Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39.480
Filtrar
1.
J Cell Biol ; 222(11)2023 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-37733372

RESUMO

Melanoma is an aggressive cancer typically arising from transformation of melanocytes residing in the basal layer of the epidermis, where they are in direct contact with surrounding keratinocytes. The role of keratinocytes in shaping the melanoma tumor microenvironment remains understudied. We previously showed that temporary loss of the keratinocyte-specific cadherin, Desmoglein 1 (Dsg1), controls paracrine signaling between normal melanocytes and keratinocytes to stimulate the protective tanning response. Here, we provide evidence that melanoma cells hijack this intercellular communication by secreting factors that keep Dsg1 expression low in the surrounding keratinocytes, which in turn generate their own paracrine signals that enhance melanoma spread through CXCL1/CXCR2 signaling. Evidence suggests a model whereby paracrine signaling from melanoma cells increases levels of the transcriptional repressor Slug, and consequently decreases expression of the Dsg1 transcriptional activator Grhl1. Together, these data support the idea that paracrine crosstalk between melanoma cells and keratinocytes resulting in chronic keratinocyte Dsg1 reduction contributes to melanoma cell movement associated with tumor progression.


Assuntos
Desmogleína 1 , Queratinócitos , Melanoma , Humanos , Movimento Celular , Desmogleína 1/genética , Epiderme , Melanoma/genética , Melanoma/patologia , Microambiente Tumoral/genética
2.
Acta Oncol ; 62(8): 880-888, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37656769

RESUMO

BACKGROUND: Lymphedema is a chronic, debilitating disease that often requires life-long management. Predicting clinical manifestations and prognosis is crucial in clinical practice because the treatment of lymphedema should be individualized for best clinical outcome. The aim of this study is to explore the location and severity of lymphedema secondary to inguinal and/or iliac lymph node dissection (LND) in patients with melanoma. METHODS: Patients with melanoma who received LND at a single tertiary medical center between 1 January 2010 and 31 September 2022 were retrospectively reviewed. Patient who received inguinal LND only were designate as the inguinal group while those who received both ilioinguinal LND were included in the ilioinguinal group. Volumetric measurement was used to objectify the severity and location of lymphedema. Clinical data was acquired for 12-15 months of follow-up. RESULTS: Among 81 patients, 43 (53%) had developed lymphedema in the lower extremities at an average of 33 days after the surgery. Initially, patients manifested with medial thigh lymphedema in the inguinal group while patients were presented with whole leg lymphedema in the ilioinguinal group. Lower leg volume of the ilioinguinal group was significantly higher than the inguinal group. After more than 12 months of lymphedema treatment, upper leg volume was higher in the ilioinguinal group than the inguinal group (12.7% vs 5.4%, p < 0.05). CONCLUSION: Lymphedema developed in early post-op period. The ilioinguinal group presented with a larger volume of lymphedema in the distal area of the legs. Even after sufficient treatment, predominant lymphedema remained in the proximal leg for the ilioinguinal group. Patients with both inguinal and iliac LND were associated with more severe lymphedema. Based on the dissection sites, the clinical manifestations and prognosis of leg lymphedema can vary widely. Thus, clinicians should consider the dissection site when approaching melanoma patients with lymphedema.


Assuntos
Linfedema , Melanoma , Neoplasias Cutâneas , Humanos , Neoplasias Cutâneas/cirurgia , Neoplasias Cutâneas/patologia , Estudos Retrospectivos , Metástase Linfática , Melanoma/complicações , Melanoma/cirurgia , Melanoma/patologia , Excisão de Linfonodo/efeitos adversos , Extremidade Inferior , Linfedema/etiologia
3.
Science ; 381(6664): 1316-1323, 2023 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-37733872

RESUMO

Although tumor growth requires the mitochondrial electron transport chain (ETC), the relative contribution of complex I (CI) and complex II (CII), the gatekeepers for initiating electron flow, remains unclear. In this work, we report that the loss of CII, but not that of CI, reduces melanoma tumor growth by increasing antigen presentation and T cell-mediated killing. This is driven by succinate-mediated transcriptional and epigenetic activation of major histocompatibility complex-antigen processing and presentation (MHC-APP) genes independent of interferon signaling. Furthermore, knockout of methylation-controlled J protein (MCJ), to promote electron entry preferentially through CI, provides proof of concept of ETC rewiring to achieve antitumor responses without side effects associated with an overall reduction in mitochondrial respiration in noncancer cells. Our results may hold therapeutic potential for tumors that have reduced MHC-APP expression, a common mechanism of cancer immunoevasion.


Assuntos
Antígenos de Neoplasias , Complexo II de Transporte de Elétrons , Complexo I de Transporte de Elétrons , Mitocôndrias , Neoplasias , Humanos , Apresentação de Antígeno , Antígenos de Neoplasias/imunologia , Complexo I de Transporte de Elétrons/genética , Complexo I de Transporte de Elétrons/metabolismo , Complexo II de Transporte de Elétrons/genética , Complexo II de Transporte de Elétrons/metabolismo , Elétrons , Técnicas de Inativação de Genes , Histonas/metabolismo , Proteínas de Choque Térmico HSP40/genética , Melanoma/imunologia , Melanoma/patologia , Metilação , Mitocôndrias/enzimologia , Neoplasias/imunologia , Neoplasias/patologia , Linhagem Celular Tumoral
4.
Science ; 381(6664): 1287-1288, 2023 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-37733861

RESUMO

Mitochondrial metabolite reduces melanoma growth by boosting antigen presentation.


Assuntos
Apresentação de Antígeno , Melanoma , Mitocôndrias , Humanos , Melanoma/imunologia , Melanoma/patologia , Mitocôndrias/metabolismo , Carcinogênese/imunologia , Carcinogênese/patologia
5.
Actas dermo-sifiliogr. (Ed. impr.) ; 114(7): 636-641, jul.- ago. 2023. ilus, tab
Artigo em Espanhol | IBECS | ID: ibc-223013

RESUMO

El melanoma sobre nevus azul o melanoma ex-blue nevus es una variedad de melanoma peculiar que tiene un perfil genético diferente al del resto de los melanomas cutáneos y sorprendentemente superponible al perfil del melanoma uveal. Aunque puede aparecer de novo, el melanoma ex-blue nevus se suele desarrollar sobre un nevus azul previo o sobre una melanocitosis dérmica. No todas las lesiones nodulares desarrolladas sobre un nevus azul o una melanocitosis dérmica son melanomas, y los hallazgos clínicos e histológicos pueden ser insuficientes para llegar a un diagnóstico de certeza. Así, cobran relevancia estudios adicionales, como la hibridación genómica comparada, pues la presencia de aberraciones cromosómicas favorece el diagnóstico de malignidad. Es de especial utilidad el estudio del gen BAP1, cuya pérdida de expresión orienta a melanoma en este espectro de lesiones. Presentamos 3casos del espectro nevus azul a melanoma ex-blue nevus con estudios de biología molecular (AU)


Melanoma arising in blue nevus, also known as melanoma ex blue nevus, is a specific form of melanoma whose genetic profile is different to that of other cutaneous melanomas and surprisingly similar to that of uveal melanoma. Although melanoma ex blue nevus can appear de novo, it usually arises in a preexisting blue nevus or dermal melanocytosis. Not all nodular lesions arising in association with blue nevus or dermal melanocytosis are melanomas, however, and because clinical and histologic findings may be insufficient for a definitive diagnosis, additional studies such as comparative genomic hybridization are important. Detection of chromosomal aberrations supports a diagnosis of malignancy. Studies of the BAP1 gene are particularly useful in this setting because loss of expression is indicative of melanoma. We present 3 cases on the spectrum of blue nevus to melanoma ex blue nevus that were studied using molecular biology techniques (AU)


Assuntos
Humanos , Masculino , Feminino , Pessoa de Meia-Idade , Melanoma/diagnóstico , Melanoma/genética , Nevo Azul/diagnóstico , Nevo Azul/genética , Neoplasias Cutâneas/diagnóstico , Neoplasias Cutâneas/genética , Prognóstico , Melanoma/patologia , Nevo Azul/patologia , Neoplasias Cutâneas/patologia , Proteínas Supressoras de Tumor/genética , Ubiquitina Tiolesterase/genética
6.
Actas dermo-sifiliogr. (Ed. impr.) ; 114(7): t636-t641, jul.- ago. 2023. ilus, tab
Artigo em Inglês | IBECS | ID: ibc-223014

RESUMO

Melanoma arising in blue nevus, also known as melanoma ex blue nevus, is a specific form of melanoma whose genetic profile is different to that of other cutaneous melanomas and surprisingly similar to that of uveal melanoma. Although melanoma ex blue nevus can appear de novo, it usually arises in a preexisting blue nevus or dermal melanocytosis. Not all nodular lesions arising in association with blue nevus or dermal melanocytosis are melanomas, however, and because clinical and histologic findings may be insufficient for a definitive diagnosis, additional studies such as comparative genomic hybridization are important. Detection of chromosomal aberrations supports a diagnosis of malignancy. Studies of the BAP1 gene are particularly useful in this setting because loss of expression is indicative of melanoma. We present 3 cases on the spectrum of blue nevus to melanoma ex blue nevus that were studied using molecular biology techniques (AU)


El melanoma sobre nevus azul o melanoma ex-blue nevus es una variedad de melanoma peculiar que tiene un perfil genético diferente al del resto de los melanomas cutáneos y sorprendentemente superponible al perfil del melanoma uveal. Aunque puede aparecer de novo, el melanoma ex-blue nevus se suele desarrollar sobre un nevus azul previo o sobre una melanocitosis dérmica. No todas las lesiones nodulares desarrolladas sobre un nevus azul o una melanocitosis dérmica son melanomas, y los hallazgos clínicos e histológicos pueden ser insuficientes para llegar a un diagnóstico de certeza. Así, cobran relevancia estudios adicionales, como la hibridación genómica comparada, pues la presencia de aberraciones cromosómicas favorece el diagnóstico de malignidad. Es de especial utilidad el estudio del gen BAP1, cuya pérdida de expresión orienta a melanoma en este espectro de lesiones. Presentamos 3casos del espectro nevus azul a melanoma ex-blue nevus con estudios de biología molecular (AU)


Assuntos
Humanos , Masculino , Feminino , Pessoa de Meia-Idade , Melanoma/diagnóstico , Melanoma/genética , Nevo Azul/diagnóstico , Nevo Azul/genética , Neoplasias Cutâneas/diagnóstico , Neoplasias Cutâneas/genética , Prognóstico , Melanoma/patologia , Nevo Azul/patologia , Neoplasias Cutâneas/patologia , Proteínas Supressoras de Tumor/genética , Ubiquitina Tiolesterase/genética
8.
Int J Mol Sci ; 24(15)2023 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-37569660

RESUMO

The Food and Drug Administration (FDA) has approved MAPK inhibitors as a treatment for melanoma patients carrying a mutation in codon V600 of the BRAF gene exclusively. However, BRAF mutations outside the V600 codon may occur in a small percentage of melanomas. Although these rare variants may cause B-RAF activation, their predictive response to B-RAF inhibitor treatments is still poorly understood. We exploited an integrated approach for mutation detection, tumor evolution tracking, and assessment of response to treatment in a metastatic melanoma patient carrying the rare p.T599dup B-RAF mutation. He was addressed to Dabrafenib/Trametinib targeted therapy, showing an initial dramatic response. In parallel, in-silico ligand-based homology modeling was set up and performed on this and an additional B-RAF rare variant (p.A598_T599insV) to unveil and justify the success of the B-RAF inhibitory activity of Dabrafenib, showing that it could adeptly bind both these variants in a similar manner to how it binds and inhibits the V600E mutant. These findings open up the possibility of broadening the spectrum of BRAF inhibitor-sensitive mutations beyond mutations at codon V600, suggesting that B-RAF V600 WT melanomas should undergo more specific investigations before ruling out the possibility of targeted therapy.


Assuntos
Melanoma , Neoplasias Cutâneas , Masculino , Humanos , Proteínas Proto-Oncogênicas B-raf/genética , Melanoma/tratamento farmacológico , Melanoma/genética , Melanoma/patologia , Imidazóis/farmacologia , Imidazóis/uso terapêutico , Oximas/farmacologia , Oximas/uso terapêutico , Mutação , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Piridonas/uso terapêutico , Pirimidinonas/uso terapêutico , Neoplasias Cutâneas/patologia
9.
Int J Mol Sci ; 24(15)2023 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-37569757

RESUMO

Pembrolizumab has been shown to increase survival in patients with metastatic melanoma. Considering the numerous oncoming studies, we decided to conduct a narrative review of the latest efficacy evidence regarding the use of pembrolizumab, alone or in combination, in patients with metastatic melanoma. A search was conducted in PubMed using "pembrolizumab," and "metastatic melanoma" as keywords, considering studies from 2022 onward. We reviewed pembrolizumab and associations, cost-effectiveness, virus, advanced acral melanoma, long-term outcomes, real-life data, biomarkers, obesity, and vaccines. In conclusion, pembrolizumab is a fundamental option in the therapy of metastatic melanoma. However, a certain group of patients do not respond and, therefore, new combination options need to be evaluated. In particular, the use of vaccines tailored to tumor epitopes could represent a breakthrough in the treatment of resistant forms. Further studies with larger sample numbers are needed to confirm the preliminary results.


Assuntos
Melanoma , Segunda Neoplasia Primária , Neoplasias Cutâneas , Humanos , Anticorpos Monoclonais Humanizados/uso terapêutico , Melanoma/patologia , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/induzido quimicamente
10.
Surgery ; 173(3): 626-632, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-37534705

RESUMO

BACKGROUND: Variability in guideline compliance for melanoma lymph node surgery is partially attributable to controversy about patient selection. Prior data has indicated suboptimal practice of sentinel lymph node biopsy and undertreatment of clinically node-positive disease, predating Multicenter Selective Lymphadenectomy Trial II publication. To minimize bias, we studied compliance with lymph node surgery guidelines in T2/T3 (intermediate-thickness) melanoma patients, where the greatest agreement exists. METHODS: T2/T3 and metastasis 0 melanoma cases were identified from 2004 to 2018 Surveillance, Epidemiology, and End Results data. Analysis used Cochran-Armitage test for trends, multivariable logistic regression, and Kaplan-Meier survival estimates. RESULTS: Of 66,319 eligible T2/T3 patients, 57,211 were clinically node negative; 2,191 were clinically node positive; 6,197 were clinical node unreported; and 19,044/66,319 (28.8%) had no lymph node surgery. Among clinically node-negative patients, 36,433 (63.7%) underwent sentinel lymph node biopsy and 31,026 (85.2%) were pathologically node negative; 1,499 clinically node-positive patients (68.4%) had a lymph node dissection. Lymph node dissection rates declined from 2004 to 2018, 79.8% to 32.0% for clinically node-negative/pathologically node-positive patients and 80.4% to 61.2% for clinically node-positive/pathologically node-positive patients (both P < .0001). For clinically node-negative patients, lymph node surgery compliance improved from 63.7% (2004) to 70.4% (2018) (P < .0001). Compliance correlated with younger age, male sex, tumor mitotic rate, and site (extremity > trunk/head/neck) in multivariable analysis and improved 5-year cancer-specific survival (90.0% vs 83.4%) (all P < .0001). CONCLUSIONS: Despite clear guidelines, one-third of intermediate-thickness melanoma patients in a recent cohort did not have recommended lymph node surgery. Lymph node status is a key determinant of the relative benefit of adjuvant systemic therapy and the need for active surveillance of pathologically node-positive/clinically node-negative patients. These data highlighted a clinical care gap. Efforts to improve guideline compliance are a logical strategy to improve cancer outcomes for intermediate-thickness melanoma patients.


Assuntos
Melanoma , Neoplasias Cutâneas , Humanos , Masculino , Neoplasias Cutâneas/epidemiologia , Neoplasias Cutâneas/cirurgia , Benchmarking , Melanoma/epidemiologia , Melanoma/cirurgia , Melanoma/patologia , Biópsia de Linfonodo Sentinela , Excisão de Linfonodo
12.
ACS Nano ; 17(16): 15590-15604, 2023 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-37530430

RESUMO

Because of the distinguished properties between nanovaccine and traditional vaccine, the precise guidelines for nanovaccines with an optimal vaccination strategy to induce ideal immunities are greatly desired for combating major diseases, including cancer and infections. Herein, we designed and synthesized a self-navigating nanoadjuvant composed of Fe-doped manganese carbonate and its nanovaccine via a facile method. First, the degradation of the nanoadjuvant under acidic milieu of immune cells in lymph nodes would generate T1 and T2 MR imaging (MRI) signals to reflect the transformation dynamics of the nanovaccine and inform us when the next vaccination needed. Under this guideline, nanovaccines with a precise vaccination strategy triggered robust antigen-specific immune responses and immunological memory to effectively prevent ovalbumin (OVA)-expressing melanoma relapse by activating dendritic cells via a stimulator of interferon genes (STING) signaling pathway and inducing antigen cross-presentation by shaping lysosome integrity with CO2 generation and upregulating transporter associated antigen processing 1 (TAP-1) transporter. This study provides a universal nanoadjuvant with imaging self-guidance, immunopotentiating, and cross-priming activities for developing precise vaccines with an optimal immunization strategy to combat major diseases.


Assuntos
Vacinas Anticâncer , Melanoma , Nanopartículas , Neoplasias , Vacinas , Humanos , Animais , Camundongos , Apresentação de Antígeno , Melanoma/patologia , Neoplasias/metabolismo , Vacinação , Imageamento por Ressonância Magnética , Células Dendríticas , Imunoterapia/métodos , Camundongos Endogâmicos C57BL , Nanopartículas/química
14.
J Neurooncol ; 164(1): 199-209, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37552363

RESUMO

PURPOSE: We aimed to assess the outcomes and patterns of toxicity in patients with melanoma brain metastases (MBM) treated with stereotactic radiosurgery (SRS) with or without immunotherapy (IO). METHODS: From a prospective registry, we reviewed MBM patients treated with single fraction Gamma Knife SRS between 2008 and 2021 at our center. We recorded all systemic therapies (chemotherapy, targeted therapy, or immunotherapy) administered before, during, or after SRS. Patients with prior brain surgery were excluded. We captured adverse events following SRS, including intralesional hemorrhage (IH), radiation necrosis (RN) and local failure (LF), as well as extracranial disease status. Distant brain failure (DBF), extracranial progression-free survival (PFS) and overall survival (OS) were determined using a cumulative Incidence function and the Kaplan-Meier method. RESULTS: Our analysis included 165 patients with 570 SRS-treated MBM. Median OS for patients who received IO was 1.41 years versus 0.79 years in patients who did not (p = 0.04). Ipilimumab monotherapy was the most frequent IO regimen (30%). In the absence of IO, the cumulative incidence of symptomatic (grade 2 +) RN was 3% at 24 months and remained unchanged with respect to the type or timing of IO. The incidence of post-SRS g2 + IH in patients who did not receive systemic therapy was 19% at 1- and 2 years compared to 7% at 1- and 2 years among patients who did (HR: 0.33, 95% CI 0.11-0.98; p = 0.046). Overall, neither timing nor type of IO correlated to rates of DBF, OS, or LF. Among patients treated with IO, the median time to extracranial PFS was 5.4 months (95% IC 3.2 - 9.1). CONCLUSION: The risk of g2 + IH exceeds that of g2 + RN in MBM patients undergoing SRS, with or without IO. IH should be considered a critical adverse event following MBM treatments.


Assuntos
Neoplasias Encefálicas , Melanoma , Lesões por Radiação , Radiocirurgia , Humanos , Neoplasias Encefálicas/radioterapia , Neoplasias Encefálicas/tratamento farmacológico , Hemorragia/complicações , Hemorragia/cirurgia , Melanoma/patologia , Necrose/etiologia , Lesões por Radiação/epidemiologia , Lesões por Radiação/etiologia , Lesões por Radiação/cirurgia , Radiocirurgia/efeitos adversos , Radiocirurgia/métodos , Estudos Retrospectivos , Resultado do Tratamento
16.
Med Oncol ; 40(9): 262, 2023 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-37544953

RESUMO

Melanoma is the most lethal malignancy in skin cancers. About 97,610 new cases of melanoma are projected to occur in the United States (US) in 2023. Artichoke is a very popular plant widely consumed in the US due to its nutrition. In recent years, it has been shown that artichoke shows powerful anti-cancer effects on cancers such as breast cancer, colon cancer, liver cancer, and leukemia. However, there is little known about its effect on melanoma. This study was designed to investigate if artichoke extract (AE) has any direct effect on the growth of melanoma. Clonogenic survival assay, cell proliferation, and caspase-3 activity kits were used to evaluate the effects AE has on cell survival, proliferation, and apoptosis of the widely studied melanoma cell line HTB-72. We further investigated the possible molecular mechanisms using RT-PCR and immunohistochemical staining. The percentage of colonies of HTB-72 melanoma cells decreased significantly after treated with AE. This was paralleled with the decrease in the optic density (OD) value of cancer cells after treatment with AE. This was further supported by the decreased expression of PCNA mRNA after treated with AE. Furthermore, the cellular caspase-3 activity increased after treated with AE. The anti-proliferative effect of AE on melanoma cells correlated with increased p21, p27, and decreased CDK4. The pro-apoptotic effect of AE on melanoma cells correlated with decreased survivin. Artichoke inhibits growth of melanoma by inhibition of proliferation and promotion of apoptosis. Such a study might be helpful to develop a new promising treatment for melanoma.


Assuntos
Cynara scolymus , Melanoma , Humanos , Cynara scolymus/metabolismo , Caspase 3/metabolismo , Inibidores do Crescimento/farmacologia , Linhagem Celular Tumoral , Melanoma/tratamento farmacológico , Melanoma/patologia , Apoptose , Proliferação de Células
17.
Pathol Res Pract ; 249: 154734, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37573619

RESUMO

Clinicians and dermatologists are challenged by accurate diagnosis of melanocytic lesions, due to melanoma's resemblance to benign skin conditions. Several methodologies have been proposed to diagnose melanoma, and to differentiate between a cancerous and a benign skin condition. First, the ABCD rule and Menzies method use skin lesion characteristics to interpret the condition. The 7-point checklist, 3-point checklist, and CASH algorithm are score-based methods. Each of these methods attributes a score point to the features found on the skin lesion. Furthermore, reflectance confocal microscopy (RCM), an integrated clinical and dermoscopic risk scoring system (iDscore), and a deep convoluted neural network (DCNN) also aids in diagnosis. RCM optically sections live tissues to reveal morphological and cellular structures. The skin lesion's clinical parameters determine iDscore's score point system. The DCNN model is based on a detailed learning algorithm. Therefore, we discuss the conventional and new methodologies for the identification of skin diseases. Moreover, our review attempts to provide clinicians with a comprehensible summary of the wide range of techniques that can help differentiate between early melanomas and melanocytic nevi.


Assuntos
Melanoma , Nevo Pigmentado , Neoplasias Cutâneas , Humanos , Neoplasias Cutâneas/diagnóstico , Neoplasias Cutâneas/patologia , Diagnóstico Diferencial , Dermoscopia/métodos , Melanoma/diagnóstico , Melanoma/patologia , Nevo Pigmentado/diagnóstico , Nevo Pigmentado/patologia , Microscopia Confocal/métodos
18.
Neoplasia ; 43: 100924, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37562257

RESUMO

Cutaneous melanoma is the deadliest form of skin neoplasm and its high mortality rates could be averted by early accurate detection. While the detection of melanoma is currently reliant upon melanin visualisation, research into melanosome biogenesis, as a key driver of pathogenesis, has not yielded technology that can reliably distinguish between atypical benign, amelanotic and melanotic lesions. The endosomal-lysosomal system has important regulatory roles in cancer cell biology, including a specific functional role in melanosome biogenesis. Herein, the involvement of the endosomal-lysosomal system in melanoma was examined by pooled secondary analysis of existing gene expression datasets. A set of differentially expressed endosomal-lysosomal genes was identified in melanoma, which were interconnected by biological function. To illustrate the protein expression of the dysregulated genes, immunohistochemistry was performed on samples from patients with cutaneous melanoma to reveal candidate markers. This study demonstrated the dysregulation of Syntenin-1, Sortilin and Rab25 may provide a differentiating feature between cutaneous melanoma and squamous cell carcinoma, while IGF2R may indicate malignant propensity in these skin cancers.


Assuntos
Carcinoma de Células Escamosas , Melanoma , Neoplasias Cutâneas , Humanos , Melanoma/patologia , Neoplasias Cutâneas/patologia , Carcinoma de Células Escamosas/patologia , Lisossomos/genética , Lisossomos/patologia , Proteínas rab de Ligação ao GTP
19.
Melanoma Res ; 33(5): 406-416, 2023 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-37534686

RESUMO

For patients with locally advanced or metastatic melanoma who have BRAF V600 activating mutations, combination therapy with BRAF and MEK inhibitors is now the standard of care. The combination of encorafenib, a highly selective adenosine triphosphate-competitive BRAF inhibitor, plus binimetinib, a potent, selective, allosteric, non-adenosine triphosphate-competitive MEK1/2 inhibitor, was approved by the US Food and Drug Administration for unresectable or metastatic melanoma with BRAF V600E or V600K mutations based on data from the phase III COLUMBUS study (NCT01909453). Clinical data evaluating BRAF and MEK inhibitor combinations in advanced melanoma indicate a specific profile of adverse events that includes serious retinopathy, skin disorders, and cardiovascular toxicities. Here we provide an overview of the rationale for combining BRAF and MEK inhibitors for the treatment of melanoma, long-term safety results from COLUMBUS, and guidance on managing the most common adverse events associated with this combination based on clinical experience. Proactive and appropriate management of adverse events can allow for longer treatment durations and may result in better treatment outcomes.


Assuntos
Melanoma , Neoplasias Cutâneas , Humanos , Melanoma/tratamento farmacológico , Melanoma/genética , Melanoma/patologia , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/patologia , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Inibidores de Proteínas Quinases/efeitos adversos , Mutação , Quinases de Proteína Quinase Ativadas por Mitógeno
20.
Cancer Med ; 12(17): 17891-17900, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37551424

RESUMO

BACKGROUND: Vascular endothelial growth factor (VEGF) was initially known as vascular permeability factor and identified as a driver of tumour angiogenesis. Recently, its role in supporting an immunosuppressive tumour microenvironment was demonstrated, and anti-VEGF treatment combined with immune checkpoint blockade is currently investigated. Further, beta-adrenergic signalling as a modifier of cancer hallmarks like immune response, angiogenesis and metastasis gained increased attention during past years. METHODS: Focusing on the aspect of immunosuppression in upregulated beta-adrenergic signalling, we investigated predictive markers in patients with metastatic melanoma who received bevacizumab monotherapy, a specific VEGF-A binding antibody. We explored the expression of beta-2 adrenergic receptor (ß2-AR), interleukin 6-receptor (IL6-R), cyclooxygenase 2 (COX2) and VEGF-A by immunohistochemistry in melanoma to assess the correlation between these proteins in melanoma cells and response to treatment. RESULTS: Strong ß2-AR expression in metastases was associated with clinical benefit of bevacizumab. Furthermore, expression of the latter was positively linked to expression of VEGF-A and COX2. ß2-AR expression in melanoma metastasis appears to distinguish a subgroup of patients that might benefit from anti-VEGF treatment. CONCLUSION: Our results strengthen further exploration of anti-VEGF therapy in combination with immune checkpoint blockade in clinical studies and the investigation of ß2-AR as predictive marker.


Assuntos
Melanoma , Fator A de Crescimento do Endotélio Vascular , Humanos , Bevacizumab/uso terapêutico , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptores Adrenérgicos beta 2 , Ciclo-Oxigenase 2 , Inibidores de Checkpoint Imunológico/uso terapêutico , Melanoma/patologia , Adrenérgicos/uso terapêutico , Microambiente Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...