Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Pigment Cell Melanoma Res ; 36(6): 588-593, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37819763

ABSTRACT

The Society for Melanoma Research (SMR) was created 20 years ago and has unequivocally contributed to the vast progress of the field, particularly for the treatment of melanoma patients with metastatic disease by facilitating synergistic collaborations between clinicians, researchers at the bench, and industry. In commemoration of the 20th anniversary of the first SMR International Congress (held in 2003 in Philadelphia), we look to the future by highlighting the perspectives of the next generation of rising stars, medical, and graduate students across six continents.


Subject(s)
Melanoma , Humans , Melanoma/therapy , Melanoma/pathology
2.
Pigment Cell Melanoma Res ; 36(6): 594-601, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37819777

ABSTRACT

Upon the 20th Anniversary of the Society for Melanoma Research, we highlight the perspectives of patients aiming to help improve future experiences, outcomes, and their quality of life over the next 20 years. Five melanoma patients generously shared their inspiring and enlightening stories of diagnosis, treatment, and outcomes. Many patients had excellent medical teams that synergistically worked together to provide an accurate diagnosis, effective treatment options, and supportive care. However, it is clear that health inequities persist in communities where people of color are predominant, affecting early detection, patient experience, and outcomes. These stories shed light on the unique challenges faced by patients and how the lack of melanoma awareness and adequate resources, especially in communities of color or low socioeconomic status, can contribute to disparate outcomes in melanoma care. We expect that these stories will raise awareness about the progress in melanoma treatment but also the existent disparities in melanoma diagnosis and treatment and the importance of early detection and prevention.


Subject(s)
Melanoma , Quality of Life , Humans , Melanoma/diagnosis , Melanoma/therapy
4.
Pigment Cell Melanoma Res ; 36(6): 576-582, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37759408

ABSTRACT

To commemorate the 20th Anniversary of the Society of Melanoma Research and the first International Melanoma Research Congress held in June of 2003, we have described in brief, how the Society for Melanoma Research (SMR) began, the purpose, goals, and governance of the SMR, and how the society has evolved to support new melanoma researchers. In celebration of the immense progress in treating melanoma patients over the last 20 years and the impact of the SMR on these advances, we have highlighted memories and insight from early SMR members and founders.


Subject(s)
Friends , Melanoma , Humans , Melanoma/therapy , Societies, Medical
6.
Pigment Cell Melanoma Res ; 36(5): 441-447, 2023 09.
Article in English | MEDLINE | ID: mdl-37093838

ABSTRACT

The inaugural Diversity and Inclusion in Science Session was held during the 2021 Society for Melanoma Research (SMR) congress. The goal of the session was to discuss diversity, equity, and inclusion in the melanoma research community and strategies to promote the advancement of underrepresented melanoma researchers. An international survey was conducted to assess the diversity, equity, and inclusion (DEI) climate among researchers and clinicians within the Society for Melanoma Research (SMR). The findings suggest there are feelings and experiences of inequity, bias, and harassment within the melanoma community that correlate with one's gender, ethnic/racial group, and/or geographic location. Notably, significant reports of inequity in opportunity, discrimination, and sexual harassment demonstrate there is much work remaining to ensure all scientists in our community experience an academic workplace culture built on mutual respect, fair access, inclusion, and equitable opportunity.


Subject(s)
Diversity, Equity, Inclusion , Melanoma , Humans
7.
bioRxiv ; 2023 Jan 21.
Article in English | MEDLINE | ID: mdl-36711814

ABSTRACT

Resistance to combination BRAF/MEK inhibitor (BRAFi/MEKi) therapy arises in nearly every patient with BRAFV600E/K melanoma, despite promising initial responses. Achieving cures in this expanding BRAFi/MEKi-resistant cohort represents one of the greatest challenges to the field; few experience additional durable benefit from immunotherapy and no alternative therapies exist. To better personalize therapy in cancer patients to address therapy relapse, umbrella trials have been initiated whereby genomic sequencing of a panel of potentially actionable targets guide therapy selection for patients; however, the superior efficacy of such approaches remains to be seen. We here test the robustness of the umbrella trial rationale by analyzing relationships between genomic status of a gene and the downstream consequences at the protein level of related pathway, which find poor relationships between mutations, copy number amplification, and protein level. To profile candidate therapeutic strategies that may offer clinical benefit in the context of acquired BRAFi/MEKi resistance, we established a repository of patient-derived xenograft models from heavily pretreated patients with resistance to BRAFi/MEKi and/or immunotherapy (R-PDX). With these R-PDXs, we executed in vivo compound repurposing screens using 11 FDA-approved agents from an NCI-portfolio with pan-RTK, non-RTK and/or PI3K-mTOR specificity. We identify dasatinib as capable of restoring BRAFi/MEKi antitumor efficacy in ~70% of R-PDX tested. A systems-biology analysis indicates elevated baseline protein expression of canonical drivers of therapy resistance (e.g., AXL, YAP, HSP70, phospho-AKT) as predictive of MAPKi/dasatinib sensitivity. We therefore propose that dasatinib-based MAPKi therapy may restore antitumor efficacy in patients that have relapsed to standard-of-care therapy by broadly targeting proteins critical in melanoma therapy escape. Further, we submit that this experimental PDX paradigm could potentially improve preclinical evaluation of therapeutic modalities and augment our ability to identify biomarker-defined patient subsets that may respond to a given clinical trial.

8.
Oncogene ; 40(37): 5590-5599, 2021 09.
Article in English | MEDLINE | ID: mdl-34304249

ABSTRACT

Targeting MAPK pathway using a combination of BRAF and MEK inhibitors is an efficient strategy to treat melanoma harboring BRAF-mutation. The development of acquired resistance is inevitable due to the signaling pathway rewiring. Combining western blotting, immunohistochemistry, and reverse phase protein array (RPPA), we aim to understanding the role of the mTORC1 signaling pathway, a center node of intracellular signaling network, in mediating drug resistance of BRAF-mutant melanoma to the combination of BRAF inhibitor (BRAFi) and MEK inhibitor (MEKi) therapy. The mTORC1 signaling pathway is initially suppressed by BRAFi and MEKi combination in melanoma but rebounds overtime after tumors acquire resistance to the combination therapy (CR) as assayed in cultured cells and PDX models. In vitro experiments showed that a subset of CR melanoma cells was sensitive to mTORC1 inhibition. The mTOR inhibitors, rapamycin and NVP-BEZ235, induced cell cycle arrest and apoptosis in CR cell lines. As a proof-of-principle, we demonstrated that rapamycin and NVP-BEZ235 treatment reduced tumor growth in CR xenograft models. Mechanistically, AKT or ERK contributes to the activation of mTORC1 in CR cells, depending on PTEN status of these cells. Our study reveals that mTOR activation is essential for drug resistance of melanoma to MAPK inhibitors, and provides insight into the rewiring of the signaling networks in CR melanoma.


Subject(s)
Proto-Oncogene Proteins B-raf , TOR Serine-Threonine Kinases , Humans
9.
Cancer Cell ; 39(5): 610-631, 2021 05 10.
Article in English | MEDLINE | ID: mdl-33545064

ABSTRACT

There is a lack of appropriate melanoma models that can be used to evaluate the efficacy of novel therapeutic modalities. Here, we discuss the current state of the art of melanoma models including genetically engineered mouse, patient-derived xenograft, zebrafish, and ex vivo and in vitro models. We also identify five major challenges that can be addressed using such models, including metastasis and tumor dormancy, drug resistance, the melanoma immune response, and the impact of aging and environmental exposures on melanoma progression and drug resistance. Additionally, we discuss the opportunity for building models for rare subtypes of melanomas, which represent an unmet critical need. Finally, we identify key recommendations for melanoma models that may improve accuracy of preclinical testing and predict efficacy in clinical trials, to help usher in the next generation of melanoma therapies.


Subject(s)
Disease Models, Animal , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Tumor Microenvironment/immunology , Animals , Humans , Immunity/immunology , Immunotherapy/methods , Melanoma/pathology , Skin Neoplasms/pathology
10.
Oncogene ; 39(36): 5811-5824, 2020 09.
Article in English | MEDLINE | ID: mdl-32733068

ABSTRACT

Telomere maintenance via telomerase reactivation is a nearly universal hallmark of cancer cells which enables replicative immortality. In contrast, telomerase activity is silenced in most adult somatic cells. Thus, telomerase represents an attractive target for highly selective cancer therapeutics. However, development of telomerase inhibitors has been challenging and thus far there are no clinically approved strategies exploiting this cancer target. The discovery of prevalent mutations in the TERT promoter region in many cancers and recent advances in telomerase biology has led to a renewed interest in targeting this enzyme. Here we discuss recent efforts targeting telomerase, including immunotherapies and direct telomerase inhibitors, as well as emerging approaches such as targeting TERT gene expression driven by TERT promoter mutations. We also address some of the challenges to telomerase-directed therapies including potential therapeutic resistance and considerations for future therapeutic applications and translation into the clinical setting. Although much work remains to be done, effective strategies targeting telomerase will have a transformative impact for cancer therapy and the prospect of clinically effective drugs is boosted by recent advances in structural models of human telomerase.


Subject(s)
Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/enzymology , Telomerase/antagonists & inhibitors , Animals , Biomarkers, Tumor , Clinical Studies as Topic , Drug Evaluation, Preclinical , Gene Expression Regulation, Neoplastic/drug effects , Humans , Neoplasms/diagnosis , Neoplasms/etiology , Telomere/genetics , Telomere/metabolism , Telomere Homeostasis , Treatment Outcome
11.
Pigment Cell Melanoma Res ; 33(2): 372-377, 2020 03.
Article in English | MEDLINE | ID: mdl-31696640

ABSTRACT

Bromodomain and extra-terminal inhibitors (BETi) delay tumor growth, in part, through tumor cell intrinsic alterations and initiation of anti-tumor CD8+ T-cell responses. By contrast, BETi effects on pro-tumoral immune responses remain unclear. Here, we show that the next-generation BETi, PLX51107, delayed tumor growth to differing degrees in Braf V600E melanoma syngeneic mouse models. These differential responses were associated with the influx of tumor-associated macrophages during BETi treatment. Tumors that were poorly responsive to PLX51107 showed increased influx of colony-stimulating factor-1 receptor (CSF-1R)-positive tumor-associated macrophages. We depleted CSF-1R+ tumor-associated macrophages with the CSF-1R inhibitor, PLX3397, in combination with PLX51107. Treatment with PLX3397 enhanced the efficacy of PLX51107 in poorly responsive Braf V600E syngeneic melanomas in vivo. These findings suggest that tumor-associated macrophage accumulation limits BETi efficacy and that co-treatment with PLX3397 can improve response to PLX51107, offering a potential novel combination therapy for metastatic melanoma patients.


Subject(s)
Aminopyridines/pharmacology , Macrophages/pathology , Melanoma/pathology , Proteins/antagonists & inhibitors , Pyrroles/pharmacology , Skin Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Macrophages/drug effects , Male , Mice, Inbred C57BL , Proteins/metabolism , Proto-Oncogene Proteins B-raf/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Treatment Outcome
12.
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
13.
Cancer Res ; 78(19): 5694-5705, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30115697

ABSTRACT

The tumor suppressor TP53 is the most frequently mutated gene in human cancer and serves to restrict tumor initiation and progression. Single-nucleotide polymorphisms (SNP) in TP53 and p53 pathway genes can have a marked impact on p53 tumor suppressor function, and some have been associated with increased cancer risk and impaired response to therapy. Approximately 6% of Africans and 1% of African Americans express a p53 allele with a serine instead of proline at position 47 (Pro47Ser). This SNP impairs p53-mediated apoptosis in response to radiation and genotoxic agents and is associated with increased cancer risk in humans and in a mouse model. In this study, we compared the ability of wild-type (WT) and S47 p53 to suppress tumor development and respond to therapy. Our goal was to find therapeutic compounds that are more, not less, efficacious in S47 tumors. We identified the superior efficacy of two agents, cisplatin and BET inhibitors, on S47 tumors compared with WT. Cisplatin caused dramatic decreases in the progression of S47 tumors by activating the p53/PIN1 axis to drive the mitochondrial cell death program. These findings serve as important proof of principle that chemotherapy can be tailored to p53 genotype.Significance: A rare African-derived radioresistant p53 SNP provides proof of principle that chemotherapy can be tailored to TP53 genotype. Cancer Res; 78(19); 5694-705. ©2018 AACR.


Subject(s)
Polymorphism, Single Nucleotide , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Africa , Black or African American/genetics , Alleles , Animals , Apoptosis , Black People/genetics , Cell Death/drug effects , Cell Line, Tumor , Cell Transformation, Neoplastic , Cisplatin/pharmacology , Disease Progression , Fibroblasts/metabolism , Genotype , Humans , Mice , Mitochondria/metabolism , Mutation/drug effects , Neoplasm Transplantation , Pharmacogenetics , Precision Medicine , Risk
14.
EMBO Mol Med ; 10(5)2018 05.
Article in English | MEDLINE | ID: mdl-29650805

ABSTRACT

Despite novel therapies for melanoma, drug resistance remains a significant hurdle to achieving optimal responses. NRAS-mutant melanoma is an archetype of therapeutic challenges in the field, which we used to test drug combinations to avert drug resistance. We show that BET proteins are overexpressed in NRAS-mutant melanoma and that high levels of the BET family member BRD4 are associated with poor patient survival. Combining BET and MEK inhibitors synergistically curbed the growth of NRAS-mutant melanoma and prolonged the survival of mice bearing tumors refractory to MAPK inhibitors and immunotherapy. Transcriptomic and proteomic analysis revealed that combining BET and MEK inhibitors mitigates a MAPK and checkpoint inhibitor resistance transcriptional signature, downregulates the transcription factor TCF19, and induces apoptosis. Our studies demonstrate that co-targeting MEK and BET can offset therapy resistance, offering a salvage strategy for melanomas with no other therapeutic options, and possibly other treatment-resistant tumor types.


Subject(s)
Drug Resistance, Neoplasm/drug effects , MAP Kinase Kinase 1/antagonists & inhibitors , Melanoma/drug therapy , Proteins/antagonists & inhibitors , Skin Neoplasms/drug therapy , Xenograft Model Antitumor Assays , Acetanilides/pharmacology , Animals , Cell Cycle Proteins , Cell Line, Tumor , Gene Expression Profiling/methods , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , MAP Kinase Kinase 1/metabolism , MAP Kinase Signaling System/drug effects , Melanoma/genetics , Melanoma/metabolism , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Kinase Inhibitors/pharmacology , Proteins/metabolism , Proteomics/methods , Salvage Therapy/methods , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
15.
Oncogene ; 37(30): 4058-4072, 2018 07.
Article in English | MEDLINE | ID: mdl-29695835

ABSTRACT

Targeting RAS is one of the greatest challenges in cancer therapy. Oncogenic mutations in NRAS are present in over 25% of melanomas and patients whose tumors harbor NRAS mutations have limited therapeutic options and poor prognosis. Thus far, there are no clinical agents available to effectively target NRAS or any other RAS oncogene. An alternative approach is to identify and target critical tumor vulnerabilities or non-oncogene addictions that are essential for tumor survival. We investigated the consequences of NRAS blockade in NRAS-mutant melanoma and show that decreased expression of the telomerase catalytic subunit, TERT, is a major consequence. TERT silencing or treatment of NRAS-mutant melanoma with the telomerase-dependent telomere uncapping agent, 6-thio-2'-deoxyguanosine (6-thio-dG), led to rapid cell death, along with evidence of both telomeric and non-telomeric DNA damage, increased ROS levels, and upregulation of a mitochondrial antioxidant adaptive response. Combining 6-thio-dG with the mitochondrial inhibitor Gamitrinib attenuated this adaptive response and more effectively suppressed NRAS-mutant melanoma. Our study uncovers a robust dependency of NRAS-mutant melanoma on TERT, and provides proof-of-principle for a new combination strategy to combat this class of tumors, which could be expanded to other tumor types.


Subject(s)
GTP Phosphohydrolases/genetics , Melanoma/genetics , Membrane Proteins/genetics , Mutation/genetics , Telomerase/genetics , Animals , Antioxidants/metabolism , Cell Line , Cell Line, Tumor , DNA Damage/genetics , Female , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mitochondria/genetics , Reactive Oxygen Species/metabolism , Up-Regulation/genetics
16.
Mol Cancer ; 16(1): 85, 2017 04 28.
Article in English | MEDLINE | ID: mdl-28454577

ABSTRACT

BACKGROUND: The BRAF protein kinase is widely studied as a cancer driver and therapeutic target. However, the regulation of its expression is not completely understood. RESULTS: Taking advantage of the RNA-seq data of more than 4800 patients belonging to 9 different cancer types, we show that BRAF mRNA exists as a pool of 3 isoforms (reference BRAF, BRAF-X1, and BRAF-X2) that differ in the last part of their coding sequences, as well as in the length (BRAF-ref: 76 nt; BRAF-X1 and BRAF-X2: up to 7 kb) and in the sequence of their 3'UTRs. The expression levels of BRAF-ref and BRAF-X1/X2 are inversely correlated, while the most prevalent among the three isoforms varies from cancer type to cancer type. In melanoma cells, the X1 isoform is expressed at the highest level in both therapy-naïve cells and cells with acquired resistance to vemurafenib driven by BRAF gene amplification or expression of the Δ[3-10] splicing variant. In addition to the BRAF-ref protein, the BRAF-X1 protein (the full length as well as the Δ[3-10] variant) is also translated. The expression levels of the BRAF-ref and BRAF-X1 proteins are similar, and together they account for BRAF functional activities. In contrast, the endogenous BRAF-X2 protein is hard to detect because the C-terminal domain is selectively recognized by the ubiquitin-proteasome pathway and targeted for degradation. CONCLUSIONS: By shedding light on the repertoire of BRAF mRNA and protein variants, and on the complex regulation of their expression, our work paves the way to a deeper understanding of a crucially important player in human cancer and to a more informed development of new therapeutic strategies.


Subject(s)
Melanoma/genetics , Neoplasms/genetics , Protein Isoforms/genetics , Proto-Oncogene Proteins B-raf/genetics , Alternative Splicing/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Exons/genetics , Gene Expression Regulation, Neoplastic/drug effects , High-Throughput Nucleotide Sequencing , Humans , Indoles/administration & dosage , Melanoma/drug therapy , Melanoma/pathology , Neoplasms/drug therapy , Neoplasms/pathology , RNA, Messenger/genetics , Sulfonamides/administration & dosage , Vemurafenib
17.
18.
Oncotarget ; 7(44): 71211-71222, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27655717

ABSTRACT

The discovery of activating BRAF mutations in approximately 50% of melanomas has led to the development of MAPK pathway inhibitors, which have transformed melanoma therapy. However, not all BRAF-V600E melanomas respond to MAPK inhibition. Therefore, it is important to understand why tumors with the same oncogenic driver have variable responses to MAPK inhibitors. Here, we show that concurrent loss of PTEN and activation of the Notch pathway is associated with poor response to the ERK inhibitor SCH772984, and that co-inhibition of Notch and ERK decreased viability in BRAF-V600E melanomas. Additionally, patients with low PTEN and Notch activation had significantly shorter progression free survival when treated with BRAF inhibitors. Our studies provide a rationale to further develop combination strategies with Notch antagonists to maximize the efficacy of MAPK inhibition in melanoma. Our findings should prompt the evaluation of combinations co-targeting MAPK/ERK and Notch as a strategy to improve current therapies and warrant further evaluation of co-occurrence of aberrant PTEN and Notch activation as predictive markers of response to therapy.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Indazoles/therapeutic use , Melanoma/drug therapy , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/genetics , Receptors, Notch/antagonists & inhibitors , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Animals , Cell Line, Tumor , Humans , Melanoma/genetics , Melanoma/pathology , Mice , PTEN Phosphohydrolase/physiology , Phosphatidylinositol 3-Kinases/physiology , Receptors, Notch/physiology
19.
ACS Chem Biol ; 11(10): 2876-2888, 2016 10 21.
Article in English | MEDLINE | ID: mdl-27571413

ABSTRACT

The BRAF kinase, within the mitogen activated protein kinase (MAPK) signaling pathway, harbors activating mutations in about half of melanomas and to a significant extent in many other cancers. A single valine to glutamic acid substitution at residue 600 (BRAFV600E) accounts for about 90% of these activating mutations. While BRAFV600E-selective small molecule inhibitors, such as debrafenib and vemurafenib, have shown therapeutic benefit, almost all patients develop resistance. Resistance often arises through reactivation of the MAPK pathway, typically through mutation of upstream RAS, downstream MEK, or splicing variants. RAF kinases signal as homo- and heterodimers, and another complication associated with small molecule BRAFV600E inhibition is drug-induced allosteric activation of a wild-type RAF subunit (BRAF or CRAF) of the kinase dimer, a process called "transactivation" or "paradoxical activation." Here, we used BRAFV600E and vemurafenib as a model system to develop chemically linked kinase inhibitors to lock RAF dimers in an inactive conformation that cannot undergo transactivation. This structure-based design effort resulted in the development of Vem-BisAmide-2, a compound containing two vemurafenib molecules connected by a bis amide linker. We show that Vem-BisAmide-2 has comparable inhibitory potency as vemurafenib to BRAFV600E both in vitro and in cells but promotes an inactive dimeric BRAFV600E conformation unable to undergo transactivation. The crystal structure of a BRAFV600E/Vem-BisAmide-2 complex and associated biochemical studies reveal the molecular basis for how Vem-BisAmide-2 mediates selectivity for an inactive over an active dimeric BRAFV600E conformation. These studies have implications for targeting BRAFV600E/RAF heterodimers and other kinase dimers for therapy.


Subject(s)
Indoles/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Sulfonamides/pharmacology , Cell Line, Tumor , Crystallography, X-Ray , Dimerization , Enzyme-Linked Immunosorbent Assay , Humans , Indoles/chemistry , Melanoma/pathology , Molecular Structure , Protein Conformation , Proto-Oncogene Proteins B-raf/chemistry , Solutions , Sulfonamides/chemistry , Vemurafenib
SELECTION OF CITATIONS
SEARCH DETAIL
...