Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.862
Filter
1.
BMC Cancer ; 24(1): 1034, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39169294

ABSTRACT

BACKGROUND: Melanoma is a highly aggressive form of skin cancer. The existence of cancer stem cells (CSCs) and tumor immune evasion are two major causes of melanoma progression, but no effective treatment has been found at present. Astragalus polysaccharide (APS) is a principal active component derived from Astragalus membranaceus, showing anti-tumor effects in various tumors including melanoma. However, the underlying mechanism is still unclear. METHODS: The regulation of APS on self-renewal ability and CSC markers expression in melanoma stem cells (MSCs) was measured by tumor sphere formation and tumorigenicity assays, RT-qPCR, and western blot. Flow cytometry was conducted to evaluate the activation of immune system by APS in melanoma mice. Further, the mechanism was explored based on PD-L1 overexpression and knock-down B16 cells. RESULTS: APS attenuated the tumor sphere formation of MSCs in vitro as well as the tumorigenicity in vivo. It also decreased the expression of CD133, BMI1 and CD47. Based on the PD-L1 overexpression and knock-down B16 cells, it was confirmed that APS inhibited the induction of MSCs by down-regulating PD-L1 expression. Meanwhile, APS increased the infiltration of CD4+ and CD8+T cells in tumor tissues because of its inhibitory effect on PD-L1. CONCLUSIONS: APS inhibited MSC induction and overcame tumor immune evasion through reducing PD-L1 expression. This study provided compelling evidence that APS could be a prospective therapeutic agent for treating melanoma.


Subject(s)
B7-H1 Antigen , Melanoma, Experimental , Neoplastic Stem Cells , Polysaccharides , B7-H1 Antigen/metabolism , Animals , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/immunology , Mice , Polysaccharides/pharmacology , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Tumor Escape/drug effects , Cell Line, Tumor , Mice, Inbred C57BL , Humans , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Skin Neoplasms/immunology , Skin Neoplasms/metabolism , Melanoma/drug therapy , Melanoma/metabolism , Melanoma/pathology , Melanoma/immunology , Astragalus Plant/chemistry , Immune Evasion
2.
Exp Biol Med (Maywood) ; 249: 10096, 2024.
Article in English | MEDLINE | ID: mdl-39170033

ABSTRACT

The current study explores the potential of ultrasound-assisted laser therapy (USaLT) to selectively destroy melanoma cells. The technology was tested on an ex vivo melanoma model, which was established by growing melanoma cells in chicken breast tissue. Ultrasound-only and laser-only treatments were used as control groups. USaLT was able to effectively destroy melanoma cells and selectively remove 66.41% of melanoma cells in the ex vivo tumor model when an ultrasound peak negative pressure of 2 MPa was concurrently applied with a laser fluence of 28 mJ/cm2 at 532 nm optical wavelength for 5 min. The therapeutic efficiency was further improved with the use of a higher laser fluence, and the treatment depth was improved to 3.5 mm with the use of 1,064 nm laser light at a fluence of 150 mJ/cm2. None of the laser-only and ultrasound-only treatments were able to remove any melanoma cells. The treatment outcome was validated with histological analyses and photoacoustic imaging. This study opens the possibility of USaLT for melanoma that is currently treated by laser therapy, but at a much lower laser fluence level, hence improving the safety potential of laser therapy.


Subject(s)
Chickens , Melanoma , Animals , Melanoma/radiotherapy , Melanoma/pathology , Ultrasonic Therapy/methods , Laser Therapy/methods , Cell Line, Tumor , Photoacoustic Techniques/methods
3.
Cell Host Microbe ; 32(8): 1217-1218, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39146792

ABSTRACT

Fecal microbiota transplants (FMTs) recently entered the cancer therapeutics field as a method to resensitize treatment-resistant melanoma patients to immune checkpoint inhibitors (ICIs). In this issue of Cell Host & Microbe, Kim and colleagues extend its utility to other solid tumors, including esophageal and hepatocellular carcinomas.1.


Subject(s)
Fecal Microbiota Transplantation , Immunotherapy , Fecal Microbiota Transplantation/methods , Humans , Immunotherapy/methods , Immune Checkpoint Inhibitors/therapeutic use , Gastrointestinal Microbiome/immunology , Melanoma/therapy , Melanoma/immunology , Neoplasms/therapy , Neoplasms/immunology , Esophageal Neoplasms/therapy , Esophageal Neoplasms/immunology , Esophageal Neoplasms/microbiology , Carcinoma, Hepatocellular/therapy , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/microbiology , Liver Neoplasms/therapy , Liver Neoplasms/immunology , Animals , Feces/microbiology
5.
Sci Rep ; 14(1): 19561, 2024 08 22.
Article in English | MEDLINE | ID: mdl-39174596

ABSTRACT

There is a critical need to non-invasively assess the PD-L1 expression in tumors as a predictive biomarker for determining the efficacy of anti-PD-1/PD-L1 immunotherapies. Non-invasive imaging modality like positron emission tomography (PET) can be a powerful tool to assess the PD-L1 expression in the whole body including multiple metastases as a patient selection criterion for the anti-PD-1/PD-L1 immunotherapy. In this study, we synthesized B11-nanobody, B11-scFv and B11-diabody fragments from the full-length anti-PD-L1 B11 IgG. Out of the three antibody fragments, B11-diabody showed higher nM affinity towards PD-L1 antigen as compared to B11-scFv and B11-nanobody. All three antibody fragments were successfully radiolabeled with 64Cu, a PET radioisotope. For radiolabeling, the antibody fragments were first conjugated with p-SCN-Bn-NOTA followed by chelation with 64Cu. All three radiolabeled antibody fragments were found to be stable in mouse and human sera for up to 24 h. Additionally, all three [64Cu]Cu-NOTA-B11-antibody fragments were evaluated in PD-L1 negative and human PD-L1 expressing cancer cells and subcutaneous tumor models. Based on the results, [64Cu]Cu-NOTA-B11-diabody has potential to be used as a PET imaging probe for assessing PD-L1 expression in tumors as early as 4 h post-injection, allowing faster assessment compared to the full length IgG based PET imaging probe.


Subject(s)
B7-H1 Antigen , Breast Neoplasms , Positron-Emission Tomography , Positron-Emission Tomography/methods , B7-H1 Antigen/metabolism , B7-H1 Antigen/immunology , Animals , Humans , Female , Mice , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/immunology , Cell Line, Tumor , Melanoma/diagnostic imaging , Melanoma/immunology , Melanoma/metabolism , Single-Chain Antibodies/immunology , Copper Radioisotopes , Immunoglobulin Fragments/immunology
6.
Cell Commun Signal ; 22(1): 410, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39175042

ABSTRACT

BACKGROUND: BRAF (v-raf murine sarcoma viral oncogene homolog B1)/MEK (mitogen-activated protein kinase kinase) inhibitors are used for melanoma treatment. Unfortunately, patients treated with this combined therapy develop resistance to treatment quite quickly, but the mechanisms underlying this phenomenon are not yet fully understood. Here, we report and characterize two melanoma cell lines (WM9 and Hs294T) resistant to BRAF (vemurafenib) and MEK (cobimetinib) inhibitors. METHODS: Cell viability was assessed via the XTT test. The level of selected proteins as well as activation of signaling pathways were evaluated using Western blotting. The expression of the chosen genes was assessed by RT-PCR. The distribution of cell cycle phases was analyzed by flow cytometry, and confocal microscopy was used to take photos of spheroids. The composition of cytokines secreted by cells was determined using a human cytokine array. RESULTS: The resistant cells had increased survival and activation of ERK kinase in the presence of BRAF/MEK inhibitors. The IC50 values for these cells were over 1000 times higher than for controls. Resistant cells also exhibited elevated activation of AKT, p38, and JNK signaling pathways with increased expression of EGFR, ErbB2, MET, and PDGFRß receptors as well as reduced expression of ErbB3 receptor. Furthermore, these cells demonstrated increased expression of genes encoding proteins involved in drug transport and metabolism. Resistant cells also exhibited features of epithelial-mesenchymal transition and cancer stem cells as well as reduced proliferation rate and elevated cytokine secretion. CONCLUSIONS: In summary, this work describes BRAF/MEK-inhibitor-resistant melanoma cells, allowing for better understanding the underlying mechanisms of resistance. The results may thus contribute to the development of new, more effective therapeutic strategies.


Subject(s)
Azetidines , Drug Resistance, Neoplasm , Melanoma , Piperidines , Protein Kinase Inhibitors , Proto-Oncogene Proteins B-raf , Vemurafenib , Humans , Melanoma/pathology , Melanoma/genetics , Melanoma/drug therapy , Melanoma/metabolism , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/metabolism , Azetidines/pharmacology , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Protein Kinase Inhibitors/pharmacology , Piperidines/pharmacology , Vemurafenib/pharmacology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/metabolism , Cell Survival/drug effects
7.
Am Soc Clin Oncol Educ Book ; 44(3): e431818, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39177646

ABSTRACT

In the field of gynecologic cancer, low-grade serous ovarian cancer (LGSOC) has been poorly understood and underinvestigated until recently. Similarly, understanding of the distinct properties and therapeutic sensitivities of gynecologic melanoma and cervical neuroendocrine tumors has recently accelerated. For each of these rare cancers, we explore the epidemiology and natural history, discuss the prognosis, diagnostic testing, and contemporary molecular classification, and then deliberate existing and emerging therapeutic strategies. In LGSOC, we focus on the clinical relevance of recent molecular studies that shed light on the importance of mitogen-activated protein kinase (MAPK) pathway gene mutation and chromosome 1 copy-number change on prognosis and MEK inhibitor sensitivity. We also discuss the relative chemoresistance of this disease and the fact that attention is shifting to combinations of molecular therapies such as endocrine agents plus cyclin-dependent kinase 4/6 inhibitors or MEK inhibitors plus FAK inhibitors. Gynecologic tract melanomas harbor a lower frequency of canonical BRAF mutations, and have lower tumor mutational burden and immune cell infiltration than cutaneous melanomas (CMs). As a result, patients with this disease are less likely to respond to BRAF/MEK or immune checkpoint inhibition than patients with CM. Emerging strategies include the combination of antiangiogenic agents with immune checkpoint inhibitors and the use of adoptive cellular therapies. In cervical neuroendocrine cancer, we discuss the use of surgery in early-stage disease, and the uncertainties regarding the role of radiotherapy. We also explore the evidence for chemotherapy and emerging investigational strategies including the use of poly (ADP-ribose) polymerase inhibitors. For all situations, we explore the shared decision-making process with the patient.


Subject(s)
Melanoma , Humans , Female , Melanoma/therapy , Melanoma/genetics , Melanoma/epidemiology , Melanoma/pathology , Genital Neoplasms, Female/therapy , Genital Neoplasms, Female/pathology , Ovarian Neoplasms/therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/epidemiology , Neoplasm Grading , Molecular Targeted Therapy , Cystadenocarcinoma, Serous/therapy , Cystadenocarcinoma, Serous/pathology , Cystadenocarcinoma, Serous/genetics , Prognosis , Neuroendocrine Tumors/therapy , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/pathology , Neuroendocrine Tumors/epidemiology
8.
Arch Dermatol Res ; 316(8): 563, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39177825

ABSTRACT

Lower extremity nodular melanoma (NM) is a common malignant tumor with a poor prognosis. We aims to identify the prognostic factors and develop a nomogram model to predict overall survival (OS) in patients with lower extremity NM. A total of 746 patients with lower extremity NM were selected and randomly divided into a training set (522 cases) and a validation set (224 cases) from the Surveillance, Epidemiology, and End Results(SEER) database. The training set underwent univariate and multivariate Cox regression analyses to identify independent prognostic factors associated with patient outcomes, and to develop a nomogram model. The effectiveness of the nomogram was subsequently validated using the validation set. Multivariable Cox regression analysis of the training set indicated that age, ulceration, radiotherapy, chemotherapy, primary site of first malignant tumor, and Breslow thickness were independent variables associated with OS. In the training set, the area under the curve (AUC) of the nomogram for predicting 3-year and 5-year OS was 0.796 and 0.811, respectively. In the validation set, the AUC for predicting 3-year and 5-year OS was 0.694 and 0.702, respectively. The Harrell's C-index for the training set and validation set were 0.754 (95% CI: 0.721-0.787) and 0.670 (95% CI: 0.607-0.733), respectively. Calibration curves for both training and validation sets showed good agreement. In this study, we develop and validate a nomogram to predict OS in patients with lower extremity NM. The nomogram demonstrated reasonable reliability and clinical applicability. Nomograms are important tools assessing prognosis and aiding clinical decision-making.


Subject(s)
Lower Extremity , Melanoma , Nomograms , SEER Program , Skin Neoplasms , Humans , Melanoma/mortality , Melanoma/diagnosis , Melanoma/pathology , Melanoma/therapy , Melanoma/epidemiology , Male , Female , SEER Program/statistics & numerical data , Middle Aged , Prognosis , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Skin Neoplasms/diagnosis , Skin Neoplasms/therapy , Aged , Adult , Aged, 80 and over
9.
Diagn Pathol ; 19(1): 112, 2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39153970

ABSTRACT

The occurrence and progression of skin cutaneous melanoma (SKCM) is strongly associated with immune cells infiltrating the tumor microenvironment (TME). This study examined the expression, prognosis, and immune relevance of SIGLEC9 in SKCM using multiple online databases. Analysis of the GEPIA2 and Ualcan databases revealed that SIGLEC9 is highly expressed in SKCM, and patients with high SIGLEC9 expression had improved overall survival (OS). Furthermore, the mutation rate of SIGLEC9 in SKCM patients was found to be 5.41%, the highest observed. The expression of SIGLEC9 was positively correlated with macrophages, neutrophils and B cells, CD8 + T cells, CD4 + T cells, and dendritic cells, according to TIMER. Based on TCGA-SKCM data, we verified that high SIGLEC9 expression is closely associated with a good prognosis for SKCM patients, including overall survival, progression-free interval, and disease-specific survival. This positive prognosis could be due to the infiltration of immune cells into the TME. Additionally, our analysis of single-cell transcriptome data revealed that SIGLEC9 not only played a role in the normal skin immune microenvironment, but is also highly expressed in immune cell subpopulations of SKCM patients, regulating the immune response to tumors. Our findings suggest that the close association between SIGLEC9 and SKCM prognosis is primarily mediated by its effect on the tumor immune microenvironment.


Subject(s)
Biomarkers, Tumor , Melanoma , Skin Neoplasms , Tumor Microenvironment , Humans , Tumor Microenvironment/immunology , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Skin Neoplasms/genetics , Skin Neoplasms/mortality , Melanoma/immunology , Melanoma/genetics , Melanoma/mortality , Melanoma/pathology , Prognosis , Biomarkers, Tumor/genetics , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Sialic Acid Binding Immunoglobulin-like Lectins/genetics , Melanoma, Cutaneous Malignant , Antigens, CD/genetics , Antigens, CD/metabolism , Lymphocytes, Tumor-Infiltrating/immunology
11.
Cancer Med ; 13(16): e70087, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39166495

ABSTRACT

BACKGROUND: Immune-checkpoint inhibitors are now used more commonly in combination than monotherapy as the first-line choice in patients with unresectable advanced melanoma. Nevertheless, for cases that progressed after the initial combination therapy, the subsequent regimen option can be very difficult. Herein, we reported the efficacy and safety of a triple combination regimen in Chinese unresectable advanced melanoma patients who had poor responses to the first-line immune therapy. METHODS: We reviewed the clinical profiles of patients diagnosed with stage IIIC-IV melanoma between June 1, 2020, and September 30, 2023. The patients who failed the prior immune therapies and received anti-PD-1 mono antibody plus interferon(IFN)-alpha 1b and anlotinib hydrochloride as the second-line therapy were enrolled in the retrospective analysis. Additionally, we examined the exhaustion of T-cells using mIHC staining in available tumor samples. RESULTS: Fifty-five patients were included in this study. The median follow-up period was 13.6 months. The objective response rate evaluated by the investigators was 9.1%(1CR, 4PR). The disease control rate was 47.3%. The median overall survival was 17.6 months, and the median progression-free survival was 2.8 months. The adverse events rate of any grade was 100%. Grade 3 or 4 irAEs were observed in 29.1% of cases. Multiplex immunohistochemical staining revealed an increased trend of TIM3 expression on tumor-infiltrating T cells in patients without objective response. CONCLUSION: PD-1 monoclonal antibody plus interferon-alpha 1b plus anlotinib showed acceptable tolerability and anticancer benefits in Chinese metastatic melanoma patients as a second-line therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Indoles , Melanoma , Programmed Cell Death 1 Receptor , Quinolines , Humans , Male , Melanoma/drug therapy , Melanoma/pathology , Melanoma/mortality , Female , Middle Aged , Retrospective Studies , Indoles/therapeutic use , Indoles/administration & dosage , Indoles/adverse effects , Aged , Adult , Quinolines/therapeutic use , Quinolines/administration & dosage , Quinolines/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/adverse effects , Immune Checkpoint Inhibitors/administration & dosage , Interferon-alpha/therapeutic use , Interferon-alpha/administration & dosage , Neoplasm Staging , Treatment Outcome
12.
Acta Derm Venereol ; 104: adv40242, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39140487

ABSTRACT

Acral melanoma is a clinical subtype of melanoma with high mortality, on which research is limited in scope. This study aimed to assess incidence trends and melanoma-specific survival rates for acral melanoma in the Swedish population from 1990 to 2020.This cross-sectional study included patients with an acral melanoma diagnosis from 1990 to 2020 from the nationwide, population-based Swedish Melanoma Registry. Analyses on acral melanoma melanoma-specific survival rates were adjusted for age, sex, histopathological subtype, and tumour thickness. Clinicopathological features and melanoma-specific survival rates were compared between diagnostic periods: 1990-1999, 2000-2009, and 2010-2020, respectively. Changes in standardized incidence rates in 1996-2020 were evaluated separately for males and females. In total, 1,000 acral melanomas in 999 patients were included in the study. No significant yearly change in standardized incidence rates for either males or females was observed, even though the absolute number of cases increased. Factors such as male sex, age ≥ 70 years, and Breslow thickness > 1.0 were independently linked to lower melanoma-specific survival. The 5-year melanoma-specific survival across the studied period ranged from 75.8% to 77.9% for females, and from 62.4% to 71.7% for males.


Subject(s)
Melanoma , Registries , Skin Neoplasms , Humans , Melanoma/mortality , Melanoma/epidemiology , Melanoma/pathology , Male , Female , Skin Neoplasms/mortality , Skin Neoplasms/epidemiology , Skin Neoplasms/pathology , Incidence , Sweden/epidemiology , Aged , Middle Aged , Cross-Sectional Studies , Adult , Time Factors , Aged, 80 and over , Survival Rate , Risk Factors , Young Adult , Sex Distribution , Adolescent , Age Distribution , Sex Factors
14.
Artif Intell Med ; 155: 102934, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39088883

ABSTRACT

BACKGROUND: Melanoma is a serious risk to human health and early identification is vital for treatment success. Deep learning (DL) has the potential to detect cancer using imaging technologies and many studies provide evidence that DL algorithms can achieve high accuracy in melanoma diagnostics. OBJECTIVES: To critically assess different DL performances in diagnosing melanoma using dermatoscopic images and discuss the relationship between dermatologists and DL. METHODS: Ovid-Medline, Embase, IEEE Xplore, and the Cochrane Library were systematically searched from inception until 7th December 2021. Studies that reported diagnostic DL model performances in detecting melanoma using dermatoscopic images were included if they had specific outcomes and histopathologic confirmation. Binary diagnostic accuracy data and contingency tables were extracted to analyze outcomes of interest, which included sensitivity (SEN), specificity (SPE), and area under the curve (AUC). Subgroup analyses were performed according to human-machine comparison and cooperation. The study was registered in PROSPERO, CRD42022367824. RESULTS: 2309 records were initially retrieved, of which 37 studies met our inclusion criteria, and 27 provided sufficient data for meta-analytical synthesis. The pooled SEN was 82 % (range 77-86), SPE was 87 % (range 84-90), with an AUC of 0.92 (range 0.89-0.94). Human-machine comparison had pooled AUCs of 0.87 (0.84-0.90) and 0.83 (0.79-0.86) for DL and dermatologists, respectively. Pooled AUCs were 0.90 (0.87-0.93), 0.80 (0.76-0.83), and 0.88 (0.85-0.91) for DL, and junior and senior dermatologists, respectively. Analyses of human-machine cooperation were 0.88 (0.85-0.91) for DL, 0.76 (0.72-0.79) for unassisted, and 0.87 (0.84-0.90) for DL-assisted dermatologists. CONCLUSIONS: Evidence suggests that DL algorithms are as accurate as senior dermatologists in melanoma diagnostics. Therefore, DL could be used to support dermatologists in diagnostic decision-making. Although, further high-quality, large-scale multicenter studies are required to address the specific challenges associated with medical AI-based diagnostics.


Subject(s)
Deep Learning , Dermoscopy , Melanoma , Skin Neoplasms , Humans , Melanoma/diagnostic imaging , Melanoma/diagnosis , Dermoscopy/methods , Skin Neoplasms/diagnostic imaging , Skin Neoplasms/pathology , Skin Neoplasms/diagnosis , Algorithms
15.
Cell ; 187(16): 4373-4388.e15, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39121849

ABSTRACT

Relatlimab (rela; anti-LAG-3) plus nivolumab (nivo; anti-PD-1) is safe and effective for treatment of advanced melanoma. We designed a trial (NCT03743766) where advanced melanoma patients received rela, nivo, or rela+nivo to interrogate the immunologic mechanisms of rela+nivo. Analysis of biospecimens from this ongoing trial demonstrated that rela+nivo led to enhanced capacity for CD8+ T cell receptor signaling and altered CD8+ T cell differentiation, leading to heightened cytotoxicity despite the retention of an exhaustion profile. Co-expression of cytotoxic and exhaustion signatures was driven by PRDM1, BATF, ETV7, and TOX. Effector function was upregulated in clonally expanded CD8+ T cells that emerged after rela+nivo. A rela+nivo intratumoral CD8+ T cell signature was associated with a favorable prognosis. This intratumoral rela+nivo signature was validated in peripheral blood as an elevated frequency of CD38+TIM3+CD8+ T cells. Overall, we demonstrated that cytotoxicity can be enhanced despite the retention of exhaustion signatures, which will inform future therapeutic strategies.


Subject(s)
CD8-Positive T-Lymphocytes , Lymphocyte Activation Gene 3 Protein , Melanoma , Programmed Cell Death 1 Receptor , Humans , Antigens, CD/metabolism , Antigens, CD/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Basic-Leucine Zipper Transcription Factors/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Differentiation , Cytotoxicity, Immunologic , High Mobility Group Proteins , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Lymphocyte Activation Gene 3 Protein/antagonists & inhibitors , Melanoma/immunology , Melanoma/drug therapy , Melanoma/genetics , Nivolumab/therapeutic use , Nivolumab/pharmacology , Positive Regulatory Domain I-Binding Factor 1/metabolism , Positive Regulatory Domain I-Binding Factor 1/genetics , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Signal Transduction
16.
Development ; 151(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39092608

ABSTRACT

Melanocytes evolved to produce the melanin that gives colour to our hair, eyes and skin. The melanocyte lineage also gives rise to melanoma, the most lethal form of skin cancer. The melanocyte lineage differentiates from neural crest cells during development, and most melanocytes reside in the skin and hair, where they are replenished by melanocyte stem cells. Because the molecular mechanisms necessary for melanocyte specification, migration, proliferation and differentiation are co-opted during melanoma initiation and progression, studying melanocyte development is directly relevant to human disease. Here, through the lens of advances in cellular omic and genomic technologies, we review the latest findings in melanocyte development and differentiation, and how these developmental pathways become dysregulated in disease.


Subject(s)
Cell Differentiation , Cell Lineage , Melanocytes , Melanoma , Melanocytes/metabolism , Melanocytes/cytology , Humans , Animals , Melanoma/pathology , Melanoma/metabolism , Melanoma/genetics , Neural Crest/metabolism , Cell Proliferation , Skin Neoplasms/pathology , Skin Neoplasms/metabolism , Skin Neoplasms/genetics
17.
Int J Mol Sci ; 25(15)2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39126026

ABSTRACT

Melanin is a crucial pigment in melanomagenesis. Its fluorescence in human tissue is exceedingly weak but can be detected through advanced laser spectroscopy techniques. The spectral profile of melanin fluorescence distinctively varies among melanocytes, nevomelanocytes, and melanoma cells, with melanoma cells exhibiting a notably "red" fluorescence spectrum. This characteristic enables the diagnosis of melanoma both in vivo and in histological samples. Neuromelanin, a brain pigment akin to melanin, shares similar fluorescence properties. Its fluorescence can also be quantified with high spectral resolution using the same laser spectroscopic methods. Documented fluorescence spectra of neuromelanin in histological samples from the substantia nigra substantiate these findings. Our research reveals that the spectral behavior of neuromelanin fluorescence mirrors that of melanin in melanomas. This indicates that the typical red fluorescence is likely influenced by the microenvironment around (neuro)melanin, rather than by direct pigment interactions. Our ongoing studies aim to further explore this distinctive "red" fluorescence. We have observed this red fluorescence spectrum in post-mortem measurements of melanin in benign nevus. The characteristic red spectrum is also evident here (unlike the benign nevus in vivo), suggesting that hypoxia may contribute to this phenomenon. Given the central role of hypoxia in both melanoma development and treatment, as well as in fundamental Parkinson's disease mechanisms, this study discusses strategies aimed at reinforcing the hypothesis that red fluorescence from (neuro)melanin serves as an indicator of hypoxia.


Subject(s)
Melanins , Melanoma , Spectrometry, Fluorescence , Humans , Hypoxia/metabolism , Melanins/metabolism , Melanocytes/metabolism , Melanoma/metabolism , Melanoma/pathology , Spectrometry, Fluorescence/methods
18.
J Transl Med ; 22(1): 758, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138582

ABSTRACT

BACKGROUND: While melanomas commonly harbor losses of 9p21, on which CDKN2A resides, the presence of additional tumor suppressor elements at this locus is incompletely characterized. Here we assess the expression levels and functional role of microRNA-876-3p (miR-876), whose gene also maps to 9p21. METHODS: Expression of miR-876 was assessed in human tissues and cell lines using quantitative miRNA reverse transcriptase polymerase chain reaction (qRT-PCR). MIR876 copy number was determined in The Cancer Genome Atlas (TCGA) melanoma cohort. The consequences of regulation of miR-876 expression were assessed on melanoma cell colony formation, migration, invasion, apoptosis, cell cycle progression, and drug sensitivity in culture, and on in vivo tumor growth in a xenograft model. Genome-wide transcriptomic changes induced by miR-876 overexpression were determined using RNA sequencing (RNA-Seq). RESULTS: miR-876 expression was significantly decreased in primary melanoma samples when compared with nevi, and in human melanoma cell lines when compared with human melanocytes. Analysis of the TCGA cohort revealed deletions in MIR876 in > 50% of melanomas. miR-876 overexpression resulted in decreased melanoma cell colony formation, migration, and invasion, which was accompanied by cell cycle arrest and increased apoptosis. Intra-tumoral injections of miR-876 significantly suppressed melanoma growth in vivo. RNA-Seq analysis of miR-876-treated tumors revealed downregulation of several growth-promoting genes, along with upregulation of tumor suppressor genes, which was confirmed by qRT-PCR analysis. Computational analyses identified MAPK1 (or ERK2) as a possible target of miR-876 action. Overexpression of miR-876 significantly suppressed luciferase expression driven by the MAPK1/ERK2 3' UTR, and resulted in decreased ERK protein expression in melanoma cells. MAPK1/ERK2 cDNA overexpression rescued the effects of miR-876 on melanoma colony formation. miR-876 overexpression sensitized melanoma cells to treatment with the BRAF inhibitor vemurafenib. CONCLUSIONS: These studies identify miR-876 as a distinct tumor suppressor on 9p21 that is inactivated in melanoma and suggest miR-876 loss as an additional mechanism to activate ERK and the mitogen activated protein kinase (MAPK) pathway in melanoma. In addition, they suggest the therapeutic potential of combining miR-876 overexpression with BRAF inhibition as a rational therapeutic strategy for melanoma.


Subject(s)
Chromosomes, Human, Pair 9 , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Melanoma , MicroRNAs , Humans , Melanoma/genetics , Melanoma/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Line, Tumor , Animals , Chromosomes, Human, Pair 9/genetics , Cell Movement/genetics , Apoptosis/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Cell Proliferation/genetics , Base Sequence , Neoplasm Invasiveness
19.
Skin Res Technol ; 30(8): e13905, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39138831

ABSTRACT

BACKGROUND: Several cathepsins have been identified as being involved in the development of cancer. Nevertheless, the connection between cathepsins and skin cancers remained highly elusive. METHODS: A bidirectional Mendelian randomization (MR) analysis was performed to investigate the causal association between cathepsins and skin malignancies. The genome-wide association studies (GWAS) data for cathepsins, malignant melanoma (MM), and basal cell carcinoma (BCC) were obtained from European research. The primary method employed was inverse variance weighted. In addition, MR-Egger, weighted median, weighted mode, and simple mode were also executed. Sensitivity analysis was performed using Cochran's Q test, MR-Egger, and MR-PRESSO. RESULTS: From univariable MR (UVMR), cathepsin H, and S were determined to have a causal relationship with BCC. Additionally, cathepsin H was identified as associated with MM. Multivariable MR (MVMR) showed that after correcting for risk factors of skin carcinoma, cathepsin H was detected to be protective against BCC, whereas cathepsin S has been observed as a risk factor for BCC. No substantial pleiotropy and heterogeneity were identified in the sensitivity analysis. CONCLUSION: This study was the first to establish a direct link between cathepsins and skin malignancies. Cathepsin H and S have the potential to serve as new biomarkers for BCC, offering valuable assistance in the prompt identification, treatment, and prevention of the disease. Nevertheless, additional clinical trials are required to validate our findings.


Subject(s)
Carcinoma, Basal Cell , Cathepsins , Genome-Wide Association Study , Melanoma , Mendelian Randomization Analysis , Skin Neoplasms , Humans , Skin Neoplasms/genetics , Cathepsins/genetics , Carcinoma, Basal Cell/genetics , Melanoma/genetics , Cathepsin H/genetics , Polymorphism, Single Nucleotide , Risk Factors , Genetic Predisposition to Disease/genetics
20.
JAMA Netw Open ; 7(8): e2426641, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39141388

ABSTRACT

IMPORTANCE: Long-term survival data from clinical trials show that survival curves of patients with advanced melanoma treated with immune checkpoint inhibitors (ICIs) gradually reach a plateau, suggesting that patients have a chance of achieving long-term survival. OBJECTIVE: To investigate long-term survival in patients with advanced melanoma treated with ICIs outside clinical trials. DESIGN, SETTING, AND PARTICIPANTS: Cohort study using prospectively collected data from the nationwide Dutch Melanoma Treatment Registry, including patients in the Netherlands with advanced melanoma treated with first-line ICIs from 2012 to 2019. Data were analyzed from January to September 2023. EXPOSURES: Patients were treated with first-line ipilimumab-nivolumab, antibodies that target programmed cell death (anti-PD-1), or ipilimumab. MAIN OUTCOMES AND MEASURES: Progression-free survival (PFS) and melanoma-specific survival were analyzed, and a Cox proportional hazards model was used to investigate factors associated with PFS after reaching partial response (PR) or complete response (CR). RESULTS: A total of 2490 patients treated with first-line ICIs were included (median [IQR] age, 65.0 [55.3-73.0] years; 1561 male patients [62.7%]). Most patients had an Eastern Cooperative Oncology Group Performance Status of 1 or lower (2202 patients [88.5%]) and normal lactate dehydrogenase levels (1715 patients [68.9%]). PFS for all patients was 23.4% (95% CI, 21.7%-25.2%) after 3 years and 19.7% (95% CI, 18.0%-21.4%) after 5 years. Overall survival for all patients was 44.0% (95% CI, 42.1%-46.1%) after 3 years and 35.9% (95% CI, 33.9%-38.0%) after 5 years. Patients with metastases in 3 or more organ sites had a significantly higher hazard of progression after reaching PR or CR (adjusted hazard ratio, 1.37; 95% CI, 1.11-1.69). CONCLUSIONS AND RELEVANCE: This cohort study of patients with advanced melanoma treated with ICIs in clinical practice showed that their survival reached a plateau, comparable with patients participating in clinical trials. These findings can be used in daily clinical practice to guide long-term surveillance strategies and inform both physicians and patients regarding long-term treatment outcomes.


Subject(s)
Immune Checkpoint Inhibitors , Melanoma , Humans , Melanoma/drug therapy , Melanoma/mortality , Male , Female , Middle Aged , Aged , Immune Checkpoint Inhibitors/therapeutic use , Netherlands/epidemiology , Ipilimumab/therapeutic use , Nivolumab/therapeutic use , Skin Neoplasms/drug therapy , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Cohort Studies , Registries , Progression-Free Survival , Prospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL