Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Cancer Immunol Res ; 7(9): 1457-1471, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31331945

ABSTRACT

The success of targeted or immune therapies is often hampered by the emergence of resistance and/or clinical benefit in only a subset of patients. We hypothesized that combining targeted therapy with immune modulation would show enhanced antitumor responses. Here, we explored the combination potential of erdafitinib, a fibroblast growth factor receptor (FGFR) inhibitor under clinical development, with PD-1 blockade in an autochthonous FGFR2K660N/p53mut lung cancer mouse model. Erdafitinib monotherapy treatment resulted in substantial tumor control but no significant survival benefit. Although anti-PD-1 alone was ineffective, the erdafitinib and anti-PD-1 combination induced significant tumor regression and improved survival. For both erdafitinib monotherapy and combination treatments, tumor control was accompanied by tumor-intrinsic, FGFR pathway inhibition, increased T-cell infiltration, decreased regulatory T cells, and downregulation of PD-L1 expression on tumor cells. These effects were not observed in a KRASG12C-mutant genetically engineered mouse model, which is insensitive to FGFR inhibition, indicating that the immune changes mediated by erdafitinib may be initiated as a consequence of tumor cell killing. A decreased fraction of tumor-associated macrophages also occurred but only in combination-treated tumors. Treatment with erdafitinib decreased T-cell receptor (TCR) clonality, reflecting a broadening of the TCR repertoire induced by tumor cell death, whereas combination with anti-PD-1 led to increased TCR clonality, suggesting a more focused antitumor T-cell response. Our results showed that the combination of erdafitinib and anti-PD-1 drives expansion of T-cell clones and immunologic changes in the tumor microenvironment to support enhanced antitumor immunity and survival.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Immunity/drug effects , Neoplasms/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Receptors, Fibroblast Growth Factor/antagonists & inhibitors , Animals , Biomarkers , Cell Line, Tumor , Disease Models, Animal , Drug Synergism , Humans , Immunophenotyping , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Mice, Transgenic , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Prognosis , Programmed Cell Death 1 Receptor/genetics , Pyrazoles/pharmacology , Quinoxalines/pharmacology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism , Signal Transduction/drug effects , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Treatment Outcome , Tumor Microenvironment
2.
Clin Cancer Res ; 23(19): 5869-5880, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28679778

ABSTRACT

Purpose: To determine whether combination therapy with NHS-muIL12 and the anti-programmed death ligand 1 (PD-L1) antibody avelumab can enhance antitumor efficacy in preclinical models relative to monotherapies.Experimental Design: BALB/c mice bearing orthotopic EMT-6 mammary tumors and µMt- mice bearing subcutaneous MC38 tumors were treated with NHS-muIL12, avelumab, or combination therapy; tumor growth and survival were assessed. Tumor recurrence following remission and rechallenge was evaluated in EMT-6 tumor-bearing mice. Immune cell populations within spleen and tumors were evaluated by FACS and IHC. Immune gene expression in tumor tissue was profiled by NanoString® assay and plasma cytokine levels were determined by multiplex cytokine assay. The frequency of tumor antigen-reactive IFNγ-producing CD8+ T cells was evaluated by ELISpot assay.Results: NHS-muIL12 and avelumab combination therapy enhanced antitumor efficacy relative to either monotherapy in both tumor models. Most EMT-6 tumor-bearing mice treated with combination therapy had complete tumor regression. Combination therapy also induced the generation of tumor-specific immune memory, as demonstrated by protection against tumor rechallenge and induction of effector and memory T cells. Combination therapy enhanced cytotoxic NK and CD8+ T-cell proliferation and T-bet expression, whereas NHS-muIL12 monotherapy induced CD8+ T-cell infiltration into the tumor. Combination therapy also enhanced plasma cytokine levels and stimulated expression of a greater number of innate and adaptive immune genes compared with either monotherapy.Conclusions: These data indicate that combination therapy with NHS-muIL12 and avelumab increased antitumor efficacy in preclinical models, and suggest that combining NHS-IL12 and avelumab may be a promising approach to treating patients with solid tumors. Clin Cancer Res; 23(19); 5869-80. ©2017 AACR.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Breast Neoplasms/drug therapy , Immunoglobulin G/administration & dosage , Immunotherapy , Interleukin-12/immunology , Recombinant Fusion Proteins/administration & dosage , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/pathology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Combined Modality Therapy , Female , Humans , Immunoglobulin G/immunology , Interleukin-12/administration & dosage , Interleukin-12/genetics , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Mice , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Recombinant Fusion Proteins/immunology
3.
Cancer Immunol Res ; 4(12): 1038-1048, 2016 12.
Article in English | MEDLINE | ID: mdl-27856426

ABSTRACT

PD-L1 immunohistochemical staining does not always predict whether a cancer will respond to treatment with PD-1 inhibitors. We sought to characterize immune cell infiltrates and the expression of T-cell inhibitor markers in PD-L1-positive and PD-L1-negative malignant pleural mesothelioma samples. We developed a method for immune cell phenotyping using flow cytometry on solid tumors that have been dissociated into single-cell suspensions and applied this technique to analyze 43 resected malignant pleural mesothelioma specimens. Compared with PD-L1-negative tumors, PD-L1-positive tumors had significantly more infiltrating CD45+ immune cells, a significantly higher proportion of infiltrating CD3+ T cells, and a significantly higher percentage of CD3+ cells displaying the activated HLA-DR+/CD38+ phenotype. PD-L1-positive tumors also had a significantly higher proportion of proliferating CD8+ T cells, a higher fraction of FOXP3+/CD4+ Tregs, and increased expression of PD-1 and TIM-3 on CD4+ and CD8+ T cells. Double-positive PD-1+/TIM-3+ CD8+ T cells were more commonly found on PD-L1-positive tumors. Compared with epithelioid tumors, sarcomatoid and biphasic mesothelioma samples were significantly more likely to be PD-L1 positive and showed more infiltration with CD3+ T cells and PD-1+/TIM-3+ CD8+ T cells. Immunologic phenotypes in mesothelioma differ based on PD-L1 status and histologic subtype. Successful incorporation of comprehensive immune profiling by flow cytometry into prospective clinical trials could refine our ability to predict which patients will respond to specific immune checkpoint blockade strategies. Cancer Immunol Res; 4(12); 1038-48. ©2016 AACR.


Subject(s)
B7-H1 Antigen/immunology , Mesothelioma/immunology , Pleural Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Female , Humans , Male
4.
JCI Insight ; 1(14): e89014, 2016 09 08.
Article in English | MEDLINE | ID: mdl-27699239

ABSTRACT

BACKGROUND. Immune checkpoint blockade improves survival in a subset of patients with non-small-cell lung cancer (NSCLC), but robust biomarkers that predict response to PD-1 pathway inhibitors are lacking. Furthermore, our understanding of the diversity of the NSCLC tumor immune microenvironment remains limited. METHODS. We performed comprehensive flow cytometric immunoprofiling on both tumor and immune cells from 51 NSCLCs and integrated this analysis with clinical and histopathologic characteristics, next-generation sequencing, mRNA expression, and PD-L1 immunohistochemistry (IHC). RESULTS. Cytometric profiling identified an immunologically "hot" cluster with abundant CD8+ T cells expressing high levels of PD-1 and TIM-3 and an immunologically "cold" cluster with lower relative abundance of CD8+ T cells and expression of inhibitory markers. The "hot" cluster was highly enriched for expression of genes associated with T cell trafficking and cytotoxic function and high PD-L1 expression by IHC. There was no correlation between immunophenotype and KRAS or EGFR mutation, or patient smoking history, but we did observe an enrichment of squamous subtype and tumors with higher mutation burden in the "hot" cluster. Additionally, approximately 20% of cases had high B cell infiltrates with a subset producing IL-10. CONCLUSIONS. Our results support the use of immune-based metrics to study response and resistance to immunotherapy in lung cancer. FUNDING. The Robert A. and Renée E. Belfer Family Foundation, Expect Miracles Foundation, Starr Cancer Consortium, Stand Up to Cancer Foundation, Conquer Cancer Foundation, International Association for the Study of Lung Cancer, National Cancer Institute (R01 CA205150), and the Damon Runyon Cancer Research Foundation.


Subject(s)
Carcinoma, Non-Small-Cell Lung/classification , Immunophenotyping , Lung Neoplasms/classification , B7-H1 Antigen/immunology , Biomarkers, Tumor/immunology , CD8-Positive T-Lymphocytes/immunology , Humans , Immunohistochemistry , Lung , Mutation
5.
Sci Rep ; 6: 31745, 2016 08 19.
Article in English | MEDLINE | ID: mdl-27539742

ABSTRACT

With the emergence of checkpoint blockade and other immunotherapeutic drugs, and the growing adoption of smaller, more flexible adaptive clinical trial designs, there is an unmet need to develop diagnostics that can rapidly immunophenotype patient tumors. The ability to longitudinally profile the tumor immune infiltrate in response to immunotherapy also presents a window of opportunity to illuminate mechanisms of resistance. We have developed a fine needle aspirate biopsy (FNA) platform to perform immune profiling on thoracic malignancies. Matching peripheral blood, bulk resected tumor, and FNA were analyzed from 13 mesothelioma patients. FNA samples yielded greater numbers of viable cells when compared to core needle biopsies. Cell numbers were adequate to perform flow cytometric analyses on T cell lineage, T cell activation and inhibitory receptor expression, and myeloid immunosuppressive checkpoint markers. FNA samples were representative of the tumor as a whole as assessed by head-to-head comparison to single cell suspensions of dissociated whole tumor. Parallel analysis of matched patient blood enabled us to establish quality assurance criteria to determine the accuracy of FNA procedures to sample tumor tissue. FNA biopsies provide a diagnostic to rapidly phenotype the tumor immune microenvironment that may be of great relevance to clinical trials.


Subject(s)
Flow Cytometry , Immune Tolerance , Mesothelioma , T-Lymphocytes , Thoracic Neoplasms , Tumor Microenvironment/immunology , Biopsy, Fine-Needle , Female , Humans , Male , Mesothelioma/diagnosis , Mesothelioma/immunology , Mesothelioma/pathology , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Thoracic Neoplasms/diagnosis , Thoracic Neoplasms/immunology , Thoracic Neoplasms/pathology
6.
Clin Cancer Res ; 22(4): 915-22, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26459174

ABSTRACT

PURPOSE: Tumor genotyping is a powerful tool for guiding non-small cell lung cancer (NSCLC) care; however, comprehensive tumor genotyping can be logistically cumbersome. To facilitate genotyping, we developed a next-generation sequencing (NGS) assay using a desktop sequencer to detect actionable mutations and rearrangements in cell-free plasma DNA (cfDNA). EXPERIMENTAL DESIGN: An NGS panel was developed targeting 11 driver oncogenes found in NSCLC. Targeted NGS was performed using a novel methodology that maximizes on-target reads, and minimizes artifact, and was validated on DNA dilutions derived from cell lines. Plasma NGS was then blindly performed on 48 patients with advanced, progressive NSCLC and a known tumor genotype, and explored in two patients with incomplete tumor genotyping. RESULTS: NGS could identify mutations present in DNA dilutions at ≥ 0.4% allelic frequency with 100% sensitivity/specificity. Plasma NGS detected a broad range of driver and resistance mutations, including ALK, ROS1, and RET rearrangements, HER2 insertions, and MET amplification, with 100% specificity. Sensitivity was 77% across 62 known driver and resistance mutations from the 48 cases; in 29 cases with common EGFR and KRAS mutations, sensitivity was similar to droplet digital PCR. In two cases with incomplete tumor genotyping, plasma NGS rapidly identified a novel EGFR exon 19 deletion and a missed case of MET amplification. CONCLUSIONS: Blinded to tumor genotype, this plasma NGS approach detected a broad range of targetable genomic alterations in NSCLC with no false positives including complex mutations like rearrangements and unexpected resistance mutations such as EGFR C797S. Through use of widely available vacutainers and a desktop sequencing platform, this assay has the potential to be implemented broadly for patient care and translational research.


Subject(s)
Carcinoma, Non-Small-Cell Lung/blood , DNA, Neoplasm/blood , Lung Neoplasms/blood , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/secondary , Cell Line, Tumor , DNA Mutational Analysis , DNA, Neoplasm/genetics , Female , High-Throughput Nucleotide Sequencing , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging
7.
J Med Chem ; 57(21): 8817-26, 2014 Nov 13.
Article in English | MEDLINE | ID: mdl-25313996

ABSTRACT

An affinity-based mass spectrometry screening technology was used to identify novel binders to both nonphosphorylated and phosphorylated ERK2. Screening of inactive ERK2 identified a pyrrolidine analogue 1 that bound to both nonphosphorylated and phosphorylated ERK2 and inhibited ERK2 kinase activity. Chemical optimization identified compound 4 as a novel, potent, and highly selective ERK1,2 inhibitor which not only demonstrated inhibition of phosphorylation of ERK substrate p90RSK but also demonstrated inhibition of ERK1,2 phosphorylation on the activation loop. X-ray cocrystallography revealed that upon binding of compound 4 to ERK2, Tyr34 undergoes a rotation (flip) along with a shift in the poly-Gly rich loop to create a new binding pocket into which 4 can bind. This new binding mode represents a novel mechanism by which high affinity ATP-competitive compounds may achieve excellent kinase selectivity.


Subject(s)
Anilides/metabolism , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Protein Kinase Inhibitors/chemical synthesis , Pyrrolidines/metabolism , Affinity Labels , Anilides/pharmacology , Crystallography, X-Ray , Inhibitory Concentration 50 , Mass Spectrometry/methods , Mitogen-Activated Protein Kinase 1/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Pyrrolidines/pharmacology , Structure-Activity Relationship
8.
Expert Opin Investig Drugs ; 22(6): 675-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23705633

ABSTRACT

In this issue of Expert Opinion on Investigational Drugs, several protein kinases families and pathways underlying cancer and other diseases are reviewed and several small molecule inhibitors that are in clinical trials are further described. Highlights of these reviews and drug evaluations are summarized in this editorial.


Subject(s)
Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinases/drug effects , Animals , Clinical Trials as Topic , Drug Design , Humans , Neoplasms/enzymology , Neoplasms/pathology , Protein Kinases/metabolism
10.
J Biochem ; 147(4): 601-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20147452

ABSTRACT

The activation of receptor tyrosine kinases (RTKs) is tightly regulated through a variety of mechanisms. Kinetic studies show that activation of c-Kit RTK occurs through an inter-molecular autophosphorylation. Phosphopeptide mapping of c-Kit reveals that 14-22 phosphates are added to each mol of wild-type (WT) c-Kit during the activation. Phosphorylation sites are found on the JM, kinase insert (KID), c-terminal domains and the activation loop (A-loop), but only the sites on the JM domain contribute to the kinase activation. The A-loop tyrosine (Y(823)) is not phosphorylated until very late in the activation (>90% completion), indicating that the A-loop phosphorylation is not required for c-Kit activation. A sunitinib-resistant mutant D816H that accelerates auto-activation by 184-fold shows no phosphorylation on the A-loop tyrosine after full activation. A loss-of-phosphorylation mutation Y823F remains fully competent in auto-activation. Similar to WT and D816H, the unactivated Y823F mutant binds sunitinib and imatinib with high affinity (K(D) = 5.9 nM). But unlike the WT and D816H where the activated enzymes lose the ability to bind the two drugs, activated Y823F binds the two inhibitors effectively. These observations suggest that the A-loop of activated Y823F remains flexible and can readily adopt unactivated conformations to accommodate DFG-out binders.


Subject(s)
Antineoplastic Agents/metabolism , Drug Resistance, Neoplasm , Enzyme Inhibitors/metabolism , Indoles/metabolism , Phosphotyrosine/physiology , Protein Interaction Domains and Motifs/physiology , Proto-Oncogene Proteins c-kit/metabolism , Pyrroles/metabolism , Amino Acid Substitution , Benzamides , Catalytic Domain , Enzyme Activation , Humans , Imatinib Mesylate , Kinetics , Microchemistry/methods , Models, Biological , Mutant Proteins/chemistry , Mutant Proteins/isolation & purification , Mutant Proteins/metabolism , Peptide Mapping , Phosphorylation , Piperazines/metabolism , Protein Binding , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/chemistry , Proto-Oncogene Proteins c-kit/genetics , Pyrimidines/metabolism , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Sunitinib
11.
Chem Biol Drug Des ; 74(6): 547-59, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19843080

ABSTRACT

The design, synthesis and utility of fluorescence probes that bind to the DFG-out conformation of p38alpha kinase are described. Probes that demonstrate good affinity for p38alpha, have been identified and one of the probes, PF-04438255, has been successfully used in an high throughput screening (HTS) assay to identify two novel non-classical p38alpha inhibitors. In addition, a cascade activity assay was utilized to validate the selective binding of these non-classical kinase inhibitors to the unactive form of the enzyme.


Subject(s)
Fluorescent Dyes/chemical synthesis , Mitogen-Activated Protein Kinase 14/metabolism , Protein Kinase Inhibitors/chemical synthesis , Binding Sites , Computer Simulation , Crystallography, X-Ray , Fluorescent Dyes/chemistry , Fluorescent Dyes/pharmacology , High-Throughput Screening Assays , Kinetics , Mitogen-Activated Protein Kinase 14/chemistry , Naphthalenes/chemistry , Naphthalenes/pharmacology , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Pyrazoles/chemistry , Pyrazoles/pharmacology , Structure-Activity Relationship
12.
Proc Natl Acad Sci U S A ; 106(5): 1542-7, 2009 Feb 03.
Article in English | MEDLINE | ID: mdl-19164557

ABSTRACT

Most gastrointestinal stromal tumors (GISTs) exhibit aberrant activation of the receptor tyrosine kinase (RTK) KIT. The efficacy of the inhibitors imatinib mesylate and sunitinib malate in GIST patients has been linked to their inhibition of these mutant KIT proteins. However, patients on imatinib can acquire secondary KIT mutations that render the protein insensitive to the inhibitor. Sunitinib has shown efficacy against certain imatinib-resistant mutants, although a subset that resides in the activation loop, including D816H/V, remains resistant. Biochemical and structural studies were undertaken to determine the molecular basis of sunitinib resistance. Our results show that sunitinib targets the autoinhibited conformation of WT KIT and that the D816H mutant undergoes a shift in conformational equilibrium toward the active state. These findings provide a structural and enzymologic explanation for the resistance profile observed with the KIT inhibitors. Prospectively, they have implications for understanding oncogenic kinase mutants and for circumventing drug resistance.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm/genetics , Gastrointestinal Stromal Tumors/drug therapy , Indoles/therapeutic use , Mutation , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , Receptor Protein-Tyrosine Kinases/metabolism , Antineoplastic Agents/metabolism , Benzamides , Gastrointestinal Stromal Tumors/enzymology , Gastrointestinal Stromal Tumors/genetics , Humans , Imatinib Mesylate , Indoles/metabolism , Phosphorylation , Piperazines/metabolism , Pyrimidines/metabolism , Pyrroles/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Spectrometry, Fluorescence , Sunitinib
13.
J Biol Chem ; 282(45): 32613-22, 2007 Nov 09.
Article in English | MEDLINE | ID: mdl-17848561

ABSTRACT

Defects in soluble NSF attachment protein receptor (SNARE)-mediated granule exocytosis occur in islet beta cells, adipocytes, and/or skeletal muscle cells correlate with increased susceptibility to insulin resistance and diabetes. The serine/threonine kinase WNK1 (with no K (lysine)) has recently been implicated in exocytosis and is expressed in all three of these cell types. To search for WNK1 substrates related to exocytosis, we conducted a WNK1 two-hybrid screen, which yielded Munc18c. Munc18c is known to be a key regulator of accessibility of the target membrane (t-SNARE) protein syntaxin 4 to participate in SNARE core complex assembly, although a paucity of Munc18c-binding factors has precluded discovery of its precise functions. To validate WNK1 as a new Munc18c-interacting partner, the direct interaction between WNK1 and Munc18c was confirmed using in vitro binding analysis, and endogenous WNK1-Munc18c complexes were detected in the cytosolic and plasma membrane compartments of the islet beta cell line MIN6. This binding interaction is mediated through the N-terminal 172 residues of Munc18c and the kinase domain residues of WNK1 (residues 159-491). Expression of either of these two minimal interaction domains resulted in inhibition of glucose-stimulated insulin secretion, consistent with a functional importance for the endogenous WNK1-Munc18c complex in exocytosis. Interestingly, Munc18c failed to serve as a WNK1 substrate in kinase activity assays, suggesting that WNK1 functions in SNARE complex assembly outside its role as a kinase. Taken together, these data support a novel role for WNK1 and a new mechanism for the regulation of SNARE complex assembly by WNK1-Munc18c complexes.


Subject(s)
Exocytosis , Munc18 Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Qa-SNARE Proteins/metabolism , SNARE Proteins/metabolism , Amino Acid Motifs , Animals , Cell Line , Exocytosis/drug effects , Humans , Insulin/pharmacology , Munc18 Proteins/genetics , Mutation/genetics , Protein Binding , Protein Serine-Threonine Kinases/genetics , Solubility
14.
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
15.
Proc Natl Acad Sci U S A ; 102(29): 10315-20, 2005 Jul 19.
Article in English | MEDLINE | ID: mdl-16006511

ABSTRACT

WNK (with no lysine [K]) kinases are serine-threonine protein kinases with an atypical placement of the catalytic lysine. Intronic deletions increase the expression of WNK1 in humans and cause pseudohypoaldosteronism type II, a form of hypertension. WNKs have been linked to ion carriers, but the underlying regulatory mechanisms are unknown. Here, we report a mechanism for the control of ion permeability by WNK1. We show that WNK1 activates the serum- and glucocorticoid-inducible protein kinase SGK1, leading to activation of the epithelial sodium channel. Increased channel activity induced by WNK1 depends on SGK1 and the E3 ubiquitin ligase Nedd4-2. This finding provides compelling evidence that this molecular mechanism contributes to the pathogenesis of hypertension in pseudohypoaldosteronism type II caused by WNK1 and, possibly, in other forms of hypertension.


Subject(s)
Immediate-Early Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Pseudohypoaldosteronism/physiopathology , Sodium Channels/metabolism , Animals , CHO Cells , Cells, Cultured , Cricetinae , Cricetulus , Endosomal Sorting Complexes Required for Transport , Enzyme Activation/physiology , Epithelial Sodium Channels , Humans , Immunoblotting , Immunoprecipitation , Intracellular Signaling Peptides and Proteins , Minor Histocompatibility Antigens , Nedd4 Ubiquitin Protein Ligases , Patch-Clamp Techniques , Pseudohypoaldosteronism/metabolism , Two-Hybrid System Techniques , Ubiquitin-Protein Ligases/metabolism , WNK Lysine-Deficient Protein Kinase 1
16.
Biochem J ; 381(Pt 3): 635-44, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15109307

ABSTRACT

MAPK (mitogen-activated protein kinase) pathways constitute major regulators of cellular transcriptional programmes. We analysed the ERK1,2 (extracellular-signal-regulated kinase 1,2) transcriptome in a non-transformed MEC (mammary epithelial cell) line, MCF-12A, utilizing rAd MEK1EE, a recombinant adenovirus encoding constitutively active MEK1 (MAPK/ERK kinase 1). rAd MEK1EE infection induced morphological changes and DNA synthesis which were inhibited by the MEK1,2 inhibitor PD184352. Hierarchical clustering of data derived from seven time points over 24 h identified 430 and 305 co-ordinately up-regulated and down-regulated genes respectively. c-Myc binding sites were identified in the promoters of most of these up-regulated genes. A total of 46 candidate effectors of the Raf/MEK/ERK1,2 pathway in MECs were identified by comparing our dataset with previously reported Raf-1-regulated genes. These analyses led to the identification of a suite of growth factors co-ordinately induced by MEK1EE, including multiple ErbB ligands, vascular endothelial growth factor and PHRP (parathyroid hormone-related protein). PHRP is the primary mediator of humoral hypercalcaemia of malignancy, and has been implicated in metastasis to bone. We demonstrate that PHRP is secreted by MEK1EE-expressing cells. This secretion is inhibited by PD184352, but not by ErbB inhibitors. Our results suggest that, in addition to anti-proliferative properties, MEK1,2 inhibitors may be anti-angiogenic and possess therapeutic utility in the treatment of PHRP-positive tumours.


Subject(s)
Breast/cytology , Breast/enzymology , Epithelial Cells/enzymology , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 3/genetics , Transcription, Genetic , Adenoviridae/genetics , Apoptosis/genetics , Binding Sites/genetics , Cell Cycle/physiology , Cell Line , Chromosome Mapping/methods , Cytoskeleton/enzymology , Cytoskeleton/metabolism , DNA Repair/genetics , Dual Specificity Phosphatase 1 , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Enzymologic/physiology , Genes/physiology , Genes, BRCA1/physiology , Genes, BRCA2/physiology , Genes, Immediate-Early/genetics , Genetic Vectors/biosynthesis , Humans , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/physiology , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Oligonucleotide Array Sequence Analysis/methods , Protein Phosphatase 1 , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/physiology , Proto-Oncogene Proteins c-raf/genetics , Transcription Factors/genetics
17.
Exp Cell Res ; 273(2): 138-46, 2002 Feb 15.
Article in English | MEDLINE | ID: mdl-11822869

ABSTRACT

Farnesyl protein transferase inhibitors (FTIs) reverse the transformed phenotype of fibroblasts expressing activated H-Ras and block anchorage-independent growth and tumorigenesis of tumor cell lines independent of their Ras mutational status. FTIs induce significant tumor regression accompanied by apoptosis in several transgenic mouse tumor models. FTI treatment of tumor cells in vitro is proapoptotic under certain cell culture conditions. Induction of apoptosis by FTIs in vitro generally requires a second death-promoting signal. To better understand FTI-induced apoptosis we analyzed the effect of SCH 66336, a tricyclic FTI, on apoptosis of Ras-transformed Rat2 fibroblasts. Treatment of H-Ras-CVLS-transformed fibroblasts with MEK1,2 inhibitors provides a pharmacological second signal to enhance FTI-induced apoptosis. Simultaneous treatment of these cells with a MEK1,2 inhibitor markedly enhanced caspase-3 activity and the apoptotic response to SCH 66336. The combination treatment resulted in a more complete and sustained inhibition of MAPK pathway activity than observed with either drug alone. Surprisingly, after treatment with either agent alone or in combination, no apoptotic response was observed in Rat2 cells transformed with a geranylgeranylated form of H-Ras (H-Ras-CVLL). Differences were also observed when SCH 66336 treatment was combined with forced suspension growth or serum withdrawal, in that an increase in drug-induced apoptosis was observed in H-Ras-CVLS-transformed Rat2 cells but not H-Ras-CVLL-transformed Rat2 cells. The lack of apoptotic effect of SCH 66336 and MEK inhibitor, alone or in combination, in H-Ras-CVLL-transformed cells suggests a difference in the reliance of cells transformed with farnesylated and geranylgeranylated forms of H-Ras on the MAPK signal transduction cascade for survival. K-Ras-transformed cells underwent apoptosis upon MEK1,2 inhibition but not in response to SCH 66336 treatment. The apoptotic response induced by MEK1,2 inhibitors is much greater in magnitude in H-Ras-transformed cells than in K-Ras-transformed cells, also pointing to differences in pathway utilization and/or dependence for these two Ras isoforms.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Apoptosis , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Protein Prenylation , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Butadienes/pharmacology , Cell Line , Cell Line, Transformed , Enzyme Inhibitors/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Flavonoids/pharmacology , MAP Kinase Kinase 1 , MAP Kinase Kinase 2 , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Nitriles/pharmacology , Phosphorylation , Piperidines/pharmacology , Proto-Oncogene Proteins p21(ras)/genetics , Pyridines/pharmacology , Rats
18.
Trends Pharmacol Sci ; 23(1): 40-5, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11804650

ABSTRACT

Mitogen-activated protein kinases [MAPKs, also called extracellular signal-regulated kinases (ERKs)] are constituents of numerous signal transduction pathways, and are activated by protein kinase cascades. Intense efforts are under way to develop and evaluate compounds that target components of MAPK pathways. In this article, the current status of inhibitors of MAPK pathways will be presented with a focus on the properties of small-molecule inhibitors of p38, MEK1 and MEK2 protein kinases. Several of these inhibitors are effective in animal models of disease and have advanced to clinical trials for the treatment of inflammatory diseases and cancer. The clinical utility of specifically targeting a subset of cellular signaling cascades and signaling cascades that regulate pleiotropic cellular processes are being evaluated. The results of these efforts have broad implications for the treatment of many diseases.


Subject(s)
Enzyme Inhibitors/pharmacology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Drug Design , Drugs, Investigational , Humans , MAP Kinase Kinase 1 , MAP Kinase Kinase 2 , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-raf/antagonists & inhibitors , Substrate Specificity , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...