Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Front Immunol ; 15: 1336566, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38510242

RESUMEN

Introduction: About 50% of cutaneous melanoma (CM) patients present activating BRAF mutations that can be effectively targeted by BRAF inhibitors (BRAFi). However, 20% of CM patients exhibit intrinsic drug resistance to BRAFi, while most of the others develop adaptive resistance over time. The mechanisms involved in BRAFi resistance are disparate and globally seem to rewire the cellular signaling profile by up-regulating different receptor tyrosine kinases (RTKs), such as the epidermal growth factor receptor (EGFR). RTKs inhibitors have not clearly demonstrated anti-tumor activity in BRAFi resistant models. To overcome this issue, we wondered whether the shared up-regulated RTK phenotype associated with BRAFi resistance could be exploited by using immune weapons as the antibody-dependent cell cytotoxicity (ADCC)-mediated effect of anti-RTKs antibodies, and kill tumor cells independently from the mechanistic roots. Methods and results: By using an in vitro model of BRAFi resistance, we detected increased membrane expression of EGFR, both at mRNA and protein level in 4 out of 9 BRAFi-resistant (VR) CM cultures as compared to their parental sensitive cells. Increased EGFR phosphorylation and AKT activation were observed in the VR CM cultures. EGFR signaling appeared dispensable for maintaining resistance, since small molecule-, antibody- and CRISPR-targeting of EGFR did not restore sensitivity of VR cells to BRAFi. Importantly, immune-targeting of EGFR by the anti-EGFR antibody cetuximab efficiently and specifically killed EGFR-expressing VR CM cells, both in vitro and in humanized mouse models in vivo, triggering ADCC by healthy donors' and patients' peripheral blood cells. Conclusion: Our data demonstrate the efficacy of immune targeting of RTKs expressed by CM relapsing on BRAFi, providing the proof-of-concept supporting the assessment of anti-RTK antibodies in combination therapies in this setting. This strategy might be expected to concomitantly trigger the crosstalk of adaptive immune response leading to a complementing T cell immune rejection of tumors.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Animales , Ratones , Humanos , Melanoma/patología , Neoplasias Cutáneas/patología , Proteínas Proto-Oncogénicas B-raf , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptores ErbB , Citotoxicidad Celular Dependiente de Anticuerpos
2.
Artículo en Inglés | MEDLINE | ID: mdl-37482577

RESUMEN

The pea aphid, Acyrthosiphon pisum, is a paradigmatic photoperiodic species that exhibits a remarkable annual life cycle, which is tightly coupled to the seasonal changes in day length. During spring and summer, characterised by longer days, aphid populations consist exclusively of viviparous females that reproduce parthenogenetically. When autumn comes and the days shorten, aphids switch their reproductive mode and generate males and oviparous sexual females, which mate and produce cold-resistant eggs that overwinter and survive the unfavourable season. While the photoperiodic responses have been well described, the nature of the timing mechanisms which underlie day length discrimination are still not completely understood. Experiments from the 1960's suggested that aphids rely on an 'hourglass' clock measuring the elapsed time during the dark night by accumulating a biochemical factor, which reaches a critical threshold at a certain night length and triggers the switch in reproduction mode. However, the photoperiodic responses of aphids can also be attributed to a strongly dampened circadian clock. Recent studies have uncovered the molecular components and the location of the circadian clock in the brain of the pea aphid and revealed that it is well connected to the neurohormonal system controlling aphid reproduction. We provide an overview of the putative mechanisms of photoperiodic control in aphids, from the photoreceptors involved in this process to the circadian clock and the neuroendocrine system.

3.
Cell Death Discov ; 9(1): 202, 2023 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-37386023

RESUMEN

Macroautophagy, hereafter referred to as autophagy, represents a highly conserved catabolic process that maintains cellular homeostasis. At present, the role of autophagy in cutaneous melanoma (CM) is still controversial, since it appears to be tumor-suppressive at early stages of malignant transformation and cancer-promoting during disease progression. Interestingly, autophagy has been found to be often increased in CM harboring BRAF mutation and to impair the response to targeted therapy. In addition to autophagy, numerous studies have recently conducted in cancer to elucidate the molecular mechanisms of mitophagy, a selective form of mitochondria autophagy, and secretory autophagy, a process that facilitates unconventional cellular secretion. Although several aspects of mitophagy and secretory autophagy have been investigated in depth, their involvement in BRAF-mutant CM biology has only recently emerged. In this review, we aim to overview autophagy dysregulation in BRAF-mutant CM, along with the therapeutic advantages that may arise from combining autophagy inhibitors with targeted therapy. In addition, the recent advances on mitophagy and secretory autophagy involvement in BRAF-mutant CM will be also discussed. Finally, since a number of autophagy-related non-coding RNAs (ncRNAs) have been identified so far, we will briefly discussed recent advances linking ncRNAs to autophagy regulation in BRAF-mutant CM.

4.
Open Biol ; 13(6): 230090, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37369351

RESUMEN

The neuropeptide pigment-dispersing factor (PDF) plays a pivotal role in the circadian clock of most Ecdysozoa and is additionally involved in the timing of seasonal responses of several photoperiodic species. The pea aphid, Acyrthosiphon pisum, is a paradigmatic photoperiodic species with an annual life cycle tightly coupled to the seasonal changes in day length. Nevertheless, PDF could not be identified in A. pisum so far. In the present study, we identified a PDF-coding gene that has undergone significant changes in the otherwise highly conserved insect C-terminal amino acid sequence. A newly generated aphid-specific PDF antibody stained four neurons in each hemisphere of the aphid brain that co-express the clock protein Period and have projections to the pars lateralis that are highly plastic and change their appearance in a daily and seasonal manner, resembling those of the fruit fly PDF neurons. Most intriguingly, the PDF terminals overlap with dendrites of the insulin-like peptide (ILP) positive neurosecretory cells in the pars intercerebralis and with putative terminals of Cryptochrome (CRY) positive clock neurons. Since ILP has been previously shown to be crucial for seasonal adaptations and CRY might serve as a circadian photoreceptor vital for measuring day length, our results suggest that PDF plays a critical role in aphid seasonal timing.


Asunto(s)
Áfidos , Relojes Circadianos , Insulinas , Animales , Áfidos/genética , Áfidos/metabolismo , Ritmo Circadiano/genética , Drosophila/fisiología , Fibrinógeno/metabolismo , Insulinas/metabolismo , Neuronas/metabolismo , Pisum sativum/metabolismo , Péptidos/metabolismo
5.
Int J Radiat Oncol Biol Phys ; 115(3): 608-621, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36202181

RESUMEN

PURPOSE: The present study aimed at evaluating the baseline immune profile and the immunomodulating effects of radical hemithoracic radiation therapy (RT) in patients affected by malignant pleural mesothelioma (MPM) to identify potential predictive biomarkers of therapy response, toxicity development, and eligibility for further immunotherapeutic treatments. METHODS AND MATERIALS: Blood samples were collected from 55 patients with MPM, enrolled in a phase 3 trial comparing radical hemithoracic RT (interventional arm, n = 28) with local palliative RT (control arm, n = 27). Immunomonitoring was performed before RT, at the end of treatment, and 1 month after therapy, characterizing natural killer cells, B and T lymphocytes, activated CD4 and CD8 T cells, interferon-γ- and tumor necrosis factor-α-producing T helper (Th) 1 cells, regulatory T cells, and Th17 and Th22 lymphocytes, through flow cytometry. Serum levels of interleukin (IL)-6, -8, -10 and mesothelin were quantified through Enzyme-Linked Immunosorbent Assay (ELISA) assays at the same time points. Variations in the immune parameters were investigated by Friedman test and Wilcoxon signed rank post hoc test with Bonferroni correction for multiple testing, while the prognostic effect of immune biomarkers was evaluated through Kaplan-Meier method and Spearman's correlation analysis. RESULTS: Major immune variations were noticed after radical RT compared with palliative treatment, in particular an improvement in activated T cells and in interferon-γ-producing Th1 cells after RT. In the interventional arm, baseline high levels of Th22 and IL-10 and an increase in T cells were associated with an improved survival, whereas a fold increase in serum mesothelin correlated with the development of severe toxicity. An improvement of immunosuppressive regulatory T cells was observed in both arms of treatment. CONCLUSIONS: The immunomonitoring performed in patients with MPM revealed potential prognostic biomarkers for radical hemithoracic RT treatment and identified specific immune signatures induced by RT immunomodulation, which could suggest a synergistic effect with an immunotherapeutic treatment.


Asunto(s)
Neoplasias Pulmonares , Mesotelioma Maligno , Mesotelioma , Neoplasias Pleurales , Humanos , Mesotelina , Mesotelioma/radioterapia , Mesotelioma/patología , Interferón gamma , Neoplasias Pulmonares/patología
6.
Mol Oncol ; 16(3): 565-593, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34080276

RESUMEN

Cutaneous melanoma (CM) is a very aggressive disease, often characterized by unresponsiveness to conventional therapies and high mortality rates worldwide. The identification of the activating BRAFV600 mutations in approximately 50% of CM patients has recently fueled the development of novel small-molecule inhibitors that specifically target BRAFV600 -mutant CM. In addition, a major progress in CM treatment has been made by monoclonal antibodies that regulate the immune checkpoint inhibitors. However, although target-based therapies and immunotherapeutic strategies have yielded promising results, CM treatment remains a major challenge. In the last decade, accumulating evidence points to the aberrant expression of different types of noncoding RNAs (ncRNAs) in CM. While studies on microRNAs have grown exponentially leading to significant insights on CM biology, the role of circular RNAs (circRNAs) and long noncoding RNAs (lncRNAs) in this tumor is less understood, and much remains to be discovered. Here, we summarize and critically review the available evidence on the molecular functions of circRNAs and lncRNAs in BRAFV600 -mutant CM and CM immunogenicity, providing recent updates on their functional role in targeted therapy and immunotherapy resistance. In addition, we also include an evaluation of several algorithms and databases for prediction and validation of circRNA and lncRNA functional interactions.


Asunto(s)
Melanoma , MicroARNs , ARN Largo no Codificante , Neoplasias Cutáneas , Humanos , Melanoma/tratamiento farmacológico , Melanoma/terapia , MicroARNs/genética , MicroARNs/metabolismo , ARN Circular/genética , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/terapia
7.
Front Physiol ; 12: 705048, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34366893

RESUMEN

Circadian clocks prepare the organism to cyclic environmental changes in light, temperature, or food availability. Here, we characterized the master clock in the brain of a strongly photoperiodic insect, the aphid Acyrthosiphon pisum, immunohistochemically with antibodies against A. pisum Period (PER), Drosophila melanogaster Cryptochrome (CRY1), and crab Pigment-Dispersing Hormone (PDH). The latter antibody detects all so far known PDHs and PDFs (Pigment-Dispersing Factors), which play a dominant role in the circadian system of many arthropods. We found that, under long days, PER and CRY are expressed in a rhythmic manner in three regions of the brain: the dorsal and lateral protocerebrum and the lamina. No staining was detected with anti-PDH, suggesting that aphids lack PDF. All the CRY1-positive cells co-expressed PER and showed daily PER/CRY1 oscillations of high amplitude, while the PER oscillations of the CRY1-negative PER neurons were of considerable lower amplitude. The CRY1 oscillations were highly synchronous in all neurons, suggesting that aphid CRY1, similarly to Drosophila CRY1, is light sensitive and its oscillations are synchronized by light-dark cycles. Nevertheless, in contrast to Drosophila CRY1, aphid CRY1 was not degraded by light, but steadily increased during the day and decreased during the night. PER was always located in the nuclei of the clock neurons, while CRY was predominantly cytoplasmic and revealed the projections of the PER/CRY1-positive neurons. We traced the PER/CRY1-positive neurons through the aphid protocerebrum discovering striking similarities with the circadian clock of D. melanogaster: The CRY1 fibers innervate the dorsal and lateral protocerebrum and putatively connect the different PER-positive neurons with each other. They also run toward the pars intercerebralis, which controls hormone release via the neurohemal organ, the corpora cardiaca. In contrast to Drosophila, the CRY1-positive fibers additionally travel directly toward the corpora cardiaca and the close-by endocrine gland, corpora allata. This suggests a direct link between the circadian clock and the photoperiodic control of hormone release that can be studied in the future.

8.
Methods Mol Biol ; 2292: 73-94, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33651353

RESUMEN

The characterization of circulating tumor cells (CTCs) is now widely studied as a promising source of cancer-derived biomarkers because of their role in tumor formation and progression. However, CTCs analysis presents some limitations and no standardized method for CTCs isolation from urine has been defined so far. In fact, besides blood, urine represents an ideal source of noninvasive biomarkers, especially for the early detection of genitourinary tumors. Besides CTCs, long noncoding RNAs (lncRNAs) have also been proposed as potential noninvasive biomarkers, and the evaluation of the diagnostic accuracy of urinary lncRNAs has dramatically increased over the last years, with many studies being published. Therefore, this review provides an update on the clinical utility of urinary lncRNAs as novel biomarkers for the diagnosis of bladder and prostate cancers.


Asunto(s)
Neoplasias de la Próstata/orina , ARN Largo no Codificante/orina , Neoplasias de la Vejiga Urinaria/orina , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/orina , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/genética , ARN Largo no Codificante/genética , Neoplasias de la Vejiga Urinaria/diagnóstico , Neoplasias de la Vejiga Urinaria/genética
9.
Cell Death Dis ; 11(5): 392, 2020 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-32444628

RESUMEN

Mitogen-activated protein kinase (MAPK) pathway activation is a central step in BRAFV600-mutant cutaneous melanoma (CM) pathogenesis. In the last years, Spry1 has been frequently described as an upstream regulator of MAPK signaling pathway. However, its specific role in BRAFV600-mutant CM is still poorly defined. Here, we report that Spry1 knockdown (Spry1KO) in three BRAFV600-mutant CM cell lines markedly induced cell cycle arrest and apoptosis, repressed cell proliferation in vitro, and impaired tumor growth in vivo. Furthermore, our findings indicated that Spry1KO reduced the expression of several markers of epithelial-mesenchymal transition, such as MMP-2 both in vitro and in vivo. These effects were associated with a sustained and deleterious phosphorylation of ERK1/2. In addition, p38 activation along with an increase in basal ROS levels were found in Spry1KO clones compared to parental CM cell lines, suggesting that BRAFV600-mutant CM may restrain the activity of Spry1 to avoid oncogenic stress and to enable tumor growth. Consistent with this hypothesis, treatment with the BRAF inhibitor (BRAFi) vemurafenib down-regulated Spry1 levels in parental CM cell lines, indicating that Spry1 expression is sustained by the MAPK/ERK signaling pathway in a positive feedback loop that safeguards cells from the potentially toxic effects of ERK1/2 hyperactivation. Disruption of this feedback loop rendered Spry1KO cells more susceptible to apoptosis and markedly improved response to BRAFi both in vitro and in vivo, as a consequence of the detrimental effect of ERK1/2 hyperactivation observed upon Spry1 abrogation. Therefore, targeting Spry1 might offer a treatment strategy for BRAFV600-mutant CM by inducing the toxic effects of ERK-mediated signaling.


Asunto(s)
Melanoma/genética , Melanoma/patología , Proteínas de la Membrana/deficiencia , Fosfoproteínas/deficiencia , Proteínas Proto-Oncogénicas B-raf/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Humanos , Melanoma/tratamiento farmacológico , Proteínas de la Membrana/genética , Fosfoproteínas/genética , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/metabolismo , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología
10.
Methods Mol Biol ; 1909: 137-162, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30580429

RESUMEN

Aberrant DNA methylation of cell-free circulating DNA (cfDNA) has recently gained attention for its use as biomarker in cancer diagnosis, prognosis, and prediction of therapeutic response. Quantification of cfDNA methylation levels requires methods with high sensitivity and specificity due to low amounts of cfDNA available in plasma, high degradation of cfDNA, and/or contamination with genomic DNA. To date, several approaches for measuring cfDNA methylation have been established, including quantitative methylation-specific PCR (qMSP), which represents a simple, fast, and cost-effective technique that can be easily implemented into clinical practice. In this chapter, we provide a detailed protocol for SYBR Green qMSP analysis which is currently used in our laboratory for cfDNA methylation detection. Useful information regarding successful qMSP primers design are also provided.


Asunto(s)
Ácidos Nucleicos Libres de Células/genética , Metilación de ADN , Reacción en Cadena de la Polimerasa/métodos , Animales , Secuencia de Bases , Recolección de Muestras de Sangre/métodos , Ácidos Nucleicos Libres de Células/sangre , Ácidos Nucleicos Libres de Células/química , Cartilla de ADN/química , Cartilla de ADN/genética , Epigénesis Genética , Humanos , Sulfitos/química
11.
Clin Epigenetics ; 9: 58, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28572862

RESUMEN

BACKGROUND: Inclusion of new biomarkers to improve a personalized treatment approach for oropharyngeal squamous cell carcinoma (OPSCC) is urgently needed. Hypomethylation of the Long interspersed nucleotide element-1 (LINE-1) repetitive elements, a widely accepted surrogate of overall genomic DNA methylation content, was found to be associated with a poor prognosis in several cancers. At present, no studies have investigated the influence of LINE-1 methylation levels on OPSCC relapse. The main goal of this study was the evaluation of the prognostic value of LINE-1 methylation status in predicting early tumor relapse in locally advanced OPSCC. METHODS: We retrospectively reviewed a cohort of 77 patients with stage III-IVB OPSCC. Methylation of LINE-1 repetitive sequences was evaluated by real-time quantitative methylation-specific PCR in formalin-fixed paraffin-embedded tissues. The prognostic relevance of LINE-1 methylation was assessed by comparing patients who relapsed within 2 years from the end of treatment (cases) with those who did not (controls). Results were validated in an independent cohort of 33 patients with OPSCC. RESULTS: With respect to early OPSCC relapse, the mean LINE-1 methylation level was significantly lower in relapsed cases than in control group (p < 0.01). Interestingly, LINE-1 methylation was lower in relapsed cases than in controls in both HPV16-negative and HPV16-positive OPSCC patients, even if statistical significance was reached only for the former group (p = 0.01). LINE-1 methylation levels were also significantly reduced in relapsed cases with respect to the controls in OPSCC current smokers (p = 0.02). Consistently, in HPV16-negative current smokers, OPSCC relapse was significantly associated with decreased levels of LINE-1 methylation (p = 0.02). Using logistic regression model, we found that patients with hypomethylated LINE-1 were associated with a 3.5 higher risk of early relapse than hypermethylated ones (OR = 3.51; 95% CI 1.03-12.00). Adjustment for potential confounders did not substantially change the risk magnitude. Results from the validation cohort confirmed the lower LINE-1 methylation in patients who early relapsed compared to relapse-free patients. CONCLUSIONS: LINE-1 hypomethylation is associated with higher risk of early relapse in stage III-IVB OPSCC. Further validation in a prospective study is needed for its application in daily clinical practice.


Asunto(s)
Carcinoma de Células Escamosas/genética , Metilación de ADN , Elementos de Nucleótido Esparcido Largo , Neoplasias Orofaríngeas/genética , Carcinoma de Células Escamosas/patología , Epigénesis Genética , Femenino , Humanos , Modelos Logísticos , Masculino , Neoplasias de la Boca , Estadificación de Neoplasias , Neoplasias Orofaríngeas/patología , Pronóstico , Recurrencia
12.
Oncotarget ; 7(35): 57327-57350, 2016 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-27329599

RESUMEN

In recent years, recurrent somatic mutations in epigenetic regulators have been identified in patients with hematological malignancies. Furthermore, chromosomal translocations in which the fusion protein partners are themselves epigenetic regulators or where epigenetic regulators are recruited/targeted by oncogenic fusion proteins have also been described. Evidence has accumulated showing that "epigenetic drugs" are likely to provide clinical benefits in several hematological malignancies, granting their approval for the treatment of myelodysplastic syndromes and cutaneous T-cell lymphomas. A large number of pre-clinical and clinical trials evaluating epigenetic drugs alone or in combination therapies are ongoing. The aim of this review is to provide a comprehensive summary of known epigenetic alterations and of the current use of epigenetic drugs for the treatment of hematological malignancies.


Asunto(s)
Epigénesis Genética , Neoplasias Hematológicas/genética , Linfoma Cutáneo de Células T/genética , Síndromes Mielodisplásicos/genética , Acetilación , Antineoplásicos/uso terapéutico , Azacitidina/análogos & derivados , Azacitidina/farmacología , Metilación de ADN , Decitabina , Femenino , Histonas/química , Humanos , Linfoma Cutáneo de Células T/terapia , Masculino , Síndromes Mielodisplásicos/terapia , Dominios Proteicos
13.
Cancer Immunol Immunother ; 62(3): 605-14, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23138873

RESUMEN

PURPOSE: Pharmacologic DNA hypomethylation holds strong promises in cancer immunotherapy due to its immunomodulatory activity on neoplastic cells. Searching for more efficient DNA hypomethylating agents to be utilized to design novel immunotherapeutic strategies in cancer, we investigated the immunomodulatory properties of the new DNA hypomethylating agent SGI-110, that is resistant to in vivo inactivation by cytidine deaminase. EXPERIMENTAL DESIGN: Cutaneous melanoma, mesothelioma, renal cell carcinoma, and sarcoma cells were treated in vitro with SGI-110. RT-PCR, quantitative RT-PCR, quantitative methylation-specific PCR, and flow cytometric analyses were performed to investigate changes induced by SGI-110 in the constitutive immune profile of cancer cells. The recognition by gp100-specific CTL of gp100-positive melanoma cells, treated or not with SGI-110, was tested by LDH release assays. RESULTS: SGI-110 induced/up-regulated the expression of investigated cancer/testis antigens (CTA) (i.e., MAGE-A1, MAGE-A2, MAGE-A3, MAGE-A4, MAGE-A10, GAGE 1-2, GAGE 1-6, NY-ESO-1, and SSX 1-5) in all cancer cell lines studied, both at mRNA and at protein levels. Quantitative methylation-specific PCR analyses identified a hypomethylation of MAGE-A1 and NY-ESO-1 promoters in SGI-110-treated neoplastic cells, demonstrating a direct role of pharmacologic DNA demethylation in CTA induction. SGI-110 also up-regulated the expression of HLA class I antigens and of ICAM-1, resulting in an improved recognition of cancer cells by gp100-specific CTL. CONCLUSIONS: Our findings show that SGI-110 is a highly attractive therapeutic agent to comprehensively increase immunogenicity and immune recognition of neoplastic cells, and provide the scientific rationale for its clinical development to design novel chemo-immunotherapeutic approaches in cancer patients.


Asunto(s)
Azacitidina/análogos & derivados , Metilación de ADN , Inmunomodulación/efectos de los fármacos , Inmunoterapia/métodos , Neoplasias/inmunología , Antígenos de Neoplasias/biosíntesis , Antígenos de Neoplasias/genética , Azacitidina/farmacología , Línea Celular Tumoral , Citotoxicidad Inmunológica , Humanos , Inmunofenotipificación , Melanoma/inmunología , Neoplasias/terapia
14.
J Transl Med ; 10: 185, 2012 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-22950745

RESUMEN

BACKGROUND: The clinical course of cutaneous melanoma (CM) can differ significantly for patients with identical stages of disease, defined clinico-pathologically, and no molecular markers differentiate patients with such a diverse prognosis. This study aimed to define the prognostic value of whole genome DNA methylation profiles in stage III CM. METHODS: Genome-wide methylation profiles were evaluated by the Illumina Human Methylation 27 BeadChip assay in short-term neoplastic cell cultures from 45 stage IIIC CM patients. Unsupervised K-means partitioning clustering was exploited to sort patients into 2 groups based on their methylation profiles. Methylation patterns related to the discovered groups were determined using the nearest shrunken centroid classification algorithm. The impact of genome-wide methylation patterns on overall survival (OS) was assessed using Cox regression and Kaplan-Meier analyses. RESULTS: Unsupervised K-means partitioning by whole genome methylation profiles identified classes with significantly different OS in stage IIIC CM patients. Patients with a "favorable" methylation profile had increased OS (P = 0.001, log-rank = 10.2) by Kaplan-Meier analysis. Median OS of stage IIIC patients with a "favorable" vs. "unfavorable" methylation profile were 31.5 and 10.4 months, respectively. The 5 year OS for stage IIIC patients with a "favorable" methylation profile was 41.2% as compared to 0% for patients with an "unfavorable" methylation profile. Among the variables examined by multivariate Cox regression analysis, classification defined by methylation profile was the only predictor of OS (Hazard Ratio = 2.41, for "unfavorable" methylation profile; 95% Confidence Interval: 1.02-5.70; P = 0.045). A 17 gene methylation signature able to correctly assign prognosis (overall error rate = 0) in stage IIIC patients on the basis of distinct methylation-defined groups was also identified. CONCLUSIONS: A discrete whole-genome methylation signature has been identified as molecular marker of prognosis for stage IIIC CM patients. Its use in daily practice is foreseeable, and promises to refine the comprehensive clinical management of stage III CM patients.


Asunto(s)
Metilación de ADN , Genoma , Melanoma/metabolismo , Análisis de Supervivencia , Adulto , Anciano , Anciano de 80 o más Años , Secuencia de Bases , Cartilla de ADN , Femenino , Humanos , Masculino , Melanoma/patología , Persona de Mediana Edad , Reacción en Cadena en Tiempo Real de la Polimerasa
15.
J Cell Physiol ; 226(10): 2595-600, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21792917

RESUMEN

No treatment prolongs the survival of malignant mesothelioma (MM) patients. Since MM elicits anti-tumor host's immune responses, immunotherapy represents a promising strategy for its control. Immunomodulatory antibodies against components of the B7 family of immunomodulatory molecules that regulate T cell activation are being investigated in human malignancies including MM. The expression of B7-H3, a new component of the B7 family was investigated in primary cultures of human mesothelial cells (HMC) and in MM cell lines by flow cytometry and molecular analyses, and in MM tissues by immunohistochemistry. The role of DNA hypomethylating agents in modulating levels of B7-H3 expression in MM cells was also studied. Reverse transcriptase-polymerase chain reaction (RT-PCR) demonstrated that B7-H3 mRNA was consistently detectable in mesothelial and MM cells investigated; however, real-time quantitative RT-PCR analyses showed highly heterogeneous levels of B7-H3 mRNA among investigated MM cells. The analysis of B7-H3 protein expression indicated that comparable levels of B7-H3 were expressed on both cell types. Treatment with the DNA hypomethylating agent 5-aza-2'-deoxycytidine did not significantly affect the expression of B7-H3 mRNA in MM cells. In vivo, while B7-H3 was expressed in all 13 tumor biopsies of the epithelial variant, with high levels in 54% of cases, it was rarely detectable in spindle type MM in which 1/5 biopsies weakly expressed B7-H3. These findings suggest that B7-H3 is a promising target for new immunotherapeutic strategies in MM, with particular emphasis in the epithelial variant.


Asunto(s)
Antígenos CD/genética , Antígenos CD/inmunología , Vacunas contra el Cáncer/uso terapéutico , Mesotelioma/terapia , Neoplasias Pleurales/terapia , Receptores Inmunológicos/genética , Receptores Inmunológicos/inmunología , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/uso terapéutico , Antígenos CD/biosíntesis , Antígenos B7 , Línea Celular Tumoral , Células Epiteliales/citología , Células Epiteliales/inmunología , Células Epiteliales/patología , Humanos , Inmunofenotipificación , Mesotelioma/genética , Mesotelioma/inmunología , Neuroblastoma/inmunología , Neuroblastoma/patología , Derrame Pleural/inmunología , Derrame Pleural/patología , Neoplasias Pleurales/genética , Neoplasias Pleurales/inmunología , Receptores Inmunológicos/biosíntesis
16.
J Transl Med ; 9: 78, 2011 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-21615918

RESUMEN

BACKGROUND: The prognosis of cutaneous melanoma (CM) differs for patients with identical clinico-pathological stage, and no molecular markers discriminating the prognosis of stage III individuals have been established. Genome-wide alterations in DNA methylation are a common event in cancer. This study aimed to define the prognostic value of genomic DNA methylation levels in stage III CM patients. METHODS: Overall level of genomic DNA methylation was measured using bisulfite pyrosequencing at three CpG sites (CpG1, CpG2, CpG3) of the Long Interspersed Nucleotide Element-1 (LINE-1) sequences in short-term CM cultures from 42 stage IIIC patients. The impact of LINE-1 methylation on overall survival (OS) was assessed using Cox regression and Kaplan-Meier analysis. RESULTS: Hypomethylation (i.e., methylation below median) at CpG2 and CpG3 sites significantly associated with improved prognosis of CM, CpG3 showing the strongest association. Patients with hypomethylated CpG3 had increased OS (P = 0.01, log-rank = 6.39) by Kaplan-Meyer analysis. Median OS of patients with hypomethylated or hypermethylated CpG3 were 31.9 and 11.5 months, respectively. The 5 year OS for patients with hypomethylated CpG3 was 48% compared to 7% for patients with hypermethylated sequences. Among the variables examined by Cox regression analysis, LINE-1 methylation at CpG2 and CpG3 was the only predictor of OS (Hazard Ratio = 2.63, for hypermethylated CpG3; 95% Confidence Interval: 1.21-5.69; P = 0.01). CONCLUSION: LINE-1 methylation is identified as a molecular marker of prognosis for CM patients in stage IIIC. Evaluation of LINE-1 promises to represent a key tool for driving the most appropriate clinical management of stage III CM patients.


Asunto(s)
Metilación de ADN/genética , Elementos de Nucleótido Esparcido Largo/genética , Melanoma/genética , Adulto , Anciano , Anciano de 80 o más Años , Secuencia de Bases , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Masculino , Melanoma/diagnóstico , Melanoma/patología , Persona de Mediana Edad , Datos de Secuencia Molecular , Estadificación de Neoplasias , Pronóstico , Modelos de Riesgos Proporcionales , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Secuencia de ADN , Neoplasias Cutáneas/diagnóstico , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología
17.
Mol Oncol ; 5(2): 164-82, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21376678

RESUMEN

Cancer testis antigens (CTA) are a large family of tumor-associated antigens expressed in human tumors of different histological origin, but not in normal tissues except for testis and placenta. This tumor-restricted pattern of expression, together with their strong in vivo immunogenicity, identified CTA as ideal targets for tumor-specific immunotherapeutic approaches, and prompted the development of several clinical trials of CTA-based vaccine therapy. Driven by this practical clinical interest, a more detailed characterization of CTA biology has been recently undertaken. So far, at least 70 families of CTA, globally accounting for about 140 members, have been identified. Most of these CTA are expressed during spermatogenesis, but their function is still largely unknown. Epigenetic events, particularly DNA methylation, appear to be the primary mechanism regulating CTA expression in both normal and transformed cells, as well as in cancer stem cells. In view of the growing interest in CTA biology, the aim of this review is to provide the most recent information on their expression, regulation and function, together with a brief summary of the major clinical trials involving CTA as therapeutic agents. The pharmacologic modulation of CTA expression profiles on neoplastic cells by DNA hypomethylating drugs will also be discussed as a feasible approach to design new combination therapies potentially able to improve the clinical efficacy of currently adopted CTA-based immunotherapeutic regimens in cancer patients.


Asunto(s)
Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Regulación Neoplásica de la Expresión Génica , Neoplasias/inmunología , Neoplasias/terapia , Metilación de ADN , Humanos , Inmunoterapia/métodos , Masculino
18.
J Transl Med ; 8: 56, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20540720

RESUMEN

Cutaneous melanoma is a very aggressive neoplasia of melanocytic origin with constantly growing incidence and mortality rates world-wide. Epigenetic modifications (i.e., alterations of genomic DNA methylation patterns, of post-translational modifications of histones, and of microRNA profiles) have been recently identified as playing an important role in melanoma development and progression by affecting key cellular pathways such as cell cycle regulation, cell signalling, differentiation, DNA repair, apoptosis, invasion and immune recognition. In this scenario, pharmacologic inhibition of DNA methyltransferases and/or of histone deacetylases were demonstrated to efficiently restore the expression of aberrantly-silenced genes, thus re-establishing pathway functions. In light of the pleiotropic activities of epigenetic drugs, their use alone or in combination therapies is being strongly suggested, and a particular clinical benefit might be expected from their synergistic activities with chemo-, radio-, and immuno-therapeutic approaches in melanoma patients. On this path, an important improvement would possibly derive from the development of new generation epigenetic drugs characterized by much reduced systemic toxicities, higher bioavailability, and more specific epigenetic effects.


Asunto(s)
Epigénesis Genética , Melanoma/tratamiento farmacológico , Melanoma/genética , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/genética , Metilación de ADN/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Epigénesis Genética/efectos de los fármacos , Histonas/metabolismo , Humanos , Melanoma/diagnóstico , MicroARNs/metabolismo , Pronóstico , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Neoplasias Cutáneas/diagnóstico
19.
J Cell Physiol ; 223(2): 352-8, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20127705

RESUMEN

The intratumoral heterogeneity of cancer testis antigens (CTA) expression, which is driven by promoter methylation status, may hamper the effectiveness of CTA-directed vaccination of melanoma patients. Thus, we investigated whether the intratumoral heterogeneity of CTA expression is inherited at cellular level, or evolves throughout cellular replication, leading to a phenotypically unstable tumor cell population with reduced immunogenicity and/or able to escape immune control. Utilizing a previously characterized ex vivo clonal model of intratumoral heterogeneity of CTA expression in melanoma, Mel 313 MAGE-A3-low clone 5 (clone 5(M3-low)) and MAGE-A3-high clone 14 (clone 14(M3-high)) were sub-cloned and analyzed for CTA profile. Molecular assays demonstrated that levels of MAGE-A3 expression were highly conserved among generated sub-clones, as compared to parental clones. A similar behavior was identified for an extensive panel of other CTA investigated. Inherited levels of MAGE-A3 expression correlated with the extent of promoter methylation among clone 5(M3-low) and clone 14(M3-high) sub-clones analyzed. Treatment of clone 5(M3-low) with a DNA hypomethylating agent (DHA) resulted in an up-regulated expression of MAGE-A3, which was inherited at single cell level, being still detectable at day 60 in its sub-clones. Bisulfite sequencing demonstrated that also MAGE-A3 promoter methylation status was inherited among sub-clones generated from DHA-treated clone 5(M3-low) and strictly correlated with MAGE-A3 expression levels in investigated sub-clones. Similar results were obtained for additional CTA studied. Altogether our findings demonstrate that constitutive and DHA-modified CTA profiles of melanoma cells are clonally inherited throughout cellular replications, thus providing relevant insights to improve the effectiveness of CTA-based immunotherapy.


Asunto(s)
Antígenos de Neoplasias/genética , Células Clonales/metabolismo , Epigénesis Genética/genética , Regulación Neoplásica de la Expresión Génica/genética , Patrón de Herencia/genética , Melanoma/genética , Melanoma/metabolismo , Antimetabolitos Antineoplásicos/farmacología , Azacitidina/análogos & derivados , Azacitidina/farmacología , División Celular/genética , Células Clonales/efectos de los fármacos , Clonación Molecular/métodos , Metilación de ADN/efectos de los fármacos , Decitabina , Epigénesis Genética/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas de Neoplasias/genética , Regiones Promotoras Genéticas/genética , Células Tumorales Cultivadas
20.
J Cell Physiol ; 215(2): 287-91, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18205182

RESUMEN

Neoplastic populations with stem cell potential have been most recently identified in human cutaneous melanoma, and initially characterized for their phenotypic profile. Being melanoma stem cells (MSC) the most desirable target of therapeutic intervention, we asked whether they express the epigenetically-regulated cancer testis antigens (CTA) on which melanoma immunotherapy is increasingly focusing. Reverse transcription-PCR analyses identified the presence of the large majority of investigated CTA (i.e., MAGE, GAGE, NY-ESO, and SSX families) in different MSC populations. MSC expressed MAGE-A proteins as detected by western blot; noteworthy, the distribution of MAGE-A proteins was highly homogeneous within given MSC populations as shown by confocal immunofluorescence. Promoter methylation studies unveiled a homogeneously-demethylated MAGE-A3 promoter that paired MAGE-A3 expression in MSC. Altogether these findings demonstrate that MSC can be efficiently targeted by CTA-directed immunotherapeutic approaches, and suggest that epigenetic patterns most likely drive the expression of CTA in MSC as previously shown for melanoma cells.


Asunto(s)
Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Melanoma/metabolismo , Células Madre/metabolismo , Testículo/inmunología , Western Blotting , Línea Celular Tumoral , Metilación de ADN , Epigénesis Genética , Técnica del Anticuerpo Fluorescente , Humanos , Masculino , Melanoma/patología , Microscopía Confocal , Regiones Promotoras Genéticas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA