Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Sci Transl Med ; 13(591)2021 04 28.
Article in English | MEDLINE | ID: mdl-33910977

ABSTRACT

Acute lung injury (ALI) causes high mortality and lacks any pharmacological intervention. Here, we found that pazopanib ameliorated ALI manifestations and reduced mortality in mouse ALI models and reduced edema in human lung transplantation recipients. Pazopanib inhibits mitogen-activated protein kinase kinase kinase 2 (MAP3K2)- and MAP3K3-mediated phosphorylation of NADPH oxidase 2 subunit p47phox at Ser208 to increase reactive oxygen species (ROS) formation in myeloid cells. Genetic inactivation of MAP3K2 and MAP3K3 in myeloid cells or hematopoietic mutation of p47phox Ser208 to alanine attenuated ALI manifestations and abrogates anti-ALI effects of pazopanib. This myeloid MAP3K2/MAP3K3-p47phox pathway acted via paracrine H2O2 to enhance pulmonary vasculature integrity and promote lung epithelial cell survival and proliferation, leading to increased pulmonary barrier function and resistance to ALI. Thus, pazopanib has the potential to be effective for treating ALI.


Subject(s)
Acute Lung Injury , Indazoles/pharmacology , MAP Kinase Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase Kinase 3/antagonists & inhibitors , Pyrimidines/pharmacology , Sulfonamides/pharmacology , Acute Lung Injury/drug therapy , Animals , Humans , Hydrogen Peroxide , Mice , NADPH Oxidases/metabolism , Phosphorylation , Reactive Oxygen Species
2.
Nat Commun ; 11(1): 5704, 2020 11 11.
Article in English | MEDLINE | ID: mdl-33177525

ABSTRACT

Neurofibromatosis type I (NF1) is characterized by prominent skeletal manifestations caused by NF1 loss. While inhibitors of the ERK activating kinases MEK1/2 are promising as a means to treat NF1, the broad blockade of the ERK pathway produced by this strategy is potentially associated with therapy limiting toxicities. Here, we have sought targets offering a more narrow inhibition of ERK activation downstream of NF1 loss in the skeleton, finding that MEKK2 is a novel component of a noncanonical ERK pathway in osteoblasts that mediates aberrant ERK activation after NF1 loss. Accordingly, despite mice with conditional deletion of Nf1 in mature osteoblasts (Nf1fl/fl;Dmp1-Cre) and Mekk2-/- each displaying skeletal defects, Nf1fl/fl;Mekk2-/-;Dmp1-Cre mice show an amelioration of NF1-associated phenotypes. We also provide proof-of-principle that FDA-approved inhibitors with activity against MEKK2 can ameliorate NF1 skeletal pathology. Thus, MEKK2 functions as a MAP3K in the ERK pathway in osteoblasts, offering a potential new therapeutic strategy for the treatment of NF1.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Imidazoles/pharmacology , MAP Kinase Kinase Kinase 2/metabolism , Neurofibromatosis 1/etiology , Pyridazines/pharmacology , Animals , Disease Models, Animal , Enzyme Activation , Extracellular Matrix Proteins/genetics , Female , Humans , MAP Kinase Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase Kinase 2/genetics , Male , Mice, Transgenic , Neurofibromatosis 1/drug therapy , Neurofibromin 1/genetics , Neurofibromin 1/metabolism , Osteoblasts/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Skull/cytology
3.
Arch. Soc. Esp. Oftalmol ; 95(10): 501-506, oct. 2020. ilus
Article in Spanish | IBECS | ID: ibc-201402

ABSTRACT

Mujer de 58 años, con antecedentes personales de melanoma metastásico en tratamiento con trametinib y dabrafenib, que presentaba disminución de agudeza visual bilateral. En la exploración se podía observar uveítis anterior, vitritis, desprendimiento de retina seroso, vasculitis y edema de disco en ambos ojos; se la diagnosticó de síndrome de Vogt-Koyanagi-Harada-like secundario a inhibición de la vía MAP quinasa. Además de la retirada de los fármacos oncológicos, se pautaron corticoides sistémicos y tópicos, pero este tratamiento consiguió solo una mejoría parcial del cuadro cuando se reintrodujo la terapia biológica; se añadió, por tanto, un implante de dexametasona intravítreo bilateral que consiguió una evolución favorable en su sintomatología y en los hallazgos de las pruebas complementarias. La inflamación intraocular es una complicación descrita tras el tratamiento con trametinib y dabrafenib. Un diagnóstico preciso unido al tratamiento corticoideo, tanto sistémico como intravítreo, nos llevó a obtener óptimos resultados


The case is presented of a 58-year-old woman with a personal history of metastatic melanoma under treatment with trametinib and dabrafenib, as well as a decrease in bilateral visual acuity. On examination, it was observed that she had an anterior uveitis, vitritis, serous retinal detachment, vasculitis and disc oedema in both eyes. She was diagnosed with a Vogt-Koyanagi-Harada-like syndrome secondary to MAP kinase pathway inhibition. In addition to the withdrawal of the oncology drugs, systemic and topical corticosteroids were prescribed, but this treatment only achieved a partial improvement of the disease when biological therapy was re-introduced. Therefore, a bilateral intravitreal dexamethasone implant was added, which led to a favourable progression in her symptomatology, as well as in the findings of complementary tests. Intraocular inflammation is a complication described after treatment with trametinib and dabrafenib. An accurate diagnosis, added to corticosteroid treatment, systemic and intravitreal, led us to obtain optimal results


Subject(s)
Humans , Female , Middle Aged , Dexamethasone/administration & dosage , Drug Implants/administration & dosage , Uveomeningoencephalitic Syndrome/drug therapy , MAP Kinase Kinase Kinases/antagonists & inhibitors , Retinal Detachment/drug therapy , Uveomeningoencephalitic Syndrome/complications , Intravitreal Injections , MAP Kinase Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase Kinase 2/antagonists & inhibitors , Visual Acuity/drug effects , Melanoma/drug therapy , Panuveitis/drug therapy
4.
Expert Opin Pharmacother ; 21(16): 1943-1953, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32880495

ABSTRACT

INTRODUCTION: RAS-RAF-MEK-ERK signaling is implicated in tumor development by promoting cell proliferation and other cancer hallmarks. MEK1/2 kinases are up-regulated in the majority of human cancers due to activation of tyrosine kinase receptors, RAS proteins, BRAF kinase, or some other members of the MAPK pathway. Targeting of MEK1/2 kinases may counterbalance cancer progression. AREAS COVERED: The authors analyze the scientific publications relevant to selumetinib (AZD6244, ARRY-142886) systematically and provide their expert opinion. EXPERT OPINION: Selumetinib is an oral selective allosteric inhibitor of MEK1 and MEK2 kinases. Single-agent selumetinib is usually administered in hydrogen sulfate capsules 75 mg twice a day; combination with other therapeutic compounds may or may not require reduced dosing of this drug. The established dose for pediatric patients is 25 mg per square meter twice a day. Selumetinib was extensively evaluated in non-small cell lung cancer (NSCLC) patients. Studies utilizing this drug as a monotherapy did not confirm its efficacy toward NSCLC. A phase II trial showed that the addition of selumetinib to docetaxel improved response rates and progression-free survival (PFS) in chemotherapy-pretreated KRAS-mutated NSCLC patients; however, a subsequent phase III study did not confirm these findings. There are several highly successful non-NSCLC selumetinib trials involving, e.g., patients with neurofibromatosis type 1 related tumors and children with low-grade BRAF-driven gliomas.


Subject(s)
Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzimidazoles/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Administration, Oral , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Benzimidazoles/administration & dosage , Benzimidazoles/adverse effects , Carcinoma, Non-Small-Cell Lung/mortality , Cell Proliferation/drug effects , Child , Clinical Trials as Topic , Drug Administration Schedule , Female , Humans , Lung Neoplasms/mortality , MAP Kinase Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase Kinase 2/antagonists & inhibitors , Progression-Free Survival
6.
Sci Rep ; 9(1): 16010, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31690749

ABSTRACT

Serine-threonine kinase 38 (STK38) is a member of the protein kinase A (PKA)/PKG/PKC-family implicated in the regulation of cell division and morphogenesis. However, the molecular mechanisms underlying STK38 stability remain largely unknown. Here, we show that treatment of cells with either heat or the calcium ionophore A23187 induced STK38 degradation. The calpain inhibitor calpeptin suppressed hyperthermia-induced degradation or the appearance of A23187-induced cleaved form of STK38. An in vitro cleavage assay was then used to demonstrate that calpain I directly cleaves STK38 at the proximal N-terminal region. Deletion of the N-terminal region of STK38 increased its stability against hyperthermia. We further demonstrated that the MAPKK kinase (MAP3K) MEKK2 prevented both heat- and calpain-induced cleavage of STK38. MEKK2 knockdown enhanced hyperthermia-induced degradation of STK38. We performed an in vitro MEKK2 assay and identified the key regulatory site in STK38 phosphorylated by MEKK2. Experiments with a phosphorylation-defective mutant demonstrated that phosphorylation of Ser 91 is important for STK38 stability, as the enzyme is susceptible to degradation by the calpain pathway unless this residue is phosphorylated. In summary, we demonstrated that STK38 is a calpain substrate and revealed a novel role of MEKK2 in the process of STK38 degradation by calpain.


Subject(s)
Calpain/metabolism , MAP Kinase Kinase Kinase 2/metabolism , Protein Serine-Threonine Kinases/metabolism , Algorithms , Calcimycin/pharmacology , Calpain/antagonists & inhibitors , Cell Line, Tumor , Dipeptides/pharmacology , Humans , MAP Kinase Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase Kinase 2/genetics , Mutagenesis, Site-Directed , Phosphorylation/drug effects , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Protein Stability , Proteolysis/drug effects , RNA Interference , RNA, Small Interfering/metabolism , Temperature
7.
Biochem Biophys Res Commun ; 496(1): 205-211, 2018 01 29.
Article in English | MEDLINE | ID: mdl-29309787

ABSTRACT

The kinase MEKK2 (MAP3K2) activates the MEK5/ERK5 cell signaling pathway and may play an important role in tumor growth and metastasis. Thus, MEKK2 may represent a novel kinase target for cancer. In order to identify inhibitors of MEKK2, we screened a library of compounds using a high throughput MEKK2 intrinsic ATPase enzyme assay. We identified two hits with validated structures and confirmed activity in the primary assay (IC50 values = 322 nM and 7.7 µM) and two orthogonal MEKK2 biochemical assays. Compound 1, the more potent hit, was the subject of further investigation. Limited structure-activity relationship (SAR) studies were performed on this iminocoumarin hit which resulted in ≥20-fold more potent analogs (e.g. 8 and 16 nM IC50). Two analogs had improved selectivity in a 50-member kinase profiling panel compared to the hit. These studies suggested that substitutions around the phenoxy ring of this scaffold can impart improved potency and selectivity for MEKK2. Analog Compound 1s (16 nM IC50) was further verified by external testing to inhibit MEKK2 and MEKK3 with similar potencies. Compound 1s displayed activity in cell-based assays in which it inhibited ERK5 pathway activation in cells and inhibited cell migration in a scratch assay. Thus, we have identified a scaffold that has promising potential to be developed into a highly selective and potent inhibitor of MEKK2. Information from these SAR studies provides specific guidance for the future design of MEKK2 inhibitor probes.


Subject(s)
Coumarins/chemistry , Coumarins/metabolism , MAP Kinase Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase Kinase 2/metabolism , Protein Interaction Mapping/methods , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Cells, Cultured , Coumarins/administration & dosage , Drug Delivery Systems/methods , Drug Discovery , Drug Evaluation, Preclinical/methods , Humans , Protein Kinase Inhibitors/administration & dosage
8.
Eur J Clin Pharmacol ; 73(6): 717-726, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28283692

ABSTRACT

PURPOSE: Emerging data on selumetinib, a MEK1/2 inhibitor in clinical development, suggest a possible difference in pharmacokinetics (PK) between Japanese and Western patients. This pooled analysis sought to assess the effect of ethnicity on selumetinib exposure in healthy Western and Asian subjects, and to identify any association between genetic variants in the UGT1A1, CYP2C19 and ABCG2 genes and observed differences in selumetinib PK. METHODS: A pooled analysis of data from ten Phase I studies, one in Asian subjects (encompassing Japanese, non-Japanese Asian and Indian Asian subjects) and nine in Western subjects, was conducted. Key findings were derived from the collective exposure data across doses of 25, 35, 50 and 75 mg selumetinib; primary variables were dose-normalized AUC and Cmax. RESULTS: PK data from 308 subjects (10 studies) were available for the pooled analysis; genetic data from 87 subjects (3 studies) were available for the pharmacogenetic analysis. Dose-normalized AUC and Cmax were 35% (95% CI: 25-47%) and 39% (95% CI: 24-56%) higher in the pooled Asian group, respectively, compared with Western subjects. PK exposure parameters were similar between the Japanese, non-Japanese Asian and Indian groups. There was no evidence that the polymorphisms assessed in the genes UGT1A1, CYP2C19 and ABCG2 account for observed PK differences. CONCLUSIONS: Selumetinib exposure was higher in healthy Asian subjects compared with Western subjects, and these data provide valuable insight for clinicians to consider when treating patients of Asian ethnicity with selumetinib.


Subject(s)
Benzimidazoles/pharmacokinetics , Pharmacogenetics , Protein Kinase Inhibitors/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Area Under Curve , Asian People , Black People , Clinical Trials, Phase I as Topic , Cytochrome P-450 CYP2C19/genetics , Glucuronosyltransferase/genetics , Humans , MAP Kinase Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase Kinase 2/antagonists & inhibitors , Neoplasm Proteins/genetics , White People
9.
Invest New Drugs ; 35(5): 616-626, 2017 10.
Article in English | MEDLINE | ID: mdl-28194539

ABSTRACT

Background Dual inhibition of activated MAPK and mTOR signaling pathways may enhance the antitumor efficacy of the MEK 1/2 inhibitor pimasertib and the mTOR inhibitor temsirolimus given in combination. Methods In this phase I study, patients with refractory advanced solid tumors (NCT01378377) received once-weekly temsirolimus plus once-daily oral pimasertib in 21-day cycles in a modified 3 + 3 dose-escalation design. The maximum tolerated dose (MTD) and recommended phase 2 dose (RP2D) of pimasertib in combination with temsirolimus, safety and pharmacokinetics (PK) were investigated. Results Of 33 patients evaluated, all experienced ≥1 treatment-emergent adverse event (TEAE) and 31 had treatment-related TEAEs, most frequently stomatitis and thrombocytopenia. TEAEs were reversible. No deaths were attributed to treatment. Nine patients had dose-limiting toxicities (stomatitis, thrombocytopenia, serum creatinine phosphokinase increase, visual impairment) and the MTD was determined as 45 mg/day pimasertib plus 25 mg/week temsirolimus. However, due to overlapping toxicities no further investigations were performed and the RP2D was not defined. PK profiles of both agents were not adversely affected. Seventeen patients (17/26 patients) had a best response of stable disease; five had stable disease lasting >12 weeks. Conclusions The RP2D was not defined and the pimasertib plus temsirolimus combination investigated did not warrant further study.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms/drug therapy , TOR Serine-Threonine Kinases/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Female , Humans , MAP Kinase Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase Kinase 2/antagonists & inhibitors , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/metabolism , Niacinamide/administration & dosage , Niacinamide/analogs & derivatives , Protein Kinase Inhibitors/administration & dosage , Sirolimus/administration & dosage , Sirolimus/analogs & derivatives , Young Adult
10.
Eur J Clin Pharmacol ; 73(2): 175-184, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27889832

ABSTRACT

PURPOSE: Two phase I, open-label trials in healthy subjects assessed whether co-administration with CYP3A4/CYP2C19 inhibitors, itraconazole/fluconazole (study A), or CYP3A4 inducer, rifampicin (study B), affects the exposure, safety/tolerability and pharmacokinetics of selumetinib and its metabolite N-desmethyl selumetinib. METHODS: In study A (n = 26), subjects received a single dose of selumetinib 25 mg and, after 4 days of washout, were randomized to treatment 1 (itraconazole 200 mg twice daily on days 1-11) or treatment 2 (fluconazole 400 mg on day 1 then 200 mg/day on days 2-11) plus co-administration of single-dose selumetinib 25 mg on day 8 (selumetinib staggered 4 h after itraconazole/fluconazole dose); Twenty-one days after discharge/washout, subjects received the alternate treatment. In study B (n = 22), subjects received a single dose of selumetinib 75 mg (day 1) then rifampicin 600 mg/day (days 4-14) plus a single dose of selumetinib 75 mg on day 12. Pharmacokinetic analysis and safety assessments were performed. RESULTS: Selumetinib co-administered with itraconazole, fluconazole (selumetinib staggered 4 h after itraconazole/fluconazole dose), or rifampicin was well tolerated. Selumetinib exposure was higher when co-administered with itraconazole or fluconazole (area under the plasma concentration-time curve (AUC) increased by 49 and 53%, respectively; maximum plasma concentration (C max) increased by 19 and 26%, respectively) but lower when co-dosed with rifampicin (AUC and C max decreased by 51 and 26%, respectively) versus selumetinib dosed alone. Co-administration with itraconazole or rifampicin decreased N-desmethyl selumetinib AUC(0-t) (11 and 55%, respectively), and C max (25 and 18%, respectively), with fluconazole, AUC(0-t) increased by 40%, but there was no effect on C max. CONCLUSIONS: Co-administration of CYP3A4/CYP2C19 inhibitors will likely increase exposure to selumetinib, while CYP3A4 inducers will likely reduce its exposure.


Subject(s)
Benzimidazoles/pharmacokinetics , Cytochrome P-450 CYP2C19 Inhibitors/pharmacology , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Adolescent , Adult , Benzimidazoles/blood , Cross-Over Studies , Cytochrome P-450 CYP2C19/metabolism , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 Enzyme Inducers/pharmacology , Female , Fluconazole/pharmacology , Healthy Volunteers , Humans , Itraconazole/pharmacology , MAP Kinase Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase Kinase 2/antagonists & inhibitors , Male , Rifampin/pharmacology , Young Adult
11.
Oncotarget ; 7(32): 52354-52363, 2016 Aug 09.
Article in English | MEDLINE | ID: mdl-27429047

ABSTRACT

Biliary tract carcinomas (BTC) are malignant tumors with limited therapeutic options. Clinical experiences with anti-EGFR therapies have produced unsatisfactory results. The strategies of combined inhibition of EGFR and MEK1/2 could be a promising therapeutic option in BTC treatment. Preclinical activity of Panitumumab and Trametinib was tested in in vitro (EGI-1, MT-CHC01 and WITT cells) and in in vivo (xenograft) BTC models with different K-RAS mutational status. Trametinib reduced MAPK phosphorylation in wild type (WT) WITT cells and in both K-RAS mutated cells; in EGI-1 was also able to switch off EGFR activation. Panitumumab reduced the activation of its target only in EGI-1 cells, and of MAPK only in WITT cells. While Trametinib inhibited cell growth in K-RAS mutated cell lines, Panitumumab had no effect on proliferation independently by K-RAS status. The addition of Panitumumab to Trametinib did not significantly potentiate its anti-proliferative effect also in mutated cells. In vivo, Trametinib was able to significantly slow the tumor growth in K-RAS mutated xenograft models, but did not have effect on K-RAS WT cells; the addition of Panitumumab potentiated the Trametinib efficacy in MT-CHC01 and overcame the resistance to the anti-EGFR in WITT cells, in which the monotherapy was ineffective. Only in K-RAS mutated xenografts Trametinib alone or in combination with Panitumumab significantly decreased Ki67 positive cell fraction and CD31 angiogenesis markers. In conclusion, this preclinical study provides a rational to plan clinical trials assessing the efficacy of Trametinib in K-RAS mutated BTC patients and the combination with anti-EGFR in WT BTC patients.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Biliary Tract Neoplasms , Pyridones/pharmacology , Pyrimidinones/pharmacology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , ErbB Receptors/antagonists & inhibitors , Female , Humans , MAP Kinase Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase Kinase 2/antagonists & inhibitors , Mice , Mice, Inbred NOD , Mice, SCID , Panitumumab , Protein Kinase Inhibitors/pharmacology , Xenograft Model Antitumor Assays
12.
Biochem Biophys Res Commun ; 463(4): 888-93, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26056008

ABSTRACT

The kinase MEKK2 (MAP3K2) may play an important role in tumor growth and metastasis for several cancer types. Thus, targeting MEKK2 may represent a novel strategy for developing more effective therapies for cancer. In order to identify small molecules with MEKK2 inhibitory activity, we screened a collection of known kinase inhibitors using a high throughput MEKK2 intrinsic ATPase enzyme assay and confirmed activity of the most potent hits with this primary assay. We also confirmed activities of these known kinase inhibitors with an MEKK2 transphosphorylation slot blot assay using MKK6 as a substrate. We observed a good correlation in potencies between the two orthogonal MEKK2 kinase activity assay formats for this set of inhibitors. We report that ponatinib, AT9283, AZD7762, JNJ-7706621, PP121 and hesperadin had potent MEKK2 enzyme inhibitory activities ranging from 4.7 to 60 nM IC50. Ponatinib is an FDA-approved drug that potently inhibited MEKK2 enzyme activity with IC50 values of 10-16 nM. AT9283 is currently in clinical trials and produced MEKK2 IC50 values of 4.7-18 nM. This set of known kinase inhibitors represents some of the most potent in vitro MEKK2 inhibitors reported to date and may be useful as research tools. Although these compounds are not selective for MEKK2, the structures of these compounds give insight into pharmacophores that potently inhibit MEKK2 and could be used as initial leads to design highly selective inhibitors of MEKK2.


Subject(s)
Imidazoles/pharmacology , MAP Kinase Kinase Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Pyridazines/pharmacology , Animals , Humans , Inhibitory Concentration 50 , Phosphorylation
13.
Endocr Relat Cancer ; 21(5): 755-67, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25012986

ABSTRACT

Poorly differentiated tumors of the thyroid gland (PDTC) are generally characterized by a poor prognosis due to their resistance to available therapeutic approaches. The relative rarity of these tumors is a major obstacle to our understanding of the molecular mechanisms leading to tumor aggressiveness and drug resistance, and consequently to the development of novel therapies. By simultaneously activating Kras and deleting p53 (Trp53) in thyroid follicular cells, we have generated a novel mouse model that develops papillary thyroid cancer invariably progressing to PDTC. In several cases, tumors further progress to anaplastic carcinomas. The poorly differentiated tumors are morphologically and functionally similar to their human counterparts and depend on MEK/ERK signaling for proliferation. Using primary carcinomas as well as carcinoma-derived cell lines, we also demonstrate that these tumors are intrinsically resistant to apoptosis due to high levels of expression of the Bcl2 family members, Bcl2a1 (Bcl2a1a) and Mcl1, and can be effectively targeted by Obatoclax, a small-molecule pan-inhibitor of the Bcl2 family. Furthermore, we show that Bcl2 family inhibition synergizes with MEK inhibition as well as with doxorubicin in inducing cell death. Thus, our studies in a novel, relevant mouse model have uncovered a promising druggable feature of aggressive thyroid cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Pyrroles/pharmacology , Thyroid Neoplasms/metabolism , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Humans , Indoles , MAP Kinase Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase Kinase 2/antagonists & inhibitors , Mice, Transgenic , Minor Histocompatibility Antigens , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyrroles/therapeutic use , RNA, Messenger/metabolism , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/pathology , Tumor Burden/drug effects
14.
J Biomol Screen ; 18(4): 388-99, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23134735

ABSTRACT

The kinase MEKK2 (MAP3K2) has recently been implicated in tumor growth and metastasis. Thus, selective inhibition of MEKK2 may be a novel strategy for cancer therapy. To identify inhibitors of MEKK2 kinase activity, we have developed a novel activity assay for MEKK2 based on the discovery that recombinant purified MEKK2 has intrinsic ATPase activity. This MEKK2 ATPase assay was validated for enzyme identity and enzymatic purity by multiple methods including mass spectrometry analysis, testing different sources of MEKK2 and comparing ATPase assay IC50 data for multiple inhibitors to literature values and to IC50 data generated using MEKK2 binding and transphosphorylation assays. Taken together, these data indicated that genuine MEKK2 activity was being measured in this assay and no other ATPases contributed to the signal. A miniaturized version of the assay was validated for high-throughput screening, and compound libraries were screened. The screening hits generated comparable potencies in the MEKK2 intrinsic ATPase, binding, and transphosphorylation assays. We identified a novel MEKK2 inhibitor and confirmed that crizotinib and bosutinib are potent in vitro inhibitors of MEKK2 activity with IC50 values of <100 nM. Thus, this assay has utility for the discovery of small-molecule inhibitors of MEKK2 activity.


Subject(s)
Adenosine Triphosphatases/metabolism , Drug Discovery , Enzyme Assays/methods , High-Throughput Screening Assays/methods , MAP Kinase Kinase Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Humans , Inhibitory Concentration 50 , MAP Kinase Kinase Kinase 2/metabolism , Phosphorylation/drug effects
15.
Eur J Cancer ; 47(4): 520-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21074412

ABSTRACT

Vascular endothelial growth factors (VEGF-C and VEGF-A) play important roles in tumour-induced lymphangiogenesis and angiogenesis, respectively, key processes implicated in promoting tumour growth and metastatic spread. Previous work from our laboratory has shown that EGFR overexpression in squamous carcinomas of the head and neck (SCCHN) is linked to high levels of VEGF-A and VEGF-C (but low levels of VEGF-D) and is associated with poor prognosis. The present study explored the signalling pathways regulating the induction of VEGF-C and VEGF-A in the SCCHN cell lines CAL 27 and Detroit 562. The addition of exogenous EGF induced the expression of VEGF-C and VEGF-A in a concentration-dependent manner and this was blocked by a selective EGFR inhibitor, gefitinib. In both cell lines stimulated with endogenous or exogenous ligand, inhibition of MEK1/2 (with U0126 or PD98059) or PI3K (with PI-103 or LY294002) resulted in a marked reduction of EGFR-induced VEGF-A expression, whereas exogenous EGF-induced VEGF-C upregulation was blocked by inhibitors of MEK but not PI3K. Inhibition of p38 MAPK suppressed EGF-induced VEGF-C upregulation in CAL 27 cells, but inhibited EGF-induced VEGF-A upregulation in Detroit 562. Taken together, our evidence suggests that both endogenous and exogenous EGFR activation induces VEGF-A expression requiring both PI3K and MAPK signalling whereas VEGF-C expression is dependent on MAPK, but not the PI3K or mTOR pathways in SCCHN cell lines. p38 MAPK appears to be differentially linked to either VEGF-A or VEGF-C regulation in different cellular contexts.


Subject(s)
Carcinoma, Squamous Cell/enzymology , Cytokines/metabolism , Head and Neck Neoplasms/enzymology , Mitogen-Activated Protein Kinases/physiology , Phosphatidylinositol 3-Kinases/physiology , Blotting, Western , Carcinoma, Squamous Cell/blood supply , Enzyme-Linked Immunosorbent Assay , Head and Neck Neoplasms/blood supply , Humans , Lymphangiogenesis/physiology , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Signaling System/physiology , Neovascularization, Pathologic/enzymology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Tumor Cells, Cultured , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor C/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
17.
Immunology ; 128(1 Suppl): e325-34, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19016906

ABSTRACT

Using two-dimensional sodium dodecyl sulphate-polyacrylamide gel electrophoresis, we found that copper/zinc superoxide dismutase (Cu/Zn-SOD, SOD-1) was induced in constructed CCR5 stably transfected HEK 293 cells, but not in mock cells, treated with CCL5. CCL5-induced SOD-1 expression was also confirmed in HEK 293-CCR5 cells and CCR5-positive granulocyte-macrophage colony-stimulating factor-induced human macrophages and murine macrophage RAW264.7 cells. CCL5 and CCR5 interaction induced SOD-1 expression mainly via MEK-ERK activation. In addition, we provided evidence that upregulation of SOD-1 by CCL5/CCR5 activation occurred in parallel with the increased release of tumour necrosis factor-alpha and nitric oxide and production of intracellular reactive oxygen species as well as enhanced nuclear factor-kappaB transcriptional activity in CCR5-positive RAW264.7 cells. Conversely, the MEK1/2 inhibitor PD98059 significantly inhibited SOD-1 expression with the decrease of these biological responses. More importantly, inhibition of SOD-1 activity by disulfiram also strongly inhibited the CCL5-induced biological effects. These data suggest that SOD-1 mediates CCR5 activation by CCL5 and that pharmacological modulation of SOD-1 may be beneficial to CCR5-associated diseases.


Subject(s)
Chemokine CCL5/metabolism , MAP Kinase Kinase Kinase 1/immunology , MAP Kinase Kinase Kinase 2/immunology , Macrophages/immunology , Receptors, CCR5/metabolism , Superoxide Dismutase/biosynthesis , Androstadienes/pharmacology , Animals , Cell Line , Chemokine CCL5/genetics , Down-Regulation/drug effects , Down-Regulation/immunology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Flavonoids/pharmacology , Humans , MAP Kinase Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase Kinase 1/metabolism , MAP Kinase Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase Kinase 2/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Nitric Oxide/agonists , Nitric Oxide/immunology , Nitric Oxide/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/drug effects , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Reactive Oxygen Species/agonists , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Receptors, CCR5/agonists , Superoxide Dismutase-1 , Transfection , Tumor Necrosis Factor-alpha/agonists , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/drug effects , Up-Regulation/immunology , Wortmannin , NF-kappaB-Inducing Kinase
18.
Mol Cancer Ther ; 5(12): 3285-93, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17172432

ABSTRACT

Epidemiologic studies have suggested an inverse correlation between dietary intake of cruciferous vegetables and cancer risk. It is thus of interest to investigate the anticancer potential of phytochemicals presented in cruciferous vegetables. In this study, methyl-3-indolylacetate (MIA), a cruciferous indole for which the bioactivity has not been previously reported, was found to significantly suppress the invasion of cancer cells stimulated by the 12-O-tetradecanoyl-phorbol-13-acetate (TPA). Our data show that MIA pretreatments inhibited matrix metalloproteinase 9 (MMP-9) expression in a concentration-dependent manner, resulting in decreased MMP-9 activity. By using real-time reverse transcription-PCR, luciferase reporter gene assay, and electrophoretic mobility shift assay, we provided convincing evidence that MIA suppresses MMP-9 gene transcription via targeting the activator protein-1 signaling but not the nuclear factor-kappaB pathway. The TPA-induced mitogen-activated protein kinase (MAPK) activation cascade was also analyzed. Despite extensive activation of major MAPKs [c-Jun NH2-terminal kinase, p38, and extracellular signal-regulated kinase-1/2 (ERK1/2)] under TPA stimulation, only the ERK1/2 activation and its consequent nuclear translocation were found to be diminished by MIA. Interestingly, MIA did not affect the TPA-induced phosphorylation of either c-Raf or MAPK/ERK kinase-1/2 (MEK1/2), two upstream kinases of ERK. Moreover, using the in vitro kinase assay, MIA was shown to inhibit the kinase activity of MEK1/2, the upstream kinases of ERK, suggesting that MEK is the major molecular target of MIA. In conclusion, data from this study provided new insight into the anticancer potential of MIA, a cruciferous vegetable-derived indole compound.


Subject(s)
Indoleacetic Acids/pharmacology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Cell Line, Tumor , HeLa Cells , Humans , MAP Kinase Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase Kinase 1/metabolism , MAP Kinase Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase Kinase 2/metabolism , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase Inhibitors , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Invasiveness , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Tetradecanoylphorbol Acetate/antagonists & inhibitors , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism , Transcription, Genetic/drug effects , Transfection
19.
Cell Prolif ; 39(4): 261-79, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16872362

ABSTRACT

Extracellular signal-regulated kinase activity is essential for mediating cell cycle progression from G(1) phase to S phase (DNA synthesis). In contrast, the role of extracellular signal-regulated kinase during G(2) phase and mitosis (M phase) is largely undefined. Previous studies have suggested that inhibition of basal extracellular signal-regulated kinase activity delays G(2)- and M-phase progression. In the current investigation, we have examined the consequence of activating the extracellular signal-regulated kinase pathway during G(2) phase on subsequent progression through mitosis. Using synchronized HeLa cells, we show that activation of the extracellular signal-regulated kinase pathway with phorbol 12-myristate 13-acetate or epidermal growth factor during G(2) phase causes a rapid cell cycle arrest in G(2) as measured by flow cytometry, mitotic indices and cyclin B1 expression. This G(2)-phase arrest was reversed by pre-treatment with bisindolylmaleimide or U0126, which are selective inhibitors of protein kinase C proteins or the extracellular signal-regulated kinase activators, MEK1/2, respectively. The extracellular signal-regulated kinase-mediated delay in M-phase entry appeared to involve de novo synthesis of the cyclin-dependent kinase inhibitor, p21(CIP1), during G(2) through a p53-independent mechanism. To establish a function for the increased expression of p21(CIP1) and delayed cell cycle progression, we show that extracellular signal-regulated kinase activation in G(2)-phase cells results in an increased number of cells containing chromosome aberrations characteristic of genomic instability. The presence of chromosome aberrations following extracellular signal-regulated kinase activation during G(2)-phase was further augmented in cells lacking p21(CIP1). These findings suggest that p21(CIP1) mediated inhibition of cell cycle progression during G(2)/M phase protects against inappropriate activation of signalling pathways, which may cause excessive chromosome damage and be detrimental to cell survival.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , G2 Phase , Mitosis , Butadienes/pharmacology , Chromosomal Instability , Cyclin B/metabolism , Cyclin B1 , Enzyme Activation , HeLa Cells , Humans , Indoles/pharmacology , MAP Kinase Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase Kinase 1/metabolism , MAP Kinase Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase Kinase 2/metabolism , Maleimides/pharmacology , Nitriles/pharmacology , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Signal Transduction , Tetradecanoylphorbol Acetate/pharmacology , Tumor Suppressor Protein p53/physiology
20.
EMBO J ; 25(1): 97-107, 2006 Jan 11.
Article in English | MEDLINE | ID: mdl-16362041

ABSTRACT

Members of the mitogen-activated protein kinase kinase kinase (MAP3K) family are crucial for the Toll-like receptor (TLR) signaling and cellular stress responses. However, the molecular mechanisms underlying the TLR- and cellular stress-mediated MAP3K activation remain largely unknown. In this study, we identified a key regulatory phosphorylation site, serine 519 and serine 526, in MAP3K MEKK2 and MEKK3, respectively. Mutation of this serine to an alanine severely impaired MEKK2/3 activation. We generated an anti-p-MEKK2/3 antibody and used this antibody to demonstrate that lipopolysaccharide induced MEKK2 and MEKK3 phosphorylation on their regulatory serine. We found that the serine phosphorylation was crucial for TLR-induced interleukin 6 production and this process is regulated by TRAF6, a key adaptor molecule for the TLR pathway. We further demonstrated that many, but not all, MAPK agonists induced the regulatory serine phosphorylation, suggesting an involvement of different MAP3Ks in activation of the MAPK cascades leading to different cellular responses. In conclusion, this study reveals a novel molecular mechanism for MEKK2/3 activation by the TLR and cellular stress pathways.


Subject(s)
MAP Kinase Kinase Kinase 2/metabolism , MAP Kinase Kinase Kinase 3/metabolism , Serine/metabolism , Toll-Like Receptors/metabolism , Amino Acid Sequence , Animals , Antibodies, Phospho-Specific/immunology , Cytokines/metabolism , Enzyme Activation , Interleukin-6/metabolism , Ligands , Lipopolysaccharides/pharmacology , MAP Kinase Kinase Kinase 2/antagonists & inhibitors , MAP Kinase Kinase Kinase 2/genetics , MAP Kinase Kinase Kinase 3/antagonists & inhibitors , MAP Kinase Kinase Kinase 3/genetics , Molecular Sequence Data , Mutation , Phosphoamino Acids/analysis , Phosphoamino Acids/immunology , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Serine/chemistry , Serine/genetics , TNF Receptor-Associated Factor 6/metabolism , Toll-Like Receptor 4/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...