Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Cell Death Discov ; 10(1): 175, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38622136

ABSTRACT

Papillary thyroid carcinoma (PTC) is the most frequent form of thyroid cancer. PTC commonly presents with mutations of the serine/threonine kinase BRAF (BRAFV600E), which drive ERK1/2 pathway activation to support growth and suppress apoptosis. PTC patients often undergo surgical resection; however, since the average age of PTC patients is under 50, adverse effects associated with prolonged maintenance therapy following total thyroidectomy are a concern. The development of mutant-selective BRAF inhibitors (BRAFi), like vemurafenib, has been efficacious in patients with metastatic melanoma, but the response rate is low for mutant BRAF PTC patients. Here, we assay the therapeutic response of BRAFi in a panel of human PTC cell lines and freshly biopsied patient samples. We observed heterogeneous responses to BRAFi, and multi-omic comparisons between susceptible and resistant mutant BRAF PTC revealed overrepresented stress response pathways and the absence of compensatory RTK activation - features that may underpin innate resistance. Importantly, resistant cell lines and patient samples had increased hallmarks of failed apoptosis; a cellular state defined by sublethal caspase activation and DNA damage. Further analysis suggests that the failed apoptotic phenotypes may have features of "minority mitochondrial outer membrane permeabilization (MOMP)" - a stress-related response characterized by fragmented and porous mitochondria known to contribute to cancer aggressiveness. We found that cells presenting with minority MOMP-like phenotypes are dependent on the apoptotic regulator, Mcl-1, as treatment with the Mcl-1 inhibitor, AZD5991, potently induced cell death in resistant cells. Furthermore, PI3K/AKT inhibitors sensitized resistant cells to BRAFi; an effect that was at least in part associated with reduced Mcl-1 levels. Together, these data implicate minority MOMP as a mechanism associated with intrinsic drug resistance and underscore the benefits of targeting Mcl-1 in mutant BRAF PTC.

2.
Mol Cancer Res ; 20(8): 1260-1271, 2022 08 05.
Article in English | MEDLINE | ID: mdl-35426938

ABSTRACT

BRCA1-associated protein 1 (BAP1) is a tumor suppressor gene that is mutated in cancer, including uveal melanoma. Loss-of-function BAP1 mutations are associated with uveal melanoma metastasis and poor prognosis, but the mechanisms underlying these effects remain unclear. Upregulation of cell-cell adhesion proteins is involved with collective migration and metastatic seeding of cancer cells. Here, we show that BAP1 loss in uveal melanoma patient samples is associated with upregulated gene expression of multiple cell adhesion molecules (CAM), including E-cadherin (CDH1), cell adhesion molecule 1 (CADM1), and syndecan-2 (SDC2). Similar findings were observed in uveal melanoma cell lines and single-cell RNA-sequencing data from uveal melanoma patient samples. BAP1 reexpression in uveal melanoma cells reduced E-cadherin and CADM1 levels. Functionally, knockdown of E-cadherin decreased spheroid cluster formation and knockdown of CADM1 decreased growth of BAP1-mutant uveal melanoma cells. Together, our findings demonstrate that BAP1 regulates the expression of CAMs which may regulate metastatic traits. IMPLICATIONS: BAP1 mutations and increased metastasis may be due to upregulation of CAMs.


Subject(s)
Melanoma , Tumor Suppressor Proteins , Ubiquitin Thiolesterase , Uveal Neoplasms , Antigens, CD , Cadherins/genetics , Cell Adhesion Molecule-1/genetics , Humans , Melanoma/pathology , Syndecan-2 , Tumor Suppressor Proteins/genetics , Ubiquitin Thiolesterase/genetics , Uveal Neoplasms/pathology
3.
Mol Cancer Res ; 19(1): 25-37, 2021 01.
Article in English | MEDLINE | ID: mdl-33004622

ABSTRACT

Prior to metastasis, modern therapeutics and surgical intervention can provide a favorable long-term survival for patients diagnosed with many types of cancers. However, prognosis is poor for patients with metastasized disease. Melanoma is the deadliest form of skin cancer, yet in situ and localized, thin melanomas can be biopsied with little to no postsurgical follow-up. However, patients with metastatic melanoma require significant clinical involvement and have a 5-year survival of only 34% to 52%, largely dependent on the site of colonization. Melanoma metastasis is a multi-step process requiring dynamic changes in cell surface proteins regulating adhesiveness to the extracellular matrix (ECM), stroma, and other cancer cells in varied tumor microenvironments. Here we will highlight recent literature to underscore how cell adhesion molecules (CAM) contribute to melanoma disease progression and metastasis.


Subject(s)
Cell Adhesion Molecules/metabolism , Disease Progression , Humans , Neoplasm Metastasis , Prognosis
4.
J Vis Exp ; (175)2021 09 22.
Article in English | MEDLINE | ID: mdl-34633392

ABSTRACT

Brain metastasis is a serious consequence of breast cancer for women as these tumors are difficult to treat and are associated with poor clinical outcomes. Preclinical mouse models of breast cancer brain metastatic (BCBM) growth are useful but are expensive, and it is difficult to track live cells and tumor cell invasion within the brain parenchyma. Presented here is a protocol for ex vivo brain slice cultures from xenografted mice containing intracranially injected breast cancer brain-seeking clonal sublines. MDA-MB-231BR luciferase tagged cells were injected intracranially into the brains of Nu/Nu female mice, and following tumor formation, the brains were isolated, sliced, and cultured ex vivo. The tumor slices were imaged to identify tumor cells expressing luciferase and monitor their proliferation and invasion in the brain parenchyma for up to 10 days. Further, the protocol describes the use of time-lapse microscopy to image the growth and invasive behavior of the tumor cells following treatment with ionizing radiation or chemotherapy. The response of tumor cells to treatments can be visualized by live-imaging microscopy, measuring bioluminescence intensity, and performing histology on the brain slice containing BCBM cells. Thus, this ex vivo slice model may be a useful platform for rapid testing of novel therapeutic agents alone or in combination with radiation to identify drugs personalized to target an individual patient's breast cancer brain metastatic growth within the brain microenvironment.


Subject(s)
Brain Neoplasms , Nervous System Physiological Phenomena , Animals , Brain , Female , Luciferases , Mice , Mice, Nude , Tumor Microenvironment
5.
Cancer Discov ; 10(2): 254-269, 2020 02.
Article in English | MEDLINE | ID: mdl-31796433

ABSTRACT

Combinations of BRAF inhibitors and MEK inhibitors (BRAFi + MEKi) are FDA-approved to treat BRAF V600E/K-mutant melanoma. Efficacy of BRAFi + MEKi associates with cancer cell death and alterations in the tumor immune microenvironment; however, the links are poorly understood. We show that BRAFi + MEKi caused durable melanoma regression in an immune-mediated manner. BRAFi + MEKi treatment promoted cleavage of gasdermin E (GSDME) and release of HMGB1, markers of pyroptotic cell death. GSDME-deficient melanoma showed defective HMGB1 release, reduced tumor-associated T cell and activated dendritic cell infiltrates in response to BRAFi + MEKi, and more frequent tumor regrowth after drug removal. Importantly, BRAFi + MEKi-resistant disease lacked pyroptosis markers and showed decreased intratumoral T-cell infiltration but was sensitive to pyroptosis-inducing chemotherapy. These data implicate BRAFi + MEKi-induced pyroptosis in antitumor immune responses and highlight new therapeutic strategies for resistant melanoma. SIGNIFICANCE: Targeted inhibitors and immune checkpoint agents have advanced the care of patients with melanoma; however, detailed knowledge of the intersection between these two research areas is lacking. We describe a molecular mechanism of targeted inhibitor regulation of an immune-stimulatory form of cell death and provide a proof-of-principle salvage therapy concept for inhibitor-resistant melanoma.See related commentary by Smalley, p. 176.This article is highlighted in the In This Issue feature, p. 161.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Melanoma/drug therapy , Proto-Oncogene Proteins B-raf/genetics , Pyroptosis/drug effects , Skin Neoplasms/drug therapy , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor/transplantation , Dendritic Cells/drug effects , Dendritic Cells/immunology , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/immunology , Female , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Male , Melanoma/genetics , Melanoma/immunology , Mice , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Mutation , Proof of Concept Study , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/metabolism , Pyroptosis/genetics , Pyroptosis/immunology , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
6.
Cell Death Dis ; 10(4): 281, 2019 03 25.
Article in English | MEDLINE | ID: mdl-30911007

ABSTRACT

Metastatic cancer remains a clinical challenge; however, patients diagnosed prior to metastatic dissemination have a good prognosis. The transcription factor, TWIST1 has been implicated in enhancing the migration and invasion steps within the metastatic cascade, but the range of TWIST1-regulated targets is poorly described. In this study, we performed expression profiling to identify the TWIST1-regulated transcriptome of melanoma cells. Gene ontology pathway analysis revealed that TWIST1 and epithelial to mesenchymal transition (EMT) were inversely correlated with levels of cell adhesion molecule 1 (CADM1). Chromatin immunoprecipitation (ChIP) studies and promoter assays demonstrated that TWIST1 physically interacts with the CADM1 promoter, suggesting TWIST1 directly represses CADM1 levels. Increased expression of CADM1 resulted in significant inhibition of motility and invasiveness of melanoma cells. In addition, elevated CADM1 elicited caspase-independent cell death in non-adherent conditions. Expression array analysis suggests that CADM1 directed non-adherent cell death is associated with loss of mitochondrial membrane potential and subsequent failure of oxidative phosphorylation pathways. Importantly, tissue microarray analysis and clinical data from TCGA indicate that CADM1 expression is inversely associated with melanoma progression and positively correlated with better overall survival in patients. Together, these data suggest that CADM1 exerts tumor suppressive functions in melanoma by reducing invasive potential and may be considered a biomarker for favorable prognosis.


Subject(s)
Cell Adhesion Molecule-1/metabolism , Melanoma/metabolism , Melanoma/pathology , Nuclear Proteins/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Twist-Related Protein 1/metabolism , Biomarkers, Tumor , Cell Adhesion Molecule-1/genetics , Cell Line, Tumor , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Humans , Membrane Potential, Mitochondrial/genetics , Neoplasm Invasiveness/genetics , Nuclear Proteins/genetics , Prognosis , Progression-Free Survival , Promoter Regions, Genetic , Tissue Array Analysis , Transfection , Twist-Related Protein 1/genetics
7.
Pigment Cell Melanoma Res ; 32(5): 687-696, 2019 09.
Article in English | MEDLINE | ID: mdl-31063649

ABSTRACT

Epigenetic agents such as bromodomain and extra-terminal region inhibitors (BETi) slow tumor growth via tumor intrinsic alterations; however, their effects on antitumor immunity remain unclear. A recent advance is the development of next-generation BETi that are potent and display a favorable half-life. Here, we tested the BETi, PLX51107, for immune-based effects on tumor growth in BRAF V600E melanoma syngeneic models. PLX51107 delayed melanoma tumor growth and increased activated, proliferating, and functional CD8+ T cells in tumors leading to CD8+ T-cell-mediated tumor growth delay. PLX51107 decreased Cox2 expression, increased dendritic cells, and lowered PD-L1, FasL, and IDO-1 expression in the tumor microenvironment. Importantly, PLX51107 delayed the growth of tumors that progressed on anti-PD-1 therapy; a response associated with decreased Cox2 levels, decreased PD-L1 expression on non-immune cells, and increased intratumoral CD8+ T cells. Thus, next-generation BETi represent a potential first-line and secondary treatment strategy for metastatic melanoma by eliciting effects, at least in part, on antitumor CD8+ T cells.


Subject(s)
Antineoplastic Agents/pharmacology , CD8-Positive T-Lymphocytes/drug effects , Melanoma/drug therapy , Oxazoles/pharmacology , Proteins/antagonists & inhibitors , Pyridines/pharmacology , Pyrroles/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Apoptosis , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Humans , Male , Melanoma/immunology , Melanoma/pathology , Mice , Mice, Inbred C57BL , Oxazoles/therapeutic use , Pyridines/therapeutic use , Pyrroles/therapeutic use , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Cell Rep ; 25(6): 1501-1510.e3, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30404005

ABSTRACT

Expression of aberrantly spliced BRAF V600E isoforms (BRAF V600E ΔEx) mediates resistance in 13%-30% of melanoma patients progressing on RAF inhibitors. BRAF V600E ΔEx confers resistance, in part, through enhanced dimerization. Here, we uncoupled BRAF V600E ΔEx dimerization from maintenance of MEK-ERK1/2 signaling. Furthermore, we show BRAF V600E ΔEx association with its substrate, MEK, is enhanced and required for RAF inhibitor resistance. RAF inhibitor treatment increased phosphorylation at serine 729 (S729) in BRAF V600E ΔEx. Mutation of S729 to a non-phosphorylatable residue reduced BRAF V600E ΔEx-MEK interaction, reduced dimerization or oligomerization, and increased RAF inhibitor sensitivity. Conversely, mutation of the BRAF dimerization domain elicited partial effects on MEK association and RAF inhibitor sensitivity. Our data implicate BRAF S729 in resistance to RAF inhibitor and underscore the importance of substrate association with BRAF V600E ΔEx. These findings may provide opportunities to target resistance driven by aberrantly spliced forms of BRAF V600E.


Subject(s)
Drug Resistance, Neoplasm/genetics , Mitogen-Activated Protein Kinase Kinases/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , RNA Splicing/genetics , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Female , Indoles/pharmacology , Mice, Nude , Mutation/genetics , Phosphorylation/drug effects , Protein Multimerization , Serine/metabolism , Substrate Specificity , Sulfonamides/pharmacology , Vemurafenib/pharmacology
9.
Mol Cancer Ther ; 17(1): 84-95, 2018 01.
Article in English | MEDLINE | ID: mdl-29133617

ABSTRACT

FDA-approved BRAF inhibitors produce high response rates and improve overall survival in patients with BRAF V600E/K-mutant melanoma, but are linked to pathologies associated with paradoxical ERK1/2 activation in wild-type BRAF cells. To overcome this limitation, a next-generation paradox-breaking RAF inhibitor (PLX8394) has been designed. Here, we show that by using a quantitative reporter assay, PLX8394 rapidly suppressed ERK1/2 reporter activity and growth of mutant BRAF melanoma xenografts. Ex vivo treatment of xenografts and use of a patient-derived explant system (PDeX) revealed that PLX8394 suppressed ERK1/2 signaling and elicited apoptosis more effectively than the FDA-approved BRAF inhibitor, vemurafenib. Furthermore, PLX8394 was efficacious against vemurafenib-resistant BRAF splice variant-expressing tumors and reduced splice variant homodimerization. Importantly, PLX8394 did not induce paradoxical activation of ERK1/2 in wild-type BRAF cell lines or PDeX. Continued in vivo dosing of xenografts with PLX8394 led to the development of acquired resistance via ERK1/2 reactivation through heterogeneous mechanisms; however, resistant cells were found to have differential sensitivity to ERK1/2 inhibitor. These findings highlight the efficacy of a paradox-breaking selective BRAF inhibitor and the use of PDeX system to test the efficacy of therapeutic agents. Mol Cancer Ther; 17(1); 84-95. ©2017 AACR.


Subject(s)
Melanoma/drug therapy , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/genetics , Animals , Cell Line, Tumor , Female , Humans , Melanoma/pathology , Mice, Nude , Protein Kinase Inhibitors/pharmacology
10.
Clin Cancer Res ; 22(7): 1550-2, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26842234

ABSTRACT

Targeted therapies have advanced the treatment options for cutaneous melanoma, but many patients will progress on drug. Patient-derived xenografts (PDX) can be used to recapitulate therapy-resistant tumors. Furthermore, PDX modeling can be utilized in combination with targeted sequencing and phosphoproteomic platforms, providing preclinical basis for second-line targeted inhibitor strategies. See related article by Krepler et al., p. 1592.


Subject(s)
Precision Medicine , Xenograft Model Antitumor Assays , Animals , Heterografts , Humans , Melanoma , Mice , Transplantation, Heterologous
11.
Mol Cancer Res ; 12(5): 795-802, 2014 May.
Article in English | MEDLINE | ID: mdl-24520098

ABSTRACT

UNLABELLED: Resistance to RAF inhibitors such as vemurafenib and dabrafenib is a major clinical problem in the treatment of melanoma. Patients with mutant BRAF melanoma that progress on RAF inhibitors have limited treatment options, and drug removal from resistant tumors may elicit multiple effects. A frequent mechanism of resistance to RAF inhibitors is caused by expression of mutant BRAF splice variants. RAF inhibitor-resistant cell lines, generated in vivo, were tested as to whether or not mutant BRAF splice variants confer a fitness advantage in the presence of RAF inhibitor. Critically, cells expressing distinct mutant BRAF splice variants grow more efficiently in vitro and in vivo in the presence of the vemurafenib analog, PLX4720, compared with in the absence of inhibitor. PLX4720-treated BRAF splice variant-expressing cells exhibited levels of phospho-extracellular signal-regulated kinase (ERK)1/2 comparable to untreated parental cells. In addition, a reduction in phospho-ERK1/2 levels following treatment with the MEK inhibitor, trametinib (GSK1120212) phenocopied the fitness benefit provided by PLX4720. These data indicate that mutant BRAF splice variant-expressing melanoma cells are benefited by defined concentrations of RAF inhibitors. IMPLICATIONS: This study provides evidence that RAF inhibitor-resistant melanoma cells benefit from continued therapy.


Subject(s)
Indoles/pharmacology , Melanoma/drug therapy , Melanoma/genetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Sulfonamides/pharmacology , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm , Female , Humans , Melanoma/enzymology , Melanoma/pathology , Mice , Mice, Nude , Protein Isoforms , Skin Neoplasms/pathology , Xenograft Model Antitumor Assays
12.
Pigment Cell Melanoma Res ; 27(3): 479-84, 2014 May.
Article in English | MEDLINE | ID: mdl-24422853

ABSTRACT

Vemurafenib and dabrafenib block MEK-ERK1/2 signaling and cause tumor regression in the majority of advanced-stage BRAF(V600E) melanoma patients; however, acquired resistance and paradoxical signaling have driven efforts for more potent and selective RAF inhibitors. Next-generation RAF inhibitors, such as PLX7904 (PB04), effectively inhibit RAF signaling in BRAF(V600E) melanoma cells without paradoxical effects in wild-type cells. Furthermore, PLX7904 blocks the growth of vemurafenib-resistant BRAF(V600E) cells that express mutant NRAS. Acquired resistance to vemurafenib and dabrafenib is also frequently driven by expression of mutation BRAF splice variants; thus, we tested the effects of PLX7904 and its clinical analog, PLX8394 (PB03), in BRAF(V600E) splice variant-mediated vemurafenib-resistant cells. We show that paradox-breaker RAF inhibitors potently block MEK-ERK1/2 signaling, G1/S cell cycle events, survival and growth of vemurafenib/PLX4720-resistant cells harboring distinct BRAF(V600E) splice variants. These data support the further investigation of paradox-breaker RAF inhibitors as a second-line treatment option for patients failing on vemurafenib or dabrafenib.


Subject(s)
Melanoma/enzymology , Neoplasm Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , raf Kinases/antagonists & inhibitors , Cell Cycle/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Enzyme Activation/drug effects , GTP Phosphohydrolases/antagonists & inhibitors , GTP Phosphohydrolases/genetics , Genes, ras , Humans , Imidazoles/pharmacology , Indoles/pharmacology , MAP Kinase Signaling System/drug effects , Melanoma/pathology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mutation, Missense , Neoplasm Proteins/genetics , Oximes/pharmacology , Phosphorylation/drug effects , Protein Isoforms/antagonists & inhibitors , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins B-raf/genetics , Signal Transduction/drug effects , Sulfonamides/pharmacology , Tumor Stem Cell Assay , Vemurafenib
13.
Cancer Res ; 74(15): 4122-32, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-25035390

ABSTRACT

ERBB3/HER3 expression and signaling are upregulated in mutant BRAF melanoma as an adaptive, prosurvival response to FDA-approved RAF inhibitors. Because compensatory ERBB3 signaling counteracts the effects of RAF inhibitors, cotargeting ERBB3 may increase the efficacy of RAF inhibitors in mutant BRAF models of melanoma. Here, we corroborate this concept by showing that the ERBB3 function-blocking monoclonal antibody huHER3-8 can inhibit neuregulin-1 activation of ERBB3 and downstream signaling in RAF-inhibited melanoma cells. Targeting mutant BRAF in combination with huHER3-8 decreased cell proliferation and increased cell death in vitro, and decreased tumor burden in vivo, compared with targeting either mutant BRAF or ERBB3 alone. Furthermore, the likelihood of a durable tumor response in vivo was increased when huHER3-8 was combined with RAF inhibitor PLX4720. Together, these results offer a preclinical proof of concept for the application of ERBB3-neutralizing antibodies to enhance the efficacy of RAF inhibitors in melanoma to delay or prevent tumor regrowth. As ERBB3 is often upregulated in response to other kinase-targeted therapeutics, these findings may have implications for other cancers as well.


Subject(s)
Antibodies, Monoclonal/pharmacology , Melanoma/drug therapy , Melanoma/enzymology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Receptor, ErbB-3/antagonists & inhibitors , Receptor, ErbB-3/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Cell Growth Processes/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Female , Heterografts , Humans , Indoles/administration & dosage , Indoles/pharmacology , Melanoma/pathology , Mice , Mice, Nude , Protein Kinase Inhibitors/administration & dosage , Pyridones/pharmacology , Pyrimidinones/pharmacology , Signal Transduction , Sulfonamides/administration & dosage , Sulfonamides/pharmacology , Vemurafenib
14.
J Invest Dermatol ; 133(8): 1928-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23856932

ABSTRACT

Despite recent advancements in the treatment of late-stage mutant BRAF (V600E/K) melanomas, a major hurdle continues to be acquired resistance to BRAF inhibitors such as vemurafenib. The mechanisms for resistance have proven to be heterogeneous, emphasizing the need to use broad therapeutic approaches. In this issue, the study "Stat3-targeted therapies overcome the acquired resistance to vemurafenib in melanomas" by Liu et al. proposes that signal transducer and activator of transcription 3 (STAT3)-paired box 3 (PAX3) signaling may be a mechanism that is used by melanomas to resist RAF inhibitors.


Subject(s)
Drug Resistance, Neoplasm/physiology , Indoles/pharmacology , Keratinocytes/drug effects , Melanoma/drug therapy , STAT3 Transcription Factor/antagonists & inhibitors , Skin Neoplasms/drug therapy , Sulfonamides/pharmacology , Humans , Male , Vemurafenib
15.
Cancer Res ; 73(23): 7101-10, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24121492

ABSTRACT

Activation of the ERK1/2 mitogen-activated protein kinases (MAPK) confers resistance to the RAF inhibitors vemurafenib and dabrafenib in mutant BRAF-driven melanomas. Methods to understand how resistance develops are important to optimize the clinical use of RAF inhibitors in patients. Here, we report the development of a novel ERK1/2 reporter system that provides a noninvasive, quantitative, and temporal analysis of RAF inhibitor efficacy in vivo. Use of this system revealed heterogeneity in the level of ERK1/2 reactivation associated with acquired resistance to RAF inhibition. We identified several distinct novel and known molecular changes in resistant tumors emerging from treatment-naïve cell populations including BRAF V600E variants and HRAS mutation, both of which were required and sufficient for ERK1/2 reactivation and drug resistance. Our work offers an advance in understanding RAF inhibitor resistance and the heterogeneity in resistance mechanisms, which emerge from a malignant cell population.


Subject(s)
Drug Resistance, Neoplasm/genetics , Genes, Reporter , Genetic Heterogeneity , Indoles/therapeutic use , MAP Kinase Signaling System/genetics , Neoplasms/genetics , Sulfonamides/therapeutic use , Animals , Enzyme Activation/genetics , Female , Genes, Reporter/physiology , Genetic Heterogeneity/drug effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Mice , Mice, Nude , Mice, Transgenic , Neoplasms/drug therapy , Neoplasms/metabolism , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Selection, Genetic/physiology , Transfection , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
16.
Sci Signal ; 6(304): ra104, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24300896

ABSTRACT

Activation of vascular endothelial growth factor receptor-2 (VEGFR-2), an endothelial cell receptor tyrosine kinase, promotes tumor angiogenesis and ocular neovascularization. We report the methylation of VEGFR-2 at multiple Lys and Arg residues, including Lys(1041), a residue that is proximal to the activation loop of the kinase domain. Methylation of VEGFR-2 was independent of ligand binding and was not regulated by ligand stimulation. Methylation of Lys(1041) enhanced tyrosine phosphorylation and kinase activity in response to ligands. Additionally, interfering with the methylation of VEGFR-2 by pharmacological inhibition or by site-directed mutagenesis revealed that methylation of Lys(1041) was required for VEGFR-2-mediated angiogenesis in zebrafish and tumor growth in mice. We propose that methylation of Lys(1041) promotes the activation of VEGFR-2 and that similar posttranslational modification could also regulate the activity of other receptor tyrosine kinases.


Subject(s)
Lysine/metabolism , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Neovascularization, Pathologic/metabolism , Protein Processing, Post-Translational , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Cell Line, Tumor , HEK293 Cells , Heterografts , Humans , Lysine/genetics , Methylation , Mice , Neoplasm Proteins/genetics , Neoplasm Transplantation , Neoplasms/genetics , Neoplasms/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Vascular Endothelial Growth Factor Receptor-2 , Zebrafish/genetics , Zebrafish Proteins/genetics
17.
Pigment Cell Melanoma Res ; 30(2): 177-178, 2017 03.
Article in English | MEDLINE | ID: mdl-27801548
SELECTION OF CITATIONS
SEARCH DETAIL