, particularly p62 (a sequestrome involved in autophagy) and STAT3 (signal transducer and activator of transcription 3; a transcription factor). Verteporfin inhibited cytokine-induced STAT3 activity and cell proliferation and reduced the viability of cultured CRC cells. Although verteporfin accumulated to a greater extent in normal cells than in tumor cells in vivo, experiments with cultured cells indicated that the normal cells efficiently cleared verteporfin-induced protein oligomers through autophagic and proteasomal pathways. Culturing CRC cells under hypoxic or nutrient-deprived conditions (modeling a typical CRC microenvironment) impaired the clearance of protein oligomers and resulted in cell death, whereas culturing cells under normoxic or glucose-replete conditions protected cell viability and proliferation in the presence of verteporfin. Furthermore, verteporfin suppressed the proliferation of other cancer cell lines even in the absence of YAP1, suggesting that verteporfin may be effective against multiple types of solid cancers.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adenocarcinoma/drug therapy , Adenoma/drug therapy , Antineoplastic Agents/pharmacology , Colonic Neoplasms/drug therapy , Neoplasm Proteins/drug effects , Phosphoproteins/antagonists & inhibitors , Porphyrins/pharmacology , Acyltransferases , Adaptor Proteins, Signal Transducing/physiology , Adenocarcinoma/pathology , Adenoma/pathology , Adenomatous Polyposis Coli/drug therapy , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/pathology , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cell Division/drug effects , Cell Line, Tumor , Colonic Neoplasms/chemically induced , Colonic Neoplasms/pathology , Genes, APC , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Weight , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/physiology , Phosphoproteins/physiology , Phosphorylation , Proteasome Endopeptidase Complex/drug effects , Protein Multimerization/drug effects , Protein Processing, Post-Translational , STAT3 Transcription Factor/antagonists & inhibitors , Transcription Factors/antagonists & inhibitors , Transcription, Genetic/drug effects , Verteporfin , Xenograft Model Antitumor Assays , YAP-Signaling Proteins
12.
Cancer Res ; 75(21): 4582-92, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26359451

ABSTRACT

Survival rates associated with pancreatic cancer remain dismal despite advancements in detection and experimental treatment strategies. Genetically engineered mouse models of pancreatic tumorigenesis have gained considerable attention based on their ability to recapitulate key clinical features of human disease including chemotherapeutic resistance and fibrosis. However, it is unclear if transgenic systems exemplified by the Kras(G12D)/Trp53(R172H)/Pdx-1-Cre (KPC) mouse model recapitulate the functional heterogeneity of human pancreatic tumors harboring distinct cells with tumorigenic properties. To facilitate tracking of heterogeneous tumor cell populations, we incorporated a luciferase-based tag into the genetic background of the KPC mouse model. We isolated pancreatic cancer cells from multiple independent tumor lines and found that roughly 1 out of 87 cells exhibited tumorigenic capability. Notably, this frequency is significantly higher than reported for human pancreatic adenocarcinomas. Cancer stem cell (CSC) markers, including CD133, CD24, Sca-1, and functional Aldefluor activity, were unable to discriminate tumorigenic from nontumorigenic cells in syngeneic transplants. Furthermore, three-dimensional spheroid cultures originating from KPC tumors did not enrich for cells with stem-like characteristics and were not significantly more tumorigenic than cells cultured as monolayers. Additionally, we did not observe significant differences in response to gemcitabine or salinomycin in several isolated subpopulations. Taken together, these studies show that the hierarchical organization of CSCs in human disease is not recapitulated in a commonly used mouse model of pancreatic cancer and therefore provide a new view of the phenotypic and functional heterogeneity of tumor cells.


Subject(s)
Neoplasms, Experimental/pathology , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Suppressor Protein p53/genetics , AC133 Antigen , Animals , Antigens, CD/metabolism , Antigens, Ly/metabolism , Antimetabolites, Antineoplastic/pharmacology , Biomarkers, Tumor/genetics , CD24 Antigen/metabolism , Cell Transformation, Neoplastic , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Disease Models, Animal , Glycoproteins/metabolism , Luciferases/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms, Experimental/genetics , Pancreas/pathology , Pancreatic Neoplasms/genetics , Peptides/metabolism , Phenotype , Pyrans/pharmacology , Spheroids, Cellular , Staining and Labeling , Tumor Cells, Cultured , Gemcitabine
13.
Am J Physiol Gastrointest Liver Physiol ; 307(7): G700-10, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25104499

ABSTRACT

High levels of cholecystokinin (CCK) can stimulate pancreatic adaptive growth in which mature acinar cells divide, leading to enhanced pancreatic mass with parallel increases in protein, DNA, RNA, and digestive enzyme content. Prolonged release of CCK can be induced by feeding trypsin inhibitor (TI) to disrupt normal feedback control. This leads to exocrine growth in a CCK-dependent manner. The extracellular signal-related kinase (ERK) pathway regulates many proliferative processes in various tissues and disease models. The aim of this study was to evaluate the role of ERK signaling in pancreatic adaptive growth using the MEK inhibitors PD-0325901 and trametinib (GSK-1120212). It was determined that PD-0325901 given two times daily by gavage or mixed into powdered chow was an effective and specific inhibitor of ERK signaling in vivo. TI-containing chow led to a robust increase in pancreatic mass, protein, DNA, and RNA content. This pancreatic adaptive growth was blocked in mice fed chow containing the MEK inhibitors. PD-0325901 blocked TI-induced ERK-regulated early response genes, cell-cycle proteins, and mitogenesis by acinar cells. It was determined that ERK signaling is necessary for the initiation of pancreatic adaptive growth but not necessary to maintain it. PD-0325901 blocked adaptive growth when given before cell-cycle initiation but not after mitogenesis had been established. Furthermore, GSK-1120212, a chemically distinct inhibitor of the ERK pathway that is now approved for clinical use, inhibited growth similar to PD-0325901. These data demonstrate that the ERK pathway is required for CCK-stimulated pancreatic adaptive growth.


Subject(s)
Cell Proliferation , Cholecystokinin/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System , Pancreas/enzymology , Animals , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , DNA Replication , Early Growth Response Transcription Factors/metabolism , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Male , Mice, Inbred ICR , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Pancreas/drug effects , Pancreas/growth & development , Protein Kinase Inhibitors/pharmacology , RNA/biosynthesis , Time Factors , Trypsin Inhibitors/pharmacology
14.
Gastroenterology ; 146(3): 822-834.e7, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24315826

ABSTRACT

BACKGROUND & AIMS: Kras signaling via mitogen-activated protein kinase (MAPK) is highly up-regulated in pancreatic cancer cells. We investigated whether MAPK signaling is required for the initiation and maintenance of pancreatic carcinogenesis in mice. METHODS: We studied the formation and maintenance of pancreatic intraepithelial neoplasia (PanINs) in p48Cre; TetO-KrasG12D; Rosa26(rtTa-IRES-EGFP) (iKras*) mice and LSL-KrasG12D mice bred with p48Cre mice (KC). Mice were given oral PD325901, a small-molecule inhibitor of MEK1 and MEK2 (factors in the MAPK signaling pathway), along with injections of cerulein to induce pancreatitis. Other mice were given PD325901 only after PanINs developed. Pancreatic tissues were collected and evaluated using histologic, immunohistochemical, immunofluorescence, and electron microscopy analyses. Acinar cells were isolated from the tissues and the effects of MEK1 and 2 inhibitors were assessed. RESULTS: PD325901 prevented PanIN formation, but not pancreatitis, in iKras* and KC mice. In iKras* or KC mice given PD325901 at 5 weeks after PanINs developed, PanINs regressed and acinar tissue regenerated. The regression occurred through differentiation of the PanIN cells to acini, accompanied by re-expression of the acinar transcription factor Mist1. CONCLUSIONS: In iKras* and KC mice, MAPK signaling is required for the initiation and maintenance of pancreatic cancer precursor lesions. MAPK signaling promotes formation of PanINs by enabling dedifferentiation of acinar cells into duct-like cells that are susceptible to transformation.


Subject(s)
Acinar Cells/pathology , Carcinoma in Situ/physiopathology , Cell Dedifferentiation/physiology , Mitogen-Activated Protein Kinase Kinases/physiology , Pancreatic Neoplasms/physiopathology , Signal Transduction/physiology , Acinar Cells/physiology , Animals , Carcinoma in Situ/pathology , Disease Models, Animal , Female , MAP Kinase Kinase 1/physiology , MAP Kinase Kinase 2/physiology , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Mutant Strains , Pancreatic Neoplasms/pathology , Pancreatitis/physiopathology , Up-Regulation/physiology
15.
Clin Cancer Res ; 17(14): 4751-60, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21527563

ABSTRACT

PURPOSE: Currently, radiologic response of brain tumors is assessed according to the Macdonald criteria 10 weeks from the start of therapy. There exists a critical need to identify nonresponding patients early in the course of their therapy for consideration of alternative treatment strategies. Our study assessed the effectiveness of the parametric response map (PRM) imaging biomarker to provide for an earlier measure of patient survival prediction. EXPERIMENTAL DESIGN: Forty-five high-grade glioma patients received concurrent chemoradiation. Quantitative MRI including apparent diffusion coefficient (ADC) and relative cerebral blood volume (rCBV) maps were acquired pretreatment and 3 weeks midtreatment on a prospective institutional-approved study. PRM, a voxel-by-voxel image analysis method, was evaluated as an early prognostic biomarker of overall survival. Clinical and conventional MR parameters were also evaluated. RESULTS: Multivariate analysis showed that PRM(ADC+) in combination with PRM(rCBV-) obtained at week 3 had a stronger correlation to 1-year and overall survival rates than any baseline clinical or treatment response imaging metric. The composite biomarker identified three distinct patient groups, nonresponders [median survival (MS) of 5.5 months, 95% CI: 4.4-6.6 months], partial responders (MS of 16 months, 95% CI: 8.6-23.4 months), and responders (MS has not yet been reached). CONCLUSIONS: Inclusion of PRM(ADC+) and PRM(rCBV-) into a single imaging biomarker metric provided early identification of patients resistant to standard chemoradiation. In comparison to the current standard of assessment of response at 10 weeks (Macdonald criteria), the composite PRM biomarker potentially provides a useful opportunity for clinicians to identify patients who may benefit from alternative treatment strategies.


Subject(s)
Biomarkers , Brain Neoplasms/diagnosis , Glioma/diagnosis , Magnetic Resonance Imaging , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/mortality , Glioma/diagnostic imaging , Glioma/mortality , Humans , Kaplan-Meier Estimate , Middle Aged , Models, Statistical , Neoplasm Staging , Prognosis , Prospective Studies , Radiography
16.
Clin Cancer Res ; 14(12): 3651-6, 2008 Jun 15.
Article in English | MEDLINE | ID: mdl-18559577

ABSTRACT

Among mammalian mitogen-activated protein kinase (MAPK) signaling cascades, the extracellular signal-related kinase (ERK) pathway has received the most attention in the oncology drug discovery arena. By virtue of its central role in promoting proliferation, survival, and metastasis, this pathway directly affects both the formation and progression of human tumors. The identification of non-ATP-competitive inhibitors of the MAPK kinase MAPK/ERK kinase (MEK) resulted in the first demonstration that the ERK pathway could be effectively shut down in a highly selective fashion. Subsequent discovery of the oncogenic nature of B-raf kinase led to the escalation of drug discovery efforts revolving around MEK and RAF. The emergence of multiple drug candidates targeting these downstream kinases provides us with the means for validating the importance of the RAS-RAF-MEK-ERK signaling cascade in human tumors. This article highlights the lessons learned in the clinical evaluation of MAPK pathway inhibitors as anticancer agents and the complexities surrounding optimization of their therapeutic potential in light of the challenges posed by genetic heterogeneity within patient populations.


Subject(s)
Drug Delivery Systems/trends , MAP Kinase Signaling System/drug effects , Neoplasms/therapy , ras Proteins/physiology , Benzamides/pharmacology , Benzimidazoles/pharmacology , Diphenylamine/analogs & derivatives , Diphenylamine/pharmacology , Drug Design , Humans , MAP Kinase Signaling System/physiology , Models, Biological , Protein Kinase Inhibitors/pharmacology , Substrate Specificity/drug effects
17.
J Med Chem ; 50(21): 5090-102, 2007 Oct 18.
Article in English | MEDLINE | ID: mdl-17880056

ABSTRACT

A new series of MEK1 inhibitors, the 4-anilino-5-carboxamido-2-pyridones, were designed and synthesized using a combination of medicinal chemistry, computational chemistry, and structural elucidation. The effect of variation in the carboxamide side chain, substitution on the pyridone nitrogen, and replacement of the 4'-iodide were all investigated. This study afforded several compounds which were either equipotent or more potent than the clinical candidate CI-1040 (1) in an isolated enzyme assay, as well as murine colon carcinoma (C26) cells, as measured by suppression of phosphorylated ERK substrate. Most notably, pyridone 27 was found to be more potent than 1 in vitro and produced a 100% response rate at a lower dose than 1, when tested for in vivo efficacy in animals bearing C26 tumors.


Subject(s)
Amides/chemical synthesis , Aniline Compounds/chemical synthesis , Antineoplastic Agents/chemical synthesis , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Pyridones/chemical synthesis , Amides/chemistry , Amides/pharmacology , Aniline Compounds/chemistry , Aniline Compounds/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Kinase 1/chemistry , MAP Kinase Kinase 2/chemistry , Male , Mice , Models, Molecular , Neoplasm Transplantation , Phosphorylation , Pyridones/chemistry , Pyridones/pharmacology , Rats , Structure-Activity Relationship
19.
Nature ; 441(7092): 457-62, 2006 May 25.
Article in English | MEDLINE | ID: mdl-16724058

ABSTRACT

The emergence of tumour-specific, molecularly targeted agents signifies a paradigm shift in cancer therapy, with less reliance on drugs that non-discriminately kill tumour and host cells. Although the diversity of targets giving rise to this new generation of anticancer drugs has expanded, many challenges persist in the design of effective treatment regimens. The complex interplay of signal-transduction pathways further complicates the customization of cancer treatments to target single mechanisms. However, despite uncertainty over precise or dominant mechanisms of action, especially for compounds targeting multiple gene products, emerging agents are producing significant therapeutic advances against a broad range of human cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Humans , Neoplasms/enzymology , Neoplasms/genetics , Phosphotransferases/antagonists & inhibitors , Phosphotransferases/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/therapeutic use , Substrate Specificity
20.
Neoplasia ; 8(1): 1-8, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16533420

ABSTRACT

The MEK-ERK growth signaling pathway is important in human hepatocellular carcinoma (HCC). To evaluate the targeting of this pathway in HCC, we characterized a novel, orally-active MEK inhibitor, PD184161, using human HCC cells (HepG2, Hep3B, PLC, and SKHep) and in vivo human tumor xenografts. PD184161 inhibited MEK activity (IC50 = 10-100 nM) in a time- and concentration-dependent manner more effectively than PD098059 or U0126. PD184161 inhibited cell proliferation and induced apoptosis at concentrations of > or = 1.0 microM in a time- and concentration-dependent manner. In vivo, tumor xenograft P-ERK levels were significantly reduced 3 to 12 hours after an oral dose of PD184161 (P < .05). Contrarily, tumor xenograft P-ERK levels following long-term (24 days) daily dosing of PD184161 were refractory to this signaling effect. PD184161 significantly suppressed tumor engraftment and initial growth (P < .0001); however, established tumors were not significantly affected. In conclusion, PD184161 has antitumor effects in HCC in vitro and in vivo that appear to correlate with suppression of MEK activity. These studies demonstrate that PD184161 is unable to suppress MEK activity in HCC xenografts in the long term. Thus, we speculate that the degree of success of MEK targeted treatment in HCC and other cancers may, in part, depend on the discovery of mechanisms governing MEK inhibitor signaling resistance.


Subject(s)
Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/enzymology , MAP Kinase Kinase Kinases/metabolism , Animals , Butadienes/pharmacology , Cell Line, Tumor , Cell Proliferation , Flavonoids/pharmacology , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Nitriles/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL