Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.692
Filter
1.
Life Sci Alliance ; 7(8)2024 Aug.
Article in English | MEDLINE | ID: mdl-38839106

ABSTRACT

Targeted therapies against mutant BRAF are effectively used in combination with MEK inhibitors (MEKi) to treat advanced melanoma. However, treatment success is affected by resistance and adverse events (AEs). Approved BRAF inhibitors (BRAFi) show high levels of target promiscuity, which can contribute to these effects. The blood vessel lining is in direct contact with high plasma concentrations of BRAFi, but effects of the inhibitors in this cell type are unknown. Hence, we aimed to characterize responses to approved BRAFi for melanoma in the vascular endothelium. We showed that clinically approved BRAFi induced a paradoxical activation of endothelial MAPK signaling. Moreover, phosphoproteomics revealed distinct sets of off-targets per inhibitor. Endothelial barrier function and junction integrity were impaired upon treatment with vemurafenib and the next-generation dimerization inhibitor PLX8394, but not with dabrafenib or encorafenib. Together, these findings provide insights into the surprisingly distinct side effects of BRAFi on endothelial signaling and functionality. Better understanding of off-target effects could help to identify molecular mechanisms behind AEs and guide the continued development of therapies for BRAF-mutant melanoma.


Subject(s)
Melanoma , Protein Kinase Inhibitors , Proto-Oncogene Proteins B-raf , Signal Transduction , Vemurafenib , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/metabolism , Humans , Protein Kinase Inhibitors/pharmacology , Melanoma/drug therapy , Melanoma/metabolism , Signal Transduction/drug effects , Vemurafenib/pharmacology , Oximes/pharmacology , Sulfonamides/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Imidazoles/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , MAP Kinase Signaling System/drug effects , Carbamates/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Cell Line, Tumor , Mutation
2.
Eur J Cancer ; 204: 114073, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38723373

ABSTRACT

BACKGROUND: Treatment with encorafenib plus binimetinib and encorafenib monotherapy is associated with improved progression-free survival (PFS) and overall survival (OS) compared with vemurafenib in patients with BRAF V600E/K-mutant metastatic melanoma. We report results from the 7-year analysis of COLUMBUS part 1 (NCT01909453) at 99.7 months (median duration between randomization and data cutoff). METHODS: 577 patients with locally advanced unresectable or metastatic BRAF V600E/K-mutant melanoma who were treatment-naive or progressed after first-line immunotherapy were randomized 1:1:1 to encorafenib 450 mg once daily (QD) plus binimetinib 45 mg twice daily (BID) (n = 192), vemurafenib 960 mg BID (n = 191), or encorafenib monotherapy 300 mg QD (n = 194). No prior BRAF/MEK inhibitor was allowed. RESULTS: Seven-year PFS and OS rates (95 % CI) were 21.2 % (14.7-28.4 %) and 27.4 % (21.2-33.9%) in the encorafenib plus binimetinib arm and 6.4 % (2.1-14.0 %) and 18.2 % (12.8-24.3 %) in the vemurafenib arm, respectively. Median melanoma-specific survival (95 % CI) was 36.8 months (27.7-51.5 months) in the encorafenib plus binimetinib arm and 19.3 months (14.8-25.9 months) in the vemurafenib arm. Thirty-four long-term responders (complete/partial response ongoing at 7 years) were identified across arms. CONCLUSIONS: This is the longest follow-up from a phase III trial of BRAF/MEK inhibitor combination in BRAF V600E/K-mutant metastatic melanoma. Safety results were consistent with the known tolerability profile of encorafenib plus binimetinib. Results support the long-term efficacy and known safety of encorafenib plus binimetinib in this population and provide new insights on long-term responders. Interactive data visualization is available at the COLUMBUS dashboard (https://clinical-trials.dimensions.ai/columbus7/).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Benzimidazoles , Carbamates , Melanoma , Mutation , Proto-Oncogene Proteins B-raf , Sulfonamides , Vemurafenib , Humans , Melanoma/drug therapy , Melanoma/genetics , Melanoma/mortality , Carbamates/administration & dosage , Carbamates/adverse effects , Proto-Oncogene Proteins B-raf/genetics , Sulfonamides/administration & dosage , Sulfonamides/adverse effects , Benzimidazoles/administration & dosage , Benzimidazoles/adverse effects , Benzimidazoles/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Male , Female , Vemurafenib/administration & dosage , Vemurafenib/adverse effects , Middle Aged , Aged , Adult , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Skin Neoplasms/mortality , Aged, 80 and over , Progression-Free Survival , Young Adult
3.
Cell Mol Life Sci ; 81(1): 226, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38775844

ABSTRACT

Vemurafenib has been used as first-line therapy for unresectable or metastatic melanoma with BRAFV600E mutation. However, overall survival is still limited due to treatment resistance after about one year. Therefore, identifying new therapeutic targets for melanoma is crucial for improving clinical outcomes. In the present study, we found that lowering intracellular cholesterol by knocking down DHCR24, the limiting synthetase, impaired tumor cell proliferation and migration and abrogated the ability to xenotransplant tumors. More importantly, administration of DHCR24 or cholesterol mediated resistance to vemurafenib and promoted the growth of melanoma spheroids. Mechanistically, we identified that 27-hydroxycholesterol (27HC), a primary metabolite of cholesterol synthesized by the enzyme cytochrome P450 27A1 (CYP27A1), reproduces the phenotypes induced by DHCR24 or cholesterol administration and activates Rap1-PI3K/AKT signaling. Accordingly, CYP27A1 is highly expressed in melanoma patients and upregulated by DHCR24 induction. Dafadine-A, a CYP27A1 inhibitor, attenuates cholesterol-induced growth of melanoma spheroids and abrogates the resistance property of vemurafenib-resistant melanoma cells. Finally, we confirmed that the effects of cholesterol on melanoma resistance require its metabolite 27HC through CYP27A1 catalysis, and that 27HC further upregulates Rap1A/Rap1B expression and increases AKT phosphorylation. Thus, our results suggest that targeting 27HC may be a useful strategy to overcome treatment resistance in metastatic melanoma.


Subject(s)
Cell Proliferation , Cholestanetriol 26-Monooxygenase , Cholesterol , Hydroxycholesterols , Melanoma , Neoplastic Stem Cells , Vemurafenib , Vemurafenib/pharmacology , Vemurafenib/therapeutic use , Humans , Melanoma/drug therapy , Melanoma/pathology , Melanoma/metabolism , Melanoma/genetics , Hydroxycholesterols/metabolism , Hydroxycholesterols/pharmacology , Animals , Cell Proliferation/drug effects , Cholestanetriol 26-Monooxygenase/metabolism , Cholestanetriol 26-Monooxygenase/genetics , Cholesterol/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Cell Line, Tumor , Mice , Drug Resistance, Neoplasm/drug effects , Signal Transduction/drug effects , Cell Movement/drug effects , Xenograft Model Antitumor Assays
4.
Cell Commun Signal ; 22(1): 282, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38778340

ABSTRACT

Extracellular vesicles (EVs) constitute a vital component of intercellular communication, exerting significant influence on metastasis formation and drug resistance mechanisms. Malignant melanoma (MM) is one of the deadliest forms of skin cancers, because of its high metastatic potential and often acquired resistance to oncotherapies. The prevalence of BRAF mutations in MM underscores the importance of BRAF-targeted therapies, such as vemurafenib and dabrafenib, alone or in combination with the MEK inhibitor, trametinib. This study aimed to elucidate the involvement of EVs in MM progression and ascertain whether EV-mediated metastasis promotion persists during single agent BRAF (vemurafenib, dabrafenib), or MEK (trametinib) and combined BRAF/MEK (dabrafenib/trametinib) inhibition.Using five pairs of syngeneic melanoma cell lines, we assessed the impact of EVs - isolated from their respective supernatants - on melanoma cell proliferation and migration. Cell viability and spheroid growth assays were employed to evaluate proliferation, while migration was analyzed through mean squared displacement (MSD) and total traveled distance (TTD) measurements derived from video microscopy and single-cell tracking.Our results indicate that while EV treatments had remarkable promoting effect on cell migration, they exerted only a modest effect on cell proliferation and spheroid growth. Notably, EVs demonstrated the ability to mitigate the inhibitory effects of BRAF inhibitors, albeit they were ineffective against a MEK inhibitor and the combination of BRAF/MEK inhibitors. In summary, our findings contribute to the understanding of the intricate role played by EVs in tumor progression, metastasis, and drug resistance in MM.


Subject(s)
Cell Movement , Extracellular Vesicles , Melanoma , Protein Kinase Inhibitors , Proto-Oncogene Proteins B-raf , Melanoma/pathology , Melanoma/drug therapy , Melanoma/metabolism , Extracellular Vesicles/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/genetics , Humans , Cell Movement/drug effects , Cell Line, Tumor , Protein Kinase Inhibitors/pharmacology , Cell Proliferation/drug effects , Vemurafenib/pharmacology , Pyrimidinones/pharmacology , Pyridones/pharmacology , Pyridones/therapeutic use , Imidazoles/pharmacology , Oximes/pharmacology
5.
Biochem Pharmacol ; 224: 116252, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38701866

ABSTRACT

The mitogen-activated protein kinase (MAPK/ERK) pathway is pivotal in controlling the proliferation and survival of melanoma cells. Several mutations, including those in BRAF, exhibit an oncogenic effect leading to increased cellular proliferation. As a result, the combination therapy of a MEK inhibitor with a BRAF inhibitor demonstrated higher efficacy and lower toxicity than BRAF inhibitor alone. This combination has become the preferred standard of care for tumors driven by BRAF mutations. Aldehyde dehydrogenase 1A1 (ALDH1A1) is a known marker of stemness involved in drug resistance in several type of tumors, including melanoma. This study demonstrates that melanoma cells overexpressing ALDH1A1 displayed resistance to vemurafenib and trametinib through the activation of PI3K/AKT signaling instead of MAPK axis. Inhibition of PI3K/AKT signaling partially rescued sensitivity to the drugs. Consistently, pharmacological inhibition of ALDH1A1 activity downregulated the activation of AKT and partially recovered responsiveness to vemurafenib and trametinib. We propose ALDH1A1 as a new potential target for treating melanoma resistant to MAPK/ERK inhibitors.


Subject(s)
Aldehyde Dehydrogenase 1 Family , Drug Resistance, Neoplasm , Melanoma , Neoplastic Stem Cells , Protein Kinase Inhibitors , Proto-Oncogene Proteins c-akt , Retinal Dehydrogenase , Humans , Melanoma/drug therapy , Melanoma/pathology , Melanoma/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/physiology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Cell Line, Tumor , Aldehyde Dehydrogenase 1 Family/metabolism , Aldehyde Dehydrogenase 1 Family/genetics , Retinal Dehydrogenase/metabolism , Protein Kinase Inhibitors/pharmacology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Pyrimidinones/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Pyridones/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Vemurafenib/pharmacology , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase/antagonists & inhibitors , Aldehyde Dehydrogenase/genetics , Antineoplastic Agents/pharmacology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Phenotype
6.
Medicine (Baltimore) ; 103(17): e37870, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38669404

ABSTRACT

RATIONALE: Erdheim-Chester disease (ECD) is a rare multisystemic disease characterized by the infiltration of multiple organs by foamy CD68 + CD1a-histiocytes. The genetic background consists of gain-of-function somatic mutations in the mitogen-activated protein kinase pathway. The purpose of the present paper is to make a contribution to the scientific literature on ECD by reporting our experience with a complex clinical case report, along with a concise review of the literature. We discussed the unusual clinical presentation, the complex diagnostic process and the comparison with other published cases. PATIENT CONCERNS: A 70-year-old man presented with arthralgia due to multiple bone areas of sclerosis, first diagnosed with metastases of a prostatic neoplasm. Sequential thorax-abdomen, femoral and homer contrast-enhanced computed tomography (CT) showed pericardial effusion, pulmonary fibrosis, and perirenal fibrous tissue as "hairy kidneys." He underwent. Three bone biopsies were unsuccessful to reach diagnosis. DIAGNOSES: A xanthelasma biopsy showed histopathological signs compatible with ECD; genetic analysis showed the mutation BRAFV600E. INTERVENTIONS: The patient underwent targeted therapy with vemurafenib (BRAF-inhibitor), discontinued 2 weeks later due to the onset of a diffuse erythematous papular rash on the trunk and limbs. OUTCOMES: At the 1-year follow-up, there was only progression of chronic kidney disease (CKD). LESSONS: The present case report describes how ECD diagnosis could represent a challenge for clinicians, owing to its heterogeneous clinical presentation. Early diagnosis followed by prompt therapy is essential for modifying the natural history of the disease.


Subject(s)
Erdheim-Chester Disease , Humans , Erdheim-Chester Disease/diagnosis , Erdheim-Chester Disease/genetics , Male , Aged , Proto-Oncogene Proteins B-raf/genetics , Vemurafenib/therapeutic use
7.
Leuk Res ; 140: 107495, 2024 May.
Article in English | MEDLINE | ID: mdl-38599153

ABSTRACT

BACKGROUND: Hairy cell leukemia (HCL) is a rare mature B-cell malignancy that is primarily treated with purine analogues. However, relapse remains a significant challenge, prompting the search for alternative therapies. The BRAF V600E mutation prevalent in HCL patients provides a target for treatment with vemurafenib. PATIENTS AND METHODS: This multicenter retrospective study included nine patients with relapsed/refractory (R/R) HCL from six different centers. Patient data included demographics, prior treatments, clinical outcomes, and adverse events. RESULTS: Patients received different treatment regimens between centers, including vemurafenib alone or in combination with rituximab. Despite the differences in protocols, all patients achieved at least a partial response, with seven patients achieving a complete response. Adverse events were generally mild with manageable side effects. The absence of myelotoxic effects and manageable side effects make BRAF inhibitors attractive, especially for patients ineligible for purine analogues or those with severe neutropenia. CONCLUSION: Single agent vemurafenib or in combination with rituximab appears to be a promising therapeutic option for R/R HCL. Further research is needed to establish standardized treatment protocols and to investigate long-term outcomes.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Leukemia, Hairy Cell , Rituximab , Vemurafenib , Humans , Leukemia, Hairy Cell/drug therapy , Leukemia, Hairy Cell/pathology , Vemurafenib/administration & dosage , Vemurafenib/therapeutic use , Vemurafenib/adverse effects , Rituximab/administration & dosage , Rituximab/therapeutic use , Rituximab/adverse effects , Male , Middle Aged , Female , Retrospective Studies , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Adult , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Treatment Outcome , Aged, 80 and over , Drug Resistance, Neoplasm
8.
Exp Cell Res ; 438(1): 114033, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38593916

ABSTRACT

Regardless of the clinical response and improved patient survival observed following treatment with BRAFi like Vemurafenib (Vem), rapid development of resistance still remains as a major obstacle in melanoma therapy. In this context, we developed and characterized two acquired Vem-resistant melanoma cell lines, A375V and SK-MEL-28V, and an intrinsically Vem-resistant cell line, RPMI-7951. Altered morphology and growth rate of the resistant cell lines displayed spindle-shaped cells with filopodia formation and enhanced proliferation rate as compared to parental cells. Further in vitro characterization in 2D models confirmed the emergence of a resistant phenotype in melanoma cells. To mimic the in vivo tumor microenvironment, spheroids were developed for both parental and resistant cell lines to recognize materialization of invadopodia structures demonstrating elevated invasiveness and proliferation of resistant cells-based spheroids, especially A375V. Importantly, we validated A375V cell line in vivo to prove its tumorigenicity and drug resistance in tumor xenograft model. Taken together, our established clinically relevant Vem-resistant tumor model could be beneficial to elucidate drug resistance mechanisms, screen and identify novel anticancer therapies to overcome BRAFi resistance in melanoma.


Subject(s)
Cell Proliferation , Drug Resistance, Neoplasm , Melanoma , Proto-Oncogene Proteins B-raf , Vemurafenib , Humans , Melanoma/drug therapy , Melanoma/pathology , Drug Resistance, Neoplasm/drug effects , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Proto-Oncogene Proteins B-raf/genetics , Vemurafenib/pharmacology , Mice , Xenograft Model Antitumor Assays , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Protein Kinase Inhibitors/pharmacology , Tumor Microenvironment/drug effects , Antineoplastic Agents/pharmacology , Mice, Nude
9.
Medicine (Baltimore) ; 103(11): e37513, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38489728

ABSTRACT

BACKGROUND: While papillary thyroid carcinoma (PTC) generally exhibits a favorable prognosis post-surgery, the poorly differentiated subtype presents elevated rates of postoperative recurrence. Certain aggressive cases demonstrate invasive behavior, compromising adjacent structures and leading to a poor prognosis. This study delineates a unique case of postoperative PTC recurrence, complicated by esophageal fistula, that showed favorable outcomes following brief Vemurafenib treatment. PATIENT DESCRIPTION: A 64-year-old female patient underwent surgical resection for PTC, subsequently experiencing rapid tumor recurrence and development of an esophageal fistula. DIAGNOSIS: The patient was confirmed to have locally advanced PTC through intraoperative cytopathology. The cancer recurred postoperatively, culminating in the formation of an esophageal fistula. METHODS: The patient was administered Vemurafenib at a dosage of 960 mg twice daily following tumor recurrence. RESULTS: A 12-month regimen of targeted Vemurafenib therapy led to a substantial reduction in tumor size. Concurrently, the esophageal fistula underwent complete healing, facilitating successful removal of the gastrostomy tube. The tumor response was classified as stable disease. CONCLUSION SUBSECTIONS: Vemurafenib demonstrates potential as a targeted therapeutic strategy for recurrent PTC harboring the BRAFV600E mutation. This approach may effectively mitigate tumor dimensions and the associated risk of esophageal and tracheal fistulas.


Subject(s)
Carcinoma, Papillary , Carcinoma , Esophageal Fistula , Thyroid Neoplasms , Female , Humans , Middle Aged , Thyroid Cancer, Papillary , Vemurafenib/therapeutic use , Thyroid Neoplasms/complications , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/surgery , Carcinoma/drug therapy , Carcinoma/surgery , Carcinoma/genetics , Carcinoma, Papillary/drug therapy , Carcinoma, Papillary/surgery , Carcinoma, Papillary/pathology , Neoplasm Recurrence, Local/pathology , Prognosis
10.
Cardiovasc Pathol ; 70: 107625, 2024.
Article in English | MEDLINE | ID: mdl-38417471

ABSTRACT

Erdheim-Chester disease (ECD) is a rare histiocytosis characterized by the foamy CD68+CD1a- histiocytes infiltrating multiple organs and tissues. ECD might be asymptomatic or present with variable manifestations. The diagnosis of ECD requires characteristic radiological findings and pathological features. Herein, we described a 52-year-old female patient who was admitted to our hospital for recurrent pericardial effusion for two months. She has a medical history of papillary thyroid carcinoma (PTC) and underwent a total thyroidectomy two years before admission. The radiological findings suggested a potential diagnosis of ECD. Cytological analysis of the effusion cytology specimen revealed CD68+CD1a- histiocytes, confirming the ECD diagnosis. The BRAF V600E mutation was identified in the histiocytes, prompting the administration of vemurafenib, a BRAF inhibitor. After two months of standard-dose vemurafenib treatment, the disease was well controlled with pericardial effusion regression.


Subject(s)
Erdheim-Chester Disease , Pericardial Effusion , Proto-Oncogene Proteins B-raf , Vemurafenib , Humans , Erdheim-Chester Disease/genetics , Erdheim-Chester Disease/drug therapy , Erdheim-Chester Disease/complications , Erdheim-Chester Disease/pathology , Erdheim-Chester Disease/diagnosis , Female , Pericardial Effusion/pathology , Pericardial Effusion/etiology , Middle Aged , Proto-Oncogene Proteins B-raf/genetics , Vemurafenib/therapeutic use , Treatment Outcome , Mutation , Histiocytes/pathology , Protein Kinase Inhibitors/therapeutic use , Predictive Value of Tests , Cytology
11.
CNS Oncol ; 13(1): CNS106, 2024 01 01.
Article in English | MEDLINE | ID: mdl-38348829

ABSTRACT

Craniopharyngiomas are tumors that arise from the remnants of Rathke's pouch along the nasopharynx to the diencephalon. Current standard of care includes maximal surgical resection versus adjuvant radiation if a maximal resection is unfeasible. Pharmacological therapy with MAPK targeted agents is an emerging therapeutic option for tumors with BRAF V600E mutations. We report a 45-year-old male with a strictly third ventricle papillary craniopharyngioma with a BRAF V600E mutation. After initial surgery with subtotal resection, the patient demonstrated durable response to targeted BRAF and MEK inhibitor therapy with vemurafenib and cobimetinib. Our report suggests that targeted therapy may reduce the need for radiation and impact surgical interventions in select cases.


Subject(s)
Azetidines , Craniopharyngioma , Piperidines , Pituitary Neoplasms , Male , Humans , Middle Aged , Vemurafenib/therapeutic use , Craniopharyngioma/drug therapy , Proto-Oncogene Proteins B-raf/genetics , Pituitary Neoplasms/diagnostic imaging , Pituitary Neoplasms/drug therapy , Pituitary Neoplasms/genetics , Mutation/genetics
12.
BMC Cancer ; 24(1): 105, 2024 Feb 12.
Article in English | MEDLINE | ID: mdl-38342891

ABSTRACT

BACKGROUND: Histiocytoses are rare disorders manifested by increased proliferation of pathogenic myeloid cells sharing histological features with macrophages or dendritic cells and accumulating in various organs, i.a., bone and skin. Pre-clinical in vitro models that could be used to determine molecular pathways of the disease are limited, hence research on histiocytoses is challenging. The current study compares cytophysiological features of progenitor, stromal-like cells derived from histiocytic lesions (sl-pHCs) of three pediatric patients with different histiocytoses types and outcomes. The characterized cells may find potential applications in drug testing. METHODS: Molecular phenotype of the cells, i.e. expression of CD1a and CD207 (langerin), was determined using flow cytometry. Cytogenetic analysis included GTG-banded metaphases and microarray (aCGH) evaluation. Furthermore, the morphology and ultrastructure of cells were evaluated using a confocal and scanning electron microscope. The microphotographs from the confocal imaging were used to reconstruct the mitochondrial network and its morphology. Basic cytophysiological parameters, such as viability, mitochondrial activity, and proliferation, were analyzed using multiple cellular assays, including Annexin V/7-AAD staining, mitopotential analysis, BrdU test, clonogenicity analysis, and distribution of cells within the cell cycle. Biomarkers potentially associated with histiocytoses progression were determined using RT-qPCR at mRNA, miRNA and lncRNA levels. Intracellular accumulation of histiocytosis-specific proteins was detected with Western blot. Cytotoxicyty and IC50 of vemurafenib and trametinib were determined with MTS assay. RESULTS: Obtained cellular models, i.e. RAB-1, HAN-1, and CHR-1, are heterogenic in terms of molecular phenotype and morphology. The cells express CD1a/CD207 markers characteristic for dendritic cells, but also show intracellular accumulation of markers characteristic for cells of mesenchymal origin, i.e. vimentin (VIM) and osteopontin (OPN). In subsequent cultures, cells remain viable and metabolically active, and the mitochondrial network is well developed, with some distinctive morphotypes noted in each cell line. Cell-specific transcriptome profile was noted, providing information on potential new biomarkers (non-coding RNAs) with diagnostic and prognostic features. The cells showed different sensitivity to vemurafenib and trametinib. CONCLUSION: Obtained and characterized cellular models of stromal-like cells derived from histiocytic lesions can be used for studies on histiocytosis biology and drug testing.


Subject(s)
Histiocytosis, Langerhans-Cell , Humans , Child , Histiocytosis, Langerhans-Cell/drug therapy , Histiocytosis, Langerhans-Cell/genetics , Histiocytosis, Langerhans-Cell/diagnosis , Vemurafenib , Macrophages/metabolism , Biomarkers , Phenotype , Antigens, CD , Lectins, C-Type/metabolism , Mannose-Binding Lectins/metabolism
13.
Nat Commun ; 15(1): 1430, 2024 Feb 16.
Article in English | MEDLINE | ID: mdl-38365756

ABSTRACT

Both targeted therapies and immunotherapies provide benefit in resected Stage III melanoma. We hypothesized that the combination of targeted and immunotherapy given prior to therapeutic lymph node dissection (TLND) would be tolerable and drive robust pathologic responses. In NeoACTIVATE (NCT03554083), a Phase II trial, patients with clinically evident resectable Stage III melanoma received either 12 weeks of neoadjuvant vemurafenib, cobimetinib, and atezolizumab (BRAF-mutated, Cohort A, n = 15), or cobimetinib and atezolizumab (BRAF-wild-type, Cohort B, n = 15) followed by TLND and 24 weeks of adjuvant atezolizumab. Here, we report outcomes from the neoadjuvant portion of the trial. Based on intent to treat analysis, pathologic response (≤50% viable tumor) and major pathologic response (complete or near-complete, ≤10% viable tumor) were observed in 86.7% and 66.7% of BRAF-mutated and 53.3% and 33.3% of BRAF-wild-type patients, respectively (primary outcome); these exceeded pre-specified benchmarks of 50% and 30% for major pathologic response. Grade 3 and higher toxicities, primarily dermatologic, occurred in 63% during neoadjuvant treatment (secondary outcome). No surgical delays nor progression to regional unresectability occurred (secondary outcome). Peripheral blood CD8 + TCM cell expansion associated with favorable pathologic responses (exploratory outcome).


Subject(s)
Antibodies, Monoclonal, Humanized , Azetidines , Melanoma , Piperidines , Skin Neoplasms , Humans , Melanoma/drug therapy , Melanoma/etiology , Vemurafenib/therapeutic use , Neoadjuvant Therapy , Proto-Oncogene Proteins B-raf/genetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Skin Neoplasms/drug therapy , Skin Neoplasms/etiology , Mutation
14.
J Nat Med ; 78(2): 342-354, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38324123

ABSTRACT

Evodiamine, a novel alkaloid, was isolated from the fruit of tetradium. It exerts a diversity of pharmacological effects and has been used to treat gastropathy, hypertension, and eczema. Several studies reported that evodiamine has various biological effects, including anti-nociceptive, anti-bacterial, anti-obesity, and anti-cancer activities. However, there is no research regarding its effects on drug-resistant cancer. This study aimed to investigate the effect of evodiamine on human vemurafenib-resistant melanoma cells (A375/R cells) proliferation ability and its mechanism. Cell activity was assessed using the cell counting kit-8 (CCK-8) method. Flow cytometry assay was used to assess cell apoptosis and cell cycle. A xenograft model was used to analyze the inhibitory effects of evodiamine on tumor growth. Bioinformatics analyses, network pharmacology, and molecular docking were used to explore the potential mechanism of evodiamine in vemurafenib-resistant melanoma. RT-qPCR and Western blotting were performed to reveal the molecular mechanism. The alkaloid extract of the fruit of tetradium, evodiamine showed the strongest tumor inhibitory effect on vemurafenib-resistant melanoma cells compared to treatment with vemurafenib alone. Evodiamine inhibited vemurafenib-resistant melanoma cell growth, proliferation, and induced apoptosis, conforming to a dose-effect relationship and time-effect relationship. Results from network pharmacology and molecular docking suggested that evodiamine might interact with IRS4 to suppress growth of human vemurafenib-resistant melanoma cells. Interestingly, evodiamine suppressed IRS4 expression and then inhibited PI3K/AKT signaling pathway, and thus had the therapeutic action on vemurafenib-resistant melanoma.


Subject(s)
Alkaloids , Antineoplastic Agents , Melanoma , Quinazolines , Humans , Vemurafenib/pharmacology , Vemurafenib/therapeutic use , Melanoma/drug therapy , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Molecular Docking Simulation , Signal Transduction , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Cell Proliferation , Alkaloids/pharmacology , Cell Line, Tumor , Insulin Receptor Substrate Proteins/metabolism
15.
AAPS PharmSciTech ; 25(1): 14, 2024 Jan 08.
Article in English | MEDLINE | ID: mdl-38191830

ABSTRACT

Vemurafenib (VMF) is a practically insoluble (< 0.1 µg/mL) and least bioavailable (1%) drug. To enhance its oral bioavailability and solubility, we formulated a reliable self-nano emulsifying drug delivery system (SNEDDS). A Quality by Design (QbD) approach was used to optimize the ratio of Capryol 90, Tween 80, and Transcutol HP. VMF-loaded SNEDDS was characterized for its size, polydispersity index (PDI), zeta potential, drug content, and transmittance. The in vitro release profile of the drug loaded in SNEDDS was compared to the free drug in two media, pH 6.8 and 1.2, and the data obtained were analyzed with different mathematical models. A reverse-phase ultra-pressure liquid chromatography (UPLC) technique with high sensitivity and selectivity was developed and validated for the quantification of VMF in analytical and bioanalytical samples. Dissolution efficiency for SNEDDS was estimated using different models, which proved that the developed novel SNEDDS formulation had a better in vitro dissolution profile than the free drug. A 2.13-fold enhanced oral bioavailability of VMF-loaded SNEDDS compared to the free drug demonstrates the superiority of the developed formulation. This work thus presents an overview of VMF-loaded SNEDDS as a promising alternative to improve the oral bioavailability of the drug.


Subject(s)
Chromatography, Reverse-Phase , Polysorbates , Biological Availability , Vemurafenib , Solubility
16.
Biol Direct ; 19(1): 6, 2024 01 04.
Article in English | MEDLINE | ID: mdl-38178263

ABSTRACT

BACKGROUND: The outcome of Acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) remain dismal despite the development of treatment. Targeted therapy is gaining more and more attention in improving prognosis. METHODS: Expression of BRAF was analyzed by RT-qPCR in AML and MDS patients. Cells viability treated by drugs was measured by CCK-8 assay. Network pharmacology and RNA-sequence were used to analyze the mechanism of drugs and verified in vitro and xenograft tumor model. RESULTS: Here we showed that BRAF was overexpressed in AML and MDS patients, and correlated with poor prognosis. The BRAF inhibitor-Vemurafenib (VEM) could significantly induce senescence, proliferation inhibition and apoptosis in AML cells, which can be enhanced by Bortezomib (BOR). This inhibitory effect was also verified in CD34 + cells derived from AML patients. Mechanistically, we showed that VEM combined with BOR could turn on HIPPO signaling pathway, thereby inducing cellular senescence in AML cells and xenograft mouse. CONCLUSIONS: Taken together, our findings demonstrate a significant upregulation of BRAF expression in AML and MDS patients, which is associated with unfavorable clinical outcomes. We also discovered that the BRAF inhibitor Vemurafenib induces cellular senescence through activation of the HIPPO signaling pathway. Analysis of BRAF expression holds promise as a prognostic indicator and potential therapeutic target for individuals with AML and MDS.


Subject(s)
Leukemia, Myeloid, Acute , Myelodysplastic Syndromes , Humans , Animals , Mice , Vemurafenib/pharmacology , Vemurafenib/therapeutic use , Hippo Signaling Pathway , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins B-raf/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/complications , Myelodysplastic Syndromes/pathology
17.
J Cancer Res Clin Oncol ; 150(1): 12, 2024 Jan 17.
Article in English | MEDLINE | ID: mdl-38231288

ABSTRACT

BACKGROUND: Langerhans cell histiocytosis (LCH) is a myeloid neoplasia with potentially fatal consequences, and about 2/3 of cases involve the BRAFV600E kinase-activated mutation. Vemurafenib, a BRAF inhibitor, has demonstrated significant clinical improvements in LCH. However, the high relapse rate of LCH following cessation of vemurafenib therapy remains a major challenge, and alternative treatment strategies require further investigation. METHODS: In this retrospective multi-center study, we evaluated the efficacy and safety of vemurafenib combined with conventional chemotherapy in patients with severe or refractory LCH. RESULTS: Seventeen patients were enrolled in the study, with eleven classified as risk organ involvement (RO +). Six received the combination therapy as the primary treatment, and eleven after being refractory to prior chemotherapy. The overall response rate was 94.1%. Progression-free survival among all 17 patients was 70.6% (12/17) at a median follow-up of 32 months, and relapse-free survival among the 15 patients with discontinuation after a response was 73.3%(11/15) at a median follow-up of 34 months. Five of six patients (83.3%) with myeloid BRAFV600E mutations demonstrated molecular remission. The overall survival rate was 100%. Adverse events were mostly classified as grades 1 or 2. CONCLUSION: Our data suggest that the combination of vemurafenib and chemotherapy can achieve sustained clinical and molecular level relief in children with LCH, and side effects are tolerable.


Subject(s)
Protein Kinase Inhibitors , Proto-Oncogene Proteins B-raf , Humans , Child , Vemurafenib , Proto-Oncogene Proteins B-raf/genetics , Protein Kinase Inhibitors/therapeutic use , Combined Modality Therapy , Mutation
18.
J Immunother Cancer ; 12(1)2024 01 04.
Article in English | MEDLINE | ID: mdl-38177075

ABSTRACT

Durable remissions are observed in 10%-20% of treated patients with advanced metastatic melanoma but the factors associated with long-term complete clinical responses are largely unknown. Here, we report the molecular characteristics of tumor evolution during disease progression along a 9-year clinical course in a patient with advanced disseminated melanoma who received different treatments, including trametinib, ipilimumab, radiation, vemurafenib, surgical tumor debulking and a second ipilimumab course, ultimately achieving complete long-term disease remission.Longitudinal analyses of therapies-resistant metastatic tumors revealed the effects of different treatments on tumor's microenvironment and immunogenicity, ultimately creating a milieu favorable to immunotherapy response. Monitoring of the temporal dynamics of T cells by analysis of the T cell receptor (TCR) repertoire in the tumor and peripheral blood during disease evolution indicated that T-cell clones with common TCR rearrangements, present at low levels at baseline, were maintained and expanded after immunotherapy, and that TCR diversity increased. Analysis of genetic, molecular, and cellular components of the tumor depicted a multistep process in which treatment with kinase inhibitors strongly conditioned the immune microenvironment creating an inflamed milieu converting cold into hot tumors, while ipilimumab impacted and increased the TCR repertoire, a requirement for tumor rejection.Since the optimal sequencing of treatment with antibodies targeting immune checkpoints and kinase inhibitors for advanced melanoma is still clinically debated, this case indicates that immunotherapy success is possible even after progression on targeted therapy.


Subject(s)
Melanoma , Humans , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Ipilimumab/therapeutic use , Vemurafenib , T-Lymphocytes/pathology , Receptors, Antigen, T-Cell/therapeutic use , Tumor Microenvironment
19.
Int J Cancer ; 154(6): 1057-1072, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38078628

ABSTRACT

About 25% of melanoma harbor activating NRAS mutations, which are associated with aggressive disease therefore requiring a rapid antitumor intervention. However, no efficient targeted therapy options are currently available for patients with NRAS-mutant melanoma. MEK inhibitors (MEKi) appear to display a moderate antitumor activity and also immunological effects in NRAS-mutant melanoma, providing an ideal backbone for combination treatments. In our study, the MEKi binimetinib, cobimetinib and trametinib combined with the BRAF inhibitors (BRAFi) encorafenib, vemurafenib and dabrafenib were investigated for their ability to inhibit proliferation, induce apoptosis and alter the expression of immune modulatory molecules in sensitive NRAS-mutant melanoma cells using two- and three-dimensional cell culture models as well as RNA sequencing analyses. Furthermore, NRAS-mutant melanoma cells resistant to the three BRAFi/MEKi combinations were established to characterize the mechanisms contributing to their resistance. All BRAFi induced a stress response in the sensitive NRAS-mutant melanoma cells thereby significantly enhancing the antiproliferative and proapoptotic activity of the MEKi analyzed. Furthermore, BRAFi/MEKi combinations upregulated immune relevant molecules, such as ICOS-L, components of antigen-presenting machinery and the "don't eat me signal" molecule CD47 in the melanoma cells. The BRAFi/MEKi-resistant, NRAS-mutant melanoma cells counteracted the molecular and immunological effects of BRAFi/MEKi by upregulating downstream mitogen-activated protein kinase pathway molecules, inhibiting apoptosis and promoting immune escape mechanisms. Together, our study reveals potent molecular and immunological effects of BRAFi/MEKi in sensitive NRAS-mutant melanoma cells that may be exploited in new combinational treatment strategies for patients with NRAS-mutant melanoma.


Subject(s)
Melanoma , Humans , Melanoma/drug therapy , Melanoma/genetics , Melanoma/metabolism , Proto-Oncogene Proteins B-raf , Vemurafenib , Protein Kinase Inhibitors/adverse effects , Mitogen-Activated Protein Kinase Kinases , Mutation , Drug Resistance, Neoplasm/genetics , Membrane Proteins/genetics , GTP Phosphohydrolases/genetics
20.
J Gene Med ; 26(1): e3644, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38072402

ABSTRACT

BACKGROUND: Melanoma, a frequently encountered cutaneous malignancy characterized by a poor prognosis, persists in presenting formidable challenges despite the advancement in molecularly targeted drugs designed to improve survival rates significantly. Unfortunately, as more therapeutic choices have developed over time, the gradual emergence of drug resistance has become a notable impediment to the effectiveness of these therapeutic interventions. The hepatocyte growth factor (HGF)/c-met signaling pathway has attracted considerable attention, associated with drug resistance stemming from multiple potential mutations within the c-met gene. The activation of the HGF/c-met pathway operates in an autocrine manner in melanoma. Notably, a key player in the regulatory orchestration of HGF/c-met activation is the long non-coding RNA MEG3. METHODS: Melanoma tissues were collected to measure MEG3 expression. In vitro validation was performed on MEG3 to prove its oncogenic roles. Bioinformatic analyses were conducted on the TCGA database to build the MEG3-related score. The immune characteristics and mutation features of the MEG3-related score were explored. RESULTS: We revealed a negative correlation between HGF and MEG3. In melanoma cells, HGF inhibited MEG3 expression by augmenting the methylation of the MEG3 promoter. Significantly, MEG3 exhibits a suppressive impact on the proliferation and migration of melanoma cells, concurrently inhibiting c-met expression. Moreover, a predictive model centered around MEG3 demonstrates notable efficacy in forecasting critical prognostic indicators, immunological profiles, and mutation statuses among melanoma patients. CONCLUSIONS: The present study highlights the potential of MEG3 as a pivotal regulator of c-met, establishing it as a promising candidate for targeted drug development in the ongoing pursuit of effective therapeutic interventions.


Subject(s)
Melanoma , Skin Neoplasms , Humans , Melanoma/drug therapy , Melanoma/genetics , Melanoma/metabolism , Vemurafenib/pharmacology , Vemurafenib/therapeutic use , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Methylation , Cell Proliferation , Cell Line, Tumor
SELECTION OF CITATIONS
SEARCH DETAIL
...