Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 218
Filtrer
2.
Cancers (Basel) ; 16(12)2024 Jun 10.
Article de Anglais | MEDLINE | ID: mdl-38927891

RÉSUMÉ

INTRODUCTION: Hairy-cell leukemia (HCL) is a rare B-cell chronic lymphoproliferative disorder (B-CLPD), whose favorable prognosis has changed with the use of purine nucleoside analogs (PNAs), such as cladribine (CDA) or pentostatin (P). However, some patients eventually relapse and over time HCL becomes resistant to chemotherapy. Many discoveries have been made in the pathophysiology of HCL during the last decade, especially in genomics, with the identification of the BRAFV600E mutation and cellular biology, including the importance of signaling pathways as well as tumor microenvironment. All of these new developments led to targeted treatments, especially BRAF inhibitors (BRAFis), MEK inhibitors (MEKis), Bruton's tyrosine kinase (BTK) inhibitors (BTKis) and recombinant anti-CD22 immunoconjugates. RESULTS: The following major changes or additions were introduced in these updated guidelines: the clinical relevance of the changes in the classification of splenic B-cell lymphomas and leukemias; the increasingly important diagnostic role of BRAFV600E mutation; and the prognostic role of the immunoglobulin (IG) variable (V) heavy chain (H) (IGHV) mutational status and repertory. We also wish to insist on the specific involvement of bones, skin, brain and/or cerebrospinal fluid (CSF) of the disease at diagnosis or during the follow-up, the novel targeted drugs (BRAFi and MEKi) used for HCL treatment, and the increasing role of minimal residual disease (MRD) assessment. CONCLUSION: Here we present recommendations for the diagnosis of HCL, treatment in first line and in relapsed/refractory patients as well as for HCL-like disorders including HCL variant (HCL-V)/splenic B-cell lymphomas/leukemias with prominent nucleoli (SBLPN) and splenic diffuse red pulp lymphoma (SDRPL).

3.
Front Oncol ; 14: 1359093, 2024.
Article de Anglais | MEDLINE | ID: mdl-38774414

RÉSUMÉ

Dabrafenib plus trametinib is a promising new therapy for patients affected by BRAFV600E-mutant glioma, with high overall response and manageable toxicity. We described a complete and long-lasting response in a case of recurrent anaplastic pleomorphic xanthoastrocytoma CNS WHO-grade 3 BRAFV600E mutated. Due to very poor prognosis, there are a few described cases of high-grade glioma (HGG) patients treated with the combined target therapy as third-line treatment. The emergence of optimized sequencing strategies and targeted agents, including multimodal and systemic therapy with dabrafenib plus trametinib, will continue to broaden personalized therapy in HGG improving patient outcomes.

4.
Expert Rev Hematol ; 17(6): 223-231, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38721670

RÉSUMÉ

INTRODUCTION: Langerhans cell histiocytosis (LCH) is a rare myeloid neoplasm, encompassing a diverse clinical spectrum ranging from localized bone or skin lesions to a multisystemic life-threatening condition. Over the past decade, there has been an expansion in understanding the molecular biology of LCH, which translated into innovative targeted therapeutic approaches. AREAS COVERED: In this article, we will review the molecular alterations observed in pediatric LCH and the relationship between these molecular changes and the clinical phenotype, as well as targeted therapies in LCH. EXPERT OPINION: Mitogen-activated protein kinase (MAPK) pathway mutation is a hallmark of LCH and is identified in 80% of the cases. Notably, BRAFV600E mutation is seen in ~50-60% of the cases, ~30% has other MAPK pathway mutations, while 15-20% have no detected mutations. While the first line therapeutic approach is vinblastine and prednisone, targeted therapies - specifically BRAF/MEK inhibitors - emerged as a promising second-line salvage strategy, particularly when a mutation is identified. Most patients respond to BRAF/MEK inhibitors but at least 75% reactivate after stopping, however, most patients respond again when restarting inhibitors.


Sujet(s)
Histiocytose à cellules de Langerhans , Thérapie moléculaire ciblée , Humains , Enfant , Histiocytose à cellules de Langerhans/génétique , Histiocytose à cellules de Langerhans/traitement médicamenteux , Histiocytose à cellules de Langerhans/thérapie , Histiocytose à cellules de Langerhans/diagnostic , Mutation , Protéines proto-oncogènes B-raf/génétique , Protéines proto-oncogènes B-raf/antagonistes et inhibiteurs , Marqueurs génétiques , Inhibiteurs de protéines kinases/usage thérapeutique , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques
5.
Int J Mol Sci ; 25(9)2024 Apr 24.
Article de Anglais | MEDLINE | ID: mdl-38731852

RÉSUMÉ

Lung cancer, despite recent advancements in survival rates, represents a significant global health burden. Non-small cell lung cancer (NSCLC), the most prevalent type, is driven largely by activating mutations in Kirsten rat sarcoma viral oncogene homologue (KRAS) and receptor tyrosine kinases (RTKs), and less in v-RAF murine sarcoma viral oncogene homolog B (BRAF) and mitogen-activated protein-kinase kinase (MEK), all key components of the RTK-RAS-mitogen-activated protein kinase (MAPK) pathway. Learning from melanoma, the identification of BRAFV600E substitution in NSCLC provided the rationale for the investigation of RAF and MEK inhibition as a therapeutic strategy. The regulatory approval of two RAF-MEK inhibitor combinations, dabrafenib-trametinib, in 2017, and encorafenib-binimetinib, in 2023, signifies a breakthrough for the management of BRAFV600E-mutant NSCLC patients. However, the almost universal emergence of acquired resistance limits their clinical benefit. New RAF and MEK inhibitors, with distinct biochemical characteristics, are in preclinical and clinical development. In this review, we aim to provide valuable insights into the current state of RAF and MEK inhibition in the management of NSCLC, fostering a deeper understanding of the potential impact on patient outcomes.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Mitogen-Activated Protein Kinase Kinases , Inhibiteurs de protéines kinases , Humains , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/génétique , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Inhibiteurs de protéines kinases/usage thérapeutique , Inhibiteurs de protéines kinases/pharmacologie , Mitogen-Activated Protein Kinase Kinases/antagonistes et inhibiteurs , Mitogen-Activated Protein Kinase Kinases/métabolisme , Animaux , Kinases raf/antagonistes et inhibiteurs , Kinases raf/métabolisme , Kinases raf/génétique , Mutation
6.
Biomed Pharmacother ; 176: 116766, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38788599

RÉSUMÉ

Activation of neuropilin-1 (NRP-1) by platelet derived growth factor (PDGF)-C sustains melanoma invasiveness. Therefore, in the search of novel agents capable of reducing melanoma spreading, PDGF-C/NRP-1 interaction was investigated as a potential druggable target. Since the PDGF-C region involved in NRP-1 binding is not yet known, based on the sequence and structural homology between PDGF-C and vascular endothelial growth factor-A (VEGF-A), we hypothesized that the NRP-1 b1 domain region involved in the interaction with VEGF-A might also be required for PDGF-C binding. Hence, this region was selected from the protein crystal structure and used as target in the molecular docking procedure. In the following virtual screening, compounds from a DrugBank database were used as query ligands to identify agents potentially capable of disrupting NRP-1/PDGF-C interaction. Among the top 45 candidates with the highest affinity, five drugs were selected based on the safety profile, lack of hormonal effects, and current availability in the market: the antipsychotic pimozide, antidiabetic gliclazide, antiallergic cromolyn sodium, anticancer tyrosine kinase inhibitor entrectinib, and antihistamine azelastine. Analysis of drug influence on PDGF-C in vitro binding to NRP-1 and PDGF-C induced migration of human melanoma cells expressing NRP-1, indicated gliclazide and entrectinib as the most specific agents that were active at clinically achievable and non-toxic concentrations. Both drugs also reverted PDGF-C ability to stimulate extracellular matrix invasion by melanoma cells resistant to BRAF inhibitors. The inhibitory effect on tumor cell motility involved a decrease of p130Cas phosphorylation, a signal transduction pathway activated by PDGF-C-mediated stimulation of NRP-1.


Sujet(s)
Lymphokines , Mélanome , Simulation de docking moléculaire , Neuropiline 1 , Facteur de croissance dérivé des plaquettes , Humains , Mélanome/traitement médicamenteux , Mélanome/anatomopathologie , Mélanome/métabolisme , Lymphokines/métabolisme , Facteur de croissance dérivé des plaquettes/métabolisme , Neuropiline 1/métabolisme , Lignée cellulaire tumorale , Liaison aux protéines , Mouvement cellulaire/effets des médicaments et des substances chimiques , Métastase tumorale , Antinéoplasiques/pharmacologie
7.
Endocrine ; 2024 May 06.
Article de Anglais | MEDLINE | ID: mdl-38709445

RÉSUMÉ

PURPOSE: Approximately 45% of anaplastic thyroid cancer (ATC) patients harbor a BRAFV600E mutation and are eligible for target therapy (TT) with BRAF and MEK inhibitors (BRAFi/MEKi), nevertheless, few data advocate for this. Hence, we've conducted a systematic review and meta-analysis investigating the effectiveness and safety of BRAFi/MEKi in BRAFV600E ATC patients. METHODS: PubMed, Embase, and the Cochrane Library were systematically searched for BRAFi/MEKi TT in BRAFV600E ATC patients. Outcomes included objective response rate (ORR), disease control rate (DCR), overall survival (OS), progression-free survival (PFS), duration of response (DOR) and adverse events (AEs). RESULTS: Nine studies with 168 patients were included. Median follow-up ranged from 2.0 to 47.9 months. 75% of patients had stage IVc. In a pooled analysis, ORR was 68.15% (95% CI 55.31-80.99, I2 = 47%) and DCR was 85.39% (95% CI 78.10-92.68, I2 = 0), with a median DOR of 14.4 months (95% CI 4.6-14.4) and a median PFS of 6.7 months (95% CI 4.7-34.2). Moreover, 1-year OS rate was 64.97% (95% CI 48.76-81.17, I2 = 84%) and 2-years OS rate was 52.08% (95% CI 35.71-68.45, I2 = 79%). Subgroup analysis showed patients in the neoadjuvant setting had higher rates of 1 and 2-years OS and observational studies tended to report higher rates of ORR than clinical trials. No new or unexpected adverse events were found. CONCLUSIONS: Our study demonstrated BRAFi/MEKi have a decent activity for BRAFV600E ATC patients, especially in the neoadjuvant setting, with a tolerable safety profile. However, further clinical trials are warranted to investigate these findings.

8.
Ups J Med Sci ; 1292024.
Article de Anglais | MEDLINE | ID: mdl-38716076

RÉSUMÉ

Background: Metastatic neuroendocrine carcinoma (NEC) is associated with short survival. Other than platinum-based chemotherapy, there is no clear standard regimen. Current guidelines suggest that combination treatment with BRAF-inhibitors should be considered for patients with BRAF V600E-mutated NEC. However, since only eight such patients have been reported in the literature, our object was to confirm the validity of this recommendation. Methods: This was a single-center retrospective cohort study conducted at Uppsala University Hospital. The included patients 1) had a histopathologically confirmed diagnosis of NEC, 2) were diagnosed between January 1st, 2018 and December 31st, 2023, 3) had tumor tissue genetically screened by a broad next-generation sequencing (NGS) panel, and 4) showed a tumor mutation for which there is a currently available targeted therapy. Results: We screened 48 patients diagnosed with NEC between January 1st, 2018 and December 31st, 2023. Twelve had been analyzed with a broad NGS-panel, and two had a targetable mutation. Both these patients harbored a BRAF V600E-mutated colon-NEC and were treated with BRAF- and MEK-inhibitors dabrafenib and trametinib in second-line. At first radiological evaluation (RECIST 1.1), both patients had a reduction of tumor size, which decreased by 31 and 40%. Both had short response periods, and their overall survival was 12 and 9 months. Conclusions: BRAF-mutated NEC is sensitive to treatment with BRAF- and MEK-inhibitor combination. These results further support that DNA sequencing should be considered as standard of care in NECs to screen for potential treatment targets.


Sujet(s)
Carcinome neuroendocrine , Oximes , Inhibiteurs de protéines kinases , Protéines proto-oncogènes B-raf , Pyridones , Pyrimidinones , Adulte , Sujet âgé , Femelle , Humains , Mâle , Adulte d'âge moyen , Carcinome neuroendocrine/génétique , Carcinome neuroendocrine/traitement médicamenteux , Séquençage nucléotidique à haut débit , Imidazoles/usage thérapeutique , Imidazoles/administration et posologie , Mutation , Oximes/usage thérapeutique , Oximes/administration et posologie , Inhibiteurs de protéines kinases/usage thérapeutique , Protéines proto-oncogènes B-raf/génétique , Pyridones/usage thérapeutique , Pyridones/administration et posologie , Pyrimidinones/usage thérapeutique , Études rétrospectives , Résultat thérapeutique
9.
Front Oncol ; 14: 1305518, 2024.
Article de Anglais | MEDLINE | ID: mdl-38549927

RÉSUMÉ

Introduction: Erdheim-Chester disease (ECD) is a rare disease that belongs to the group of Dendritic and histiocytic neoplasms. Only 2000 cases have been reported worldwide. It can present with a wide range of symptoms, making a differential diagnosis especially difficult. The primary and most important diagnostic tool is a biopsy of the affected organ/tissue. Nowadays the analysis of different mutations affecting the BRAF and MAPK pathways makes it possible to use targeted treatments, such as vemurafenib, dabrafenib, or cobimetinib. Objective: Our aim is to present the results of three male patients treated in our hematology department. Results: Our BRAF mutation-positive patient presented with retroperitoneal tissue proliferation and diabetes insipidus. The initial therapy of choice was dabrafenib. After 3 months of treatment, 18F-fluoro-deoxyglucose positron emission tomography (FDG-PET)/computed tomography (CT) scans showed regression, and after 2 years of treatment, no disease activity was detected. In our second patient, a recurrent febrile state (not explained by other reasons) and diabetes insipidus suggested the diagnosis. A femoral bone biopsy confirmed BRAF-negative ECD. The first-line therapy was interferon-alpha. After 3 months of treatment, no response was observed on 18FDG-PET/CT, and treatment with cobimetinib was started. The control 18FDG-PET/CT imaging was negative. Our third patient was evaluated for dyspnea, and a CT scan showed fibrosis with hilar lymphadenomegaly. A lung biopsy confirmed BRAF-negative ECD. We started treatment with interferon-alpha, but unfortunately, no improvement was observed. Second-line treatment with cobimetinib resulted in a partial metabolic response (PMR) according to control 18FDG-PET/CT. Conclusions: Our results demonstrate that an appropriately chosen treatment can lead to a good therapeutic response, but dose reduction may be necessary due to side effects. With advanced targeted therapeutic treatment options, survival and quality of life are significantly improved.

10.
Mol Cancer ; 23(1): 64, 2024 03 26.
Article de Anglais | MEDLINE | ID: mdl-38532456

RÉSUMÉ

Previous studies have shown the clinical benefit of rechallenging the RAF pathway in melanoma patients previously treated with BRAF inhibitors. 44 patients with multiple tumors harboring RAF alterations were rechallenged with a second RAF inhibitor, either as monotherapy or in combination with other therapies, after prior therapy with a first RAF inhibitor. This retrospective observational study results showed that rechallenging with RAFi(s) led to an overall response rate of 18.1% [PR in thyroid (1 anaplastic; 3 papillary), 1 ovarian, 2 melanoma, 1 cholangiocarcinoma, and 1 anaplastic astrocytoma]. The clinical benefit rate was 54.5%; more than 30% of patients had durable responses with PR and SD lasting > 6 months. The median progression-free survival on therapy with second RAF inhibitor in the rechallenge setting either as monotherapy or combination was shorter at 2.7 months (0.9-30.1 m) compared to 8.6 months (6.5-11.5 m) with RAF-1i. However, the median PFS with RAF-2i responders (PFS-2) improved at 12.8 months compared to 11.4 months with RAF-1i responders. The median OS from retreatment with RAF-2i was 15.5 months (11.1-30.8 m). Further prospective studies are needed to validate these results and expand targeted therapy options for RAF-aberrant cancers.


Sujet(s)
Mélanome , Humains , Mélanome/anatomopathologie , Protéines proto-oncogènes B-raf/génétique , Inhibiteurs de protéines kinases/usage thérapeutique , Études rétrospectives , Études prospectives , Mutation
11.
EClinicalMedicine ; 69: 102447, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38333370

RÉSUMÉ

Background: BRAF V600 mutations are common in melanoma, thyroid, and non-small-cell lung cancers. Despite dabrafenib and trametinib being standard treatments for certain cancers, their efficacy across various solid tumours remains unelucidated. The BELIEVE trial assessed the efficacy of dabrafenib and trametinib in solid tumours with BRAF V600E/R or non-V600 BRAF mutations. Methods: Between October 1, 2019, and June 2022, at least 50 patients with measurable and seven without measurable diseases examined were enrolled in a subcohort of the BELIEVE trial (NCCH1901, jRCTs031190104). BRAF mutated solid tumour cases other than BRAF V600E mutated colorectal cancer, melanoma, and non-small cell lung cancer cases were included. Patients with solid tumours received dabrafenib (150 mg) twice daily and trametinib (2 mg) once daily until disease progression or intolerable toxicity was observed. The primary endpoint was overall response rate (ORR), and secondary endpoints included progression-free survival (PFS), 6-month PFS, and overall survival (OS). Bayesian analysis was performed using a prior distribution with a 30% expected response rate [Beta (0.6, 1.4)]. Findings: Fourty-seven patients with measurable disease, mainly with the BRAF V600E mutation (94%), and three others with non-V600E BRAF mutations (V600R, G466A, and N486_P490del) were enrolled. The primary sites included the thyroid gland, central nervous system, liver, bile ducts, colorectum, and pancreas. The confirmed ORR was 28.0%; the expected value of posterior distribution [Beta (14.6, 37.4)] was 28.1%, although the primary endpoint was achieved, not exceeding an unexpectedly high response rate of 60% obtained using Bayesian analysis. The disease control rate (DCR) was 84.0%. The median PFS was 6.5 months (95% confidence interval [CI]; 4.2-7.2 months, 87.8% at 6 months). Responses were observed across seven tumour types. Median OS was 9.7 months (95% CI, 7.5-12.2 months). Additional patients without measurable diseases had a median PFS of 4.5 months. Adverse events (AEs) were consistent with previous reports, with 45.6% of patients experiencing grade ≥3 AEs. Interpretation: This study reported promising efficacy against BRAF V600-mutant tumours. Dabrafenib and trametinib would offer a new therapeutic option for rare cancers, such as high-grade gliomas, biliary tract cancer, and thyroid cancer. Funding: This study was funded by the Japan Agency for Medical Research and Development (22ck0106622h0003) and a Health and Labour Sciences Research Grant (19EA1008).

12.
CNS Oncol ; 13(1): CNS106, 2024 01 01.
Article de Anglais | MEDLINE | ID: mdl-38348829

RÉSUMÉ

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.


Sujet(s)
Azétidines , Craniopharyngiome , Pipéridines , Tumeurs de l'hypophyse , Mâle , Humains , Adulte d'âge moyen , Vémurafénib/usage thérapeutique , Craniopharyngiome/traitement médicamenteux , Protéines proto-oncogènes B-raf/génétique , Tumeurs de l'hypophyse/imagerie diagnostique , Tumeurs de l'hypophyse/traitement médicamenteux , Tumeurs de l'hypophyse/génétique , Mutation/génétique
13.
Healthcare (Basel) ; 12(1)2024 Jan 02.
Article de Anglais | MEDLINE | ID: mdl-38201012

RÉSUMÉ

BACKGROUND: Melanoma cancer represents the most lethal type of skin cancer originating from the malignant transformation of melanocyte cells. Almost 50% of melanomas show the activation of BRAF mutations. The identification and characterization of BRAF mutations led to the development of specific drugs that radically changed the therapeutic approach to melanoma. METHODS: We conducted a narrative review of the literature according to a written protocol before conducting the study. This article is based on previously conducted studies. We identified articles by searching electronic databases (Medline, Google Scholar and PubMed). We used a combination of "melanoma", "Braf-Mek inhibitors", " targeted therapy" and "oral side effects". RESULTS: Eighteen studies were reported in this article showing the relationship between the use of targeted therapy in melanoma cancer and the development of oral side effects, such as mucositis, hyperkeratosis and cellular proliferation. CONCLUSION: Targeted therapy plays an important role in the treatment of melanoma cancer, showing a notable increase in response rate, prolonged progression-free survival and overall survival in BRAF-mutated melanoma patients. Oral side effects represent a common finding over the course of treatment. However, these adverse effects can be easily managed in a multidisciplinary approach involving collaboration between medical oncologists and dental doctors.

14.
Oncotarget ; 15: 36-48, 2024 Jan 24.
Article de Anglais | MEDLINE | ID: mdl-38275291

RÉSUMÉ

A subset of thyroid cancers present at advanced stage or with dedifferentiated histology and have limited response to standard therapy. Tumors harboring the BRAF V600E mutation may be treated with BRAF inhibitors; however, tumor response is often short lived due to multiple compensatory resistance mechanisms. One mode of resistance is the transition to an alternative cell state, which on rare occasions can correspond to tumor dedifferentiation. DNA sequencing and RNA expression profiling show that thyroid tumors that dedifferentiate after BRAF inhibition are enriched in known genetic alterations that mediate resistance to BRAF blockade, and may also drive tumor dedifferentiation, including mutations in the PI3K/AKT/MTOR (PIK3CA, MTOR), MAP/ERK (MET, NF2, NRAS, RASA1), SWI/SNF chromatin remodeling complex (ARID2, PBRM1), and JAK/STAT pathways (JAK1). Given these findings, recent investigations have evaluated the efficacy of dual-target therapies; however, continued lack of long-term tumor control illustrates the complex and multifactorial nature of these compensatory mechanisms. Transition to an immune-suppressed state is another correlate of BRAF inhibitor resistance and tumor dedifferentiation, suggesting a possible role for concurrent targeted therapy with immunotherapy. Investigations into combined targeted and immunotherapy are ongoing, but early results with checkpoint inhibitors, viral therapies, and CAR T-cells suggest enhanced anti-tumor immune activity with these combinations.


Sujet(s)
Protéines proto-oncogènes B-raf , Tumeurs de la thyroïde , Humains , Protéines proto-oncogènes B-raf/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Tumeurs de la thyroïde/traitement médicamenteux , Tumeurs de la thyroïde/génétique , Tumeurs de la thyroïde/métabolisme , Mutation , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Sérine-thréonine kinases TOR , Lignée cellulaire tumorale , Protéine p120 d'activation de la ras GTPase/génétique
15.
Actas dermo-sifiliogr. (Ed. impr.) ; 115(1): 48-55, jan. 2024. graf, tab
Article de Espagnol | IBECS | ID: ibc-229338

RÉSUMÉ

El paciente con melanoma avanzado, metastásico o de alto riesgo, cuenta con opciones de tratamiento sistémico, inmunoterapia y terapias dirigidas, que han mejorado significativamente su supervivencia. El 50% de los pacientes con melanoma presentan mutación del gen BRAF. La toma de decisiones en cuanto a la secuencia óptima de tratamiento sistémico debe tener en cuenta factores relacionados con el medicamento, factores clínicos del paciente, así como los propios del tumor. Aunque la combinación ipilimumab-nivolumab es la que proporciona mejores resultados de supervivencia en todos los pacientes, la toxicidad asociada y el perfil de las terapias diana las puede hacer recomendables como primera línea en pacientes en determinadas situaciones clínicas. El objetivo de esta revisión es proporcionar un algoritmo de toma de decisiones en cuanto a la primera línea de tratamiento sistémico, inmunoterapia vs. terapias dirigidas, en el paciente con melanoma avanzado con mutación BRAF (AU)


Systemic treatment with immunotherapy or targeted therapy can significantly improve survival in patients with advanced (metastatic or high-risk) melanoma. Fifty percent of patients with melanoma have a BRAF mutation. Decisions on optimal sequencing of systemic treatments should take into account drug- and tumor-related factors and patient characteristics. Although the combination of ipilimumab and nivolumab is associated with the best survival outcomes, it is associated with significant toxicity. Targeted therapy may be a more favorable option in certain clinical situations. We review the literature on immunotherapy and targeted therapy in melanoma and present an algorithm for guiding decision-making on their use as first-line systemic treatments for advanced BRAF-mutated melanoma (AU)


Sujet(s)
Humains , Immunothérapie/méthodes , Mélanome/traitement médicamenteux , Mélanome/génétique , Thérapie moléculaire ciblée , Nivolumab/usage thérapeutique , Tumeurs cutanées/traitement médicamenteux , Tumeurs cutanées/génétique , Mutation
16.
Actas dermo-sifiliogr. (Ed. impr.) ; 115(1): t48-t55, jan. 2024. ilus, tab, graf
Article de Anglais | IBECS | ID: ibc-229340

RÉSUMÉ

Systemic treatment with immunotherapy or targeted therapy can significantly improve survival in patients with advanced (metastatic or high-risk) melanoma. Fifty percent of patients with melanoma have a BRAF mutation. Decisions on optimal sequencing of systemic treatments should take into account drug- and tumor-related factors and patient characteristics. Although the combination of ipilimumab and nivolumab is associated with the best survival outcomes, it is associated with significant toxicity. Targeted therapy may be a more favorable option in certain clinical situations. We review the literature on immunotherapy and targeted therapy in melanoma and present an algorithm for guiding decision-making on their use as first-line systemic treatments for advanced BRAF-mutated melanoma (AU)


El paciente con melanoma avanzado, metastásico o de alto riesgo, cuenta con opciones de tratamiento sistémico, inmunoterapia y terapias dirigidas, que han mejorado significativamente su supervivencia. El 50% de los pacientes con melanoma presentan mutación del gen BRAF. La toma de decisiones en cuanto a la secuencia óptima de tratamiento sistémico debe tener en cuenta factores relacionados con el medicamento, factores clínicos del paciente, así como los propios del tumor. Aunque la combinación ipilimumab-nivolumab es la que proporciona mejores resultados de supervivencia en todos los pacientes, la toxicidad asociada y el perfil de las terapias diana las puede hacer recomendables como primera línea en pacientes en determinadas situaciones clínicas. El objetivo de esta revisión es proporcionar un algoritmo de toma de decisiones en cuanto a la primera línea de tratamiento sistémico, inmunoterapia vs. terapias dirigidas, en el paciente con melanoma avanzado con mutación BRAF (AU)


Sujet(s)
Humains , Immunothérapie/méthodes , Mélanome/traitement médicamenteux , Mélanome/génétique , Thérapie moléculaire ciblée , Nivolumab/usage thérapeutique , Tumeurs cutanées/traitement médicamenteux , Tumeurs cutanées/génétique , Mutation
17.
Actas Dermosifiliogr ; 115(1): 48-55, 2024 Jan.
Article de Anglais, Espagnol | MEDLINE | ID: mdl-37321549

RÉSUMÉ

Systemic treatment with immunotherapy or targeted therapy can significantly improve survival in patients with advanced (metastatic or high-risk) melanoma. Fifty percent of patients with melanoma have a BRAF mutation. Decisions on optimal sequencing of systemic treatments should take into account drug- and tumor-related factors and patient characteristics. Although the combination of ipilimumab and nivolumab is associated with the best survival outcomes, it is associated with significant toxicity. Targeted therapy may be a more favorable option in certain clinical situations. We review the literature on immunotherapy and targeted therapy in melanoma and present an algorithm for guiding decision-making on their use as first-line systemic treatments for advanced BRAF-mutated melanoma.


Sujet(s)
Mélanome , Tumeurs cutanées , Humains , Mélanome/traitement médicamenteux , Mélanome/génétique , Protéines proto-oncogènes B-raf/génétique , Nivolumab/usage thérapeutique , Nivolumab/génétique , Immunothérapie , Mutation , Tumeurs cutanées/traitement médicamenteux , Tumeurs cutanées/génétique , Thérapie moléculaire ciblée , Inhibiteurs de protéines kinases/usage thérapeutique
18.
Actas Dermosifiliogr ; 115(1): T48-T55, 2024 Jan.
Article de Anglais, Espagnol | MEDLINE | ID: mdl-37923078

RÉSUMÉ

Systemic treatment with immunotherapy or targeted therapy can significantly improve survival in patients with advanced (metastatic or high-risk) melanoma. Fifty percent of patients with melanoma have a BRAF mutation. Decisions on optimal sequencing of systemic treatments should take into account drug- and tumor-related factors and patient characteristics. Although the combination of ipilimumab and nivolumab is associated with the best survival outcomes, it is associated with significant toxicity. Targeted therapy may be a more favorable option in certain clinical situations. We review the literature on immunotherapy and targeted therapy in melanoma and present an algorithm for guiding decision-making on their use as first-line systemic treatments for advanced BRAF-mutated melanoma.


Sujet(s)
Mélanome , Tumeurs cutanées , Humains , Mélanome/traitement médicamenteux , Mélanome/génétique , Protéines proto-oncogènes B-raf/génétique , Nivolumab/usage thérapeutique , Nivolumab/génétique , Immunothérapie , Mutation , Tumeurs cutanées/traitement médicamenteux , Tumeurs cutanées/génétique , Thérapie moléculaire ciblée
19.
Eur J Cancer ; 196: 113425, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-38039778

RÉSUMÉ

BACKGROUND: Targeted therapy (TT) of BRAF V600 mutated unresectable melanoma with inhibitors of the MAPK pathway achieves response rates of up to 76%, but most patients develop secondary resistance. Albeit TT is strikingly efficacious during the first days of treatment, even in advanced cases, long-term survival is highly unlikely, especially in patients with unfavorable baseline characteristics like elevated lactate dehydrogenase (LDH). In patients treated with anti-PD-1 immune checkpoint inhibitors, elevated baseline metastatic growth rate (MGR) was the most important prognostic factor. Here, we aimed at investigating the prognostic impact of MGR in patients with unresectable melanoma receiving TT. METHODS: Clinical records of 242 patients with at least one measurable target lesion (TL) receiving TT at seven skin cancer centers were reviewed. Baseline MGR was determined measuring the largest TL at baseline and at one earlier timepoint. RESULTS: Overall survival (OS) and progression-free survival (PFS) were significantly impaired in patients with an MGR > 3.9 mm/month (median OS: 11.4 vs. 35.5 months, P < 0.0001; median PFS: 4.8 vs. 9.2 months, P < 0.0001). Multivariable analysis of OS and PFS revealed that the prognostic impact of elevated MGR was independent of LDH, presence of brain and liver metastases, tumor burden, and line of treatment. The prognostic significance of elevated MGR was highest in patients with normal LDH. CONCLUSIONS: Baseline MGR is an important independent prognostic marker for OS and PFS in melanoma patients treated with TT. Its implementation in clinical routine is easy and could facilitate the prognostic stratification.


Sujet(s)
Mélanome , Tumeurs cutanées , Humains , Mélanome/traitement médicamenteux , Mélanome/génétique , Mélanome/métabolisme , Protéines proto-oncogènes B-raf/génétique , Pronostic , Tumeurs cutanées/traitement médicamenteux , Tumeurs cutanées/génétique , Tumeurs cutanées/anatomopathologie , Survie sans progression , Études rétrospectives , Mutation
20.
Pathol Oncol Res ; 29: 1611378, 2023.
Article de Anglais | MEDLINE | ID: mdl-38025907

RÉSUMÉ

Introduction: Hairy cell leukemia (HCL) is an indolent B-cell lymphoproliferative disease. BRAF V600E mutation is detected in nearly all classical HCL cases which offers the possibility of targeted therapy. Objective: The aim of our study was to assess the efficacy of low-dose vemurafenib as well as to assess the long term outcome of HCL patients treated with this drug at the Department of Internal Medicine and Oncology at Semmelweis University. Methods: We report on 10 patients with classical HCL treated with low-dose vemurafenib at our Department between 2013 and 2022. Results: As a result of fixed time low-dose vemurafenib treatment, 5 of 10 patients (5/10) achieved partial remission, 4 (4/10) had stable disease, and 1 (1/10) had MRD positivity. No patients achieved complete remission. The median progression-free survival was 28.5 months while the overall survival was 82 months. Conclusion: We confirm that low dose of vemurafenib is effective and safe in the vast majority of patients with HCL. This small-molecule oral treatment allows to gain valuable time-months or even years-before further, usually parenteral treatment options have to be given or before previous treatment has to be repeated. There are also promising data supporting the combination of vemurafenib with other drugs for the treatment of HCL patients which could provide even further possibility to bridge treatment.


Sujet(s)
Antinéoplasiques , Leucémie à tricholeucocytes , Humains , Vémurafénib/usage thérapeutique , Leucémie à tricholeucocytes/traitement médicamenteux , Leucémie à tricholeucocytes/génétique , Antinéoplasiques/usage thérapeutique , Études de suivi , Universités , Protéines proto-oncogènes B-raf/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...