Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Nat Cancer ; 4(2): 240-256, 2023 02.
Article in English | MEDLINE | ID: mdl-36759733

ABSTRACT

BRAFV600E mutation confers a poor prognosis in metastatic colorectal cancer (CRC) despite combinatorial targeted therapies based on the latest understanding of signaling circuitry. To identify parallel resistance mechanisms induced by BRAF-MEK-EGFR co-targeting, we used a high-throughput kinase activity mapping platform. Here we show that SRC kinases are systematically activated in BRAFV600E CRC following targeted inhibition of BRAF ± EGFR and that coordinated targeting of SRC with BRAF ± EGFR increases treatment efficacy in vitro and in vivo. SRC drives resistance to BRAF ± EGFR targeted therapy independently of ERK signaling by inducing transcriptional reprogramming through ß-catenin (CTNNB1). The EGFR-independent compensatory activation of SRC kinases is mediated by an autocrine prostaglandin E2 loop that can be blocked with cyclooxygenase-2 (COX2) inhibitors. Co-targeting of COX2 with BRAF + EGFR promotes durable suppression of tumor growth in patient-derived tumor xenograft models. COX2 inhibition represents a drug-repurposing strategy to overcome therapeutic resistance in BRAFV600E CRC.


Subject(s)
Colorectal Neoplasms , Proto-Oncogene Proteins B-raf , Humans , Cyclooxygenase 2/genetics , Cyclooxygenase 2/therapeutic use , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , MAP Kinase Signaling System , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , ErbB Receptors/genetics , src-Family Kinases/genetics , src-Family Kinases/therapeutic use
2.
Nat Cell Biol ; 21(6): 778-790, 2019 06.
Article in English | MEDLINE | ID: mdl-31160710

ABSTRACT

Phosphorylation networks intimately regulate mechanisms of response to therapies. Mapping the phospho-catalytic profile of kinases in cells or tissues remains a challenge. Here, we introduce a practical high-throughput system to measure the enzymatic activity of kinases using biological peptide targets as phospho-sensors to reveal kinase dependencies in tumour biopsies and cell lines. A 228-peptide screen was developed to detect the activity of >60 kinases, including ABLs, AKTs, CDKs and MAPKs. Focusing on BRAFV600E tumours, we found mechanisms of intrinsic resistance to BRAFV600E-targeted therapy in colorectal cancer, including targetable parallel activation of PDPK1 and PRKCA. Furthermore, mapping the phospho-catalytic signatures of melanoma specimens identifies RPS6KB1 and PIM1 as emerging druggable vulnerabilities predictive of poor outcome in BRAFV600E patients. The results show that therapeutic resistance can be caused by the concerted upregulation of interdependent pathways. Our kinase activity-mapping system is a versatile strategy that innovates the exploration of actionable kinases for precision medicine.


Subject(s)
3-Phosphoinositide-Dependent Protein Kinases/genetics , Colorectal Neoplasms/drug therapy , Melanoma/drug therapy , Protein Kinase C-alpha/genetics , Proto-Oncogene Proteins c-pim-1/genetics , Adult , Aged , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indoles/chemistry , Kaplan-Meier Estimate , MAP Kinase Signaling System/genetics , Male , Melanoma/genetics , Melanoma/pathology , Middle Aged , Peptides/chemistry , Peptides/therapeutic use , Phosphorylation/drug effects , Protein Kinase Inhibitors/therapeutic use , Sulfonamides/therapeutic use
3.
Nat Microbiol ; 3(12): 1472-1485, 2018 12.
Article in English | MEDLINE | ID: mdl-30478389

ABSTRACT

Plasma membrane integrity is essential for the viability of eukaryotic cells. In response to bacterial pore-forming toxins, disrupted regions of the membrane are rapidly repaired. However, the pathways that mediate plasma membrane repair are unclear. Here we show that autophagy-related (ATG) protein ATG16L1 and its binding partners ATG5 and ATG12 are required for plasma membrane repair through a pathway independent of macroautophagy. ATG16L1 is required for lysosome fusion with the plasma membrane and blebbing responses that promote membrane repair. ATG16L1 deficiency causes accumulation of cholesterol in lysosomes that contributes to defective membrane repair. Cell-to-cell spread by Listeria monocytogenes requires membrane damage by the bacterial toxin listeriolysin O, which is restricted by ATG16L1-dependent membrane repair. Cells harbouring the ATG16L1 T300A allele associated with inflammatory bowel disease were also found to accumulate cholesterol and be defective in repair, linking a common inflammatory disease to plasma membrane integrity. Thus, plasma membrane repair could be an important therapeutic target for the treatment of bacterial infections and inflammatory disorders.


Subject(s)
Autophagy-Related Proteins/metabolism , Autophagy-Related Proteins/pharmacology , Cell Membrane/drug effects , Cell Membrane/metabolism , Listeria monocytogenes/drug effects , Animals , Autophagy , Autophagy-Related Protein 12/metabolism , Autophagy-Related Protein 5/metabolism , Autophagy-Related Proteins/genetics , Bacterial Toxins/toxicity , Carrier Proteins/genetics , Carrier Proteins/metabolism , Carrier Proteins/pharmacology , Cholesterol/metabolism , Disease Models, Animal , Exocytosis , HeLa Cells , Heat-Shock Proteins/toxicity , Hemolysin Proteins/toxicity , Humans , Listeria monocytogenes/metabolism , Lysosomes , Male , Mice
4.
Cell ; 173(6): 1413-1425.e14, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29754815

ABSTRACT

BRAF(V600E) mutant melanomas treated with inhibitors of the BRAF and MEK kinases almost invariably develop resistance that is frequently caused by reactivation of the mitogen activated protein kinase (MAPK) pathway. To identify novel treatment options for such patients, we searched for acquired vulnerabilities of MAPK inhibitor-resistant melanomas. We find that resistance to BRAF+MEK inhibitors is associated with increased levels of reactive oxygen species (ROS). Subsequent treatment with the histone deacetylase inhibitor vorinostat suppresses SLC7A11, leading to a lethal increase in the already-elevated levels of ROS in drug-resistant cells. This causes selective apoptotic death of only the drug-resistant tumor cells. Consistently, treatment of BRAF inhibitor-resistant melanoma with vorinostat in mice results in dramatic tumor regression. In a study in patients with advanced BRAF+MEK inhibitor-resistant melanoma, we find that vorinostat can selectively ablate drug-resistant tumor cells, providing clinical proof of concept for the novel therapy identified here.


Subject(s)
Drug Resistance, Neoplasm , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Amino Acid Transport System y+/metabolism , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , MAP Kinase Kinase 1/metabolism , MAP Kinase Signaling System , Melanoma/genetics , Mice , Mutation , Neoplasm Transplantation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Reactive Oxygen Species/metabolism , Skin Neoplasms/genetics , Treatment Outcome , Vorinostat/pharmacology
5.
Mol Cancer Ther ; 17(4): 849-857, 2018 04.
Article in English | MEDLINE | ID: mdl-29440296

ABSTRACT

The majority of high-risk neuroblastoma patients are refractory to, or relapse on, current treatment regimens, resulting in 5-year survival rates of less than 50%. This emphasizes the urgent need to identify novel therapeutic targets. Here, we report that high PIM kinase expression is correlated with poor overall survival. Treatment of neuroblastoma cell lines with the pan-PIM inhibitors AZD1208 or PIM-447 suppressed proliferation through inhibition of mTOR signaling. In a panel of neuroblastoma cell lines, we observed a marked binary response to PIM inhibition, suggesting that specific genetic lesions control responses to PIM inhibition. Using a genome-wide CRISPR-Cas9 genetic screen, we identified NF1 loss as the major resistance mechanism to PIM kinase inhibitors. Treatment with AZD1208 impaired the growth of NF1 wild-type xenografts, while NF1 knockout cells were insensitive. Thus, our data indicate that PIM inhibition may be a novel targeted therapy in NF1 wild-type neuroblastoma. Mol Cancer Ther; 17(4); 849-57. ©2018 AACR.


Subject(s)
Neoplasm Recurrence, Local/pathology , Neuroblastoma/pathology , Neurofibromin 1/metabolism , Proto-Oncogene Proteins c-pim-1/metabolism , Animals , Apoptosis , Biomarkers, Tumor , Biphenyl Compounds/pharmacology , CRISPR-Cas Systems , Cell Proliferation , Female , Follow-Up Studies , Humans , Infant , Male , Mice , Mice, Nude , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/metabolism , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Neurofibromin 1/genetics , Prognosis , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/genetics , Survival Rate , Thiazolidines/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
6.
Nat Commun ; 8: 15580, 2017 05 31.
Article in English | MEDLINE | ID: mdl-28561042

ABSTRACT

Two genes are synthetically lethal (SL) when defects in both are lethal to a cell but a single defect is non-lethal. SL partners of cancer mutations are of great interest as pharmacological targets; however, identifying them by cell line-based methods is challenging. Here we develop MiSL (Mining Synthetic Lethals), an algorithm that mines pan-cancer human primary tumour data to identify mutation-specific SL partners for specific cancers. We apply MiSL to 12 different cancers and predict 145,891 SL partners for 3,120 mutations, including known mutation-specific SL partners. Comparisons with functional screens show that MiSL predictions are enriched for SLs in multiple cancers. We extensively validate a SL interaction identified by MiSL between the IDH1 mutation and ACACA in leukaemia using gene targeting and patient-derived xenografts. Furthermore, we apply MiSL to pinpoint genetic biomarkers for drug sensitivity. These results demonstrate that MiSL can accelerate precision oncology by identifying mutation-specific targets and biomarkers.


Subject(s)
Algorithms , Computational Biology/methods , Leukemia, Myeloid, Acute/genetics , Synthetic Lethal Mutations/genetics , Animals , Cell Line, Tumor , Female , Humans , MCF-7 Cells , Male , Mice , Neoplasm Transplantation , Precision Medicine/methods , RNA Interference , RNA, Small Interfering/genetics , Transplantation, Heterologous
8.
Oncotarget ; 7(25): 37407-37419, 2016 Jun 21.
Article in English | MEDLINE | ID: mdl-27270648

ABSTRACT

Although conventional therapies for acute myeloid leukemia (AML) and diffuse large B-cell lymphoma (DLBCL) are effective in inducing remission, many patients relapse upon treatment. Hence, there is an urgent need for novel therapies. PIM kinases are often overexpressed in AML and DLBCL and are therefore an attractive therapeutic target. However, in vitro experiments have demonstrated that intrinsic resistance to PIM inhibition is common. It is therefore likely that only a minority of patients will benefit from single agent PIM inhibitor treatment. In this study, we performed an shRNA-based genetic screen to identify kinases whose suppression is synergistic with PIM inhibition. Here, we report that suppression of p38α (MAPK14) is synthetic lethal with the PIM kinase inhibitor AZD1208. PIM inhibition elevates reactive oxygen species (ROS) levels, which subsequently activates p38α and downstream AKT/mTOR signaling. We found that p38α inhibitors sensitize hematological tumor cell lines to AZD1208 treatment in vitro and in vivo. These results were validated in ex vivo patient-derived AML cells. Our findings provide mechanistic and translational evidence supporting the rationale to test a combination of p38α and PIM inhibitors in clinical trials for AML and DLBCL.


Subject(s)
Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/enzymology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , K562 Cells , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 14/antagonists & inhibitors , Mitogen-Activated Protein Kinase 14/metabolism , Signal Transduction , Xenograft Model Antitumor Assays
9.
Clin Cancer Res ; 22(15): 3884-93, 2016 08 01.
Article in English | MEDLINE | ID: mdl-26936917

ABSTRACT

PURPOSE: FGFR1 is a promising therapeutic target in multiple types of solid tumors, including head and neck squamous cell carcinoma (HNSCC). FGFR inhibitors have shown great therapeutic value in preclinical models. However, resistance remains a major setback. In this study, we have investigated the prognostic value of FGFR1 expression in HNSCC, the therapeutic relevance of targeting FGFR with AZD4547, and potential resistant mechanisms. EXPERIMENTAL DESIGN: IHC and FISH were applied on tissue microarrays to investigate FGFR1 protein expression and FGFR1 gene copy numbers in 452 HNSCCs. The sensitivity of HNSCC cell lines to AZD4547, either as single or combination treatment with the EGFR inhibitor gefitinib, was assessed using long-term colony formation assays, short-term viability assays, and biochemical analysis. RESULTS: FGFR1 protein overexpression occurred in 82% (36/44) of human papillomavirus (HPV)-positive HNSCC and 75% (294/392) of HPV-negative HNSCC and relates with poor overall survival and disease-free survival in HPV-negative HNSCC [HR, 3.07; 95% confidence interval (CI), 1.74-6.90; P = 0.001 and HR, 1.53; 95% CI, 1.04-2.39; P = 0.033]. Moreover, the FGFR1 gene was amplified in 3% (3/110) of HPV-negative HNSCC. Treatment of the high FGFR1-expressing cell line CCL30 with AZD4547 reduced cell proliferation and FGFR signaling. Two FGFR-amplified cell lines, SCC147 and BICR16, were resistant to AZD4547 treatment due to EGFR signaling. Combined AZD4547 and gefitinib treatment synergistically inhibited the proliferation of resistant cell lines. CONCLUSIONS: Here, we identify high FGFR1 expression as a candidate prognostic biomarker in HPV-negative HNSCC. Furthermore, we provide a rationale for treating FGFR1-expressing HNSCC with the FGFR inhibitor AZD4547 and for combining AZD4547 and gefitinib in FGFR inhibitor-resistant HNSCC patients. Clin Cancer Res; 22(15); 3884-93. ©2016 AACR.


Subject(s)
Biomarkers, Tumor , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/mortality , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/mortality , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Benzamides/pharmacology , Benzamides/therapeutic use , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor , Combined Modality Therapy , Female , Gefitinib , Gene Amplification , Gene Expression , Head and Neck Neoplasms/diagnosis , Head and Neck Neoplasms/drug therapy , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Molecular Targeted Therapy , Neoplasm Metastasis , Neoplasm Staging , Piperazines/pharmacology , Piperazines/therapeutic use , Prognosis , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Quinazolines/pharmacology , Quinazolines/therapeutic use , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/genetics , Squamous Cell Carcinoma of Head and Neck
10.
Cell Cycle ; 12(18): 2960-8, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23974105

ABSTRACT

The transforming growth factor ß (TGF-ß) pathway acts as a double-edged sword in tumorigenesis. By constraining epithelial cell growth, TGF-ß is a potent tumor suppressor. However, TGF-ß also acts as a key player in the induction of epithelial-to-mesenchymal transition (EMT), thereby enhancing invasiveness and metastasis. Furthermore, TGF-ß signaling has recently been correlated with resistance against both targeted and conventional anticancer agents. Here, we present data demonstrating a role for TGF-ß in chemotherapy resistance in colorectal cancer (CRC). We discuss these results in the context of recent findings indicating TGF-ß signaling as an emerging player in cancer drug resistance.


Subject(s)
Drug Resistance, Neoplasm , Transforming Growth Factor beta/metabolism , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition , Humans , Mediator Complex/antagonists & inhibitors , Mediator Complex/genetics , Mediator Complex/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/antagonists & inhibitors
11.
PLoS One ; 6(4): e19047, 2011 Apr 20.
Article in English | MEDLINE | ID: mdl-21533107

ABSTRACT

Maintenance of Foxp3 protein expression in regulatory T cells (Treg) is crucial for a balanced immune response. We have previously demonstrated that Foxp3 protein stability can be regulated through acetylation, however the specific mechanisms underlying this observation remain unclear. Here we demonstrate that SIRT1 a member of the lysine deacetylase Sirtuin (SIRT) family, but not the related SIRTs 2-7, co-localize with Foxp3 in the nucleus. Ectopic expression of SIRT1, but not SIRTs 2-7 results in decreased Foxp3 acetylation, while conversely inhibition of endogenous SIRT activity increased Foxp3 acetylation. We show that SIRT1 inhibition decreases Foxp3 poly-ubiquitination, thereby increasing Foxp3 protein levels. Co-transfection of SIRT1 with Foxp3 results in increased Foxp3 proteasomal degradation, while SIRT inhibition increases FOXP3 transcriptional activity in human Treg. Taken together, these data support a central role for SIRT1 in the regulation of Foxp3 protein levels and thereby in regulation of Treg suppressive capacity. Pharmacological modulation of SIRT1 activity in Treg may therefore provide a novel therapeutic strategy for controlling immune responses.


Subject(s)
Forkhead Transcription Factors/metabolism , Sirtuin 1/metabolism , Acetylation , Blotting, Western , Cell Line , Humans , Hydrolysis , Immunoprecipitation , Kinetics , Microscopy, Confocal , T-Lymphocytes, Regulatory/metabolism , Ubiquitination
12.
AIDS ; 24(5): 669-73, 2010 Mar 13.
Article in English | MEDLINE | ID: mdl-19926962

ABSTRACT

OBJECTIVE: Bevirimat is the first drug of a new class of antivirals that hamper the maturation of HIV. The objective of this study was to evaluate the sequence variability of the gag region targeted by bevirimat in HIV subtype-B isolates. METHODS: Of 484 HIV subtype-B isolates, the gag region comprising amino acids 357-382 was sequenced. Of the patients included, 270 were treatment naive and 214 were treatment experienced. In the latter group, 48 HIV isolates harboured mutations associated with reverse transcriptase inhibitor resistance only, and 166 HIV isolates carried mutations associated with protease inhibitor resistance. RESULTS: In the treatment-naive patient population, approximately 30% harboured an HIV isolate with at least one mutation associated with a reduced susceptibility to bevirimat (H358Y, L363M, Q369H, V370A/M/del and T371del). In HIV isolates with protease inhibitor resistance, the prevalence of bevirimat resistance mutations increased to 45%. Accumulation of mutations at four positions in the bevirimat target region, S368C, Q369H, V370A and S373P, was significantly observed. Mutations associated with bevirimat resistance were detected more frequently in HIV isolates with three or more protease inhibitor resistance mutations than in those with less than three protease inhibitor mutations. CONCLUSION: Reduced bevirimat activity can be expected in one-third of treatment-naive HIV subtype-B isolates and significantly more in protease inhibitor-resistant HIV. These data indicate that screening for bevirimat resistance mutations before administration of the drug is essential.


Subject(s)
Drug Resistance, Viral/drug effects , HIV Infections/drug therapy , HIV Protease Inhibitors/pharmacology , HIV-1/drug effects , Mutation/genetics , Succinates/pharmacology , Triterpenes/pharmacology , gag Gene Products, Human Immunodeficiency Virus/genetics , Drug Resistance, Viral/genetics , Genotype , HIV Infections/genetics , HIV-1/genetics , Humans , Molecular Sequence Data , Prevalence , Sequence Analysis, DNA , gag Gene Products, Human Immunodeficiency Virus/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...