Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Cancers (Basel) ; 16(11)2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38893253

RESUMEN

This review discusses the topic of prevention of brain metastases from the most frequent solid tumor types, i.e., lung cancer, breast cancer and melanoma. Within each tumor type, the risk of brain metastasis is related to disease status and molecular subtype (i.e., EGFR-mutant non-small cell lung cancer, HER2-positive and triple-negative breast cancer, BRAF and NRAF-mutant melanoma). Prophylactic cranial irradiation is the standard of care in patients in small cell lung cancer responsive to chemotherapy but at the price of late neurocognitive decline. More recently, several molecular agents with the capability to target molecular alterations driving tumor growth have proven as effective in the prevention of secondary relapse into the brain in clinical trials. This is the case for EGFR-mutant or ALK-rearranged non-small cell lung cancer inhibitors, tucatinib and trastuzumab-deruxtecan for HER2-positive breast cancer and BRAF inhibitors for melanoma. The need for screening with an MRI in asymptomatic patients at risk of brain metastases is emphasized.

2.
Melanoma Res ; 34(2): 175-181, 2024 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-38265469

RESUMEN

Melanoma diagnosed within 1 year of pregnancy is defined as pregnancy-associated melanoma (PAM). No robust data on how pregnancy influences melanoma nor guidelines for PAM management exist. With IRB approval, female patients with a pathology-confirmed melanoma diagnosis within 1 year of pregnancy treated at our institution from 2000 to 2020 were identified. Controls from the cancer registry were matched 1 : 4 when available on decade of age, year of surgery (±5), and stage. We identified 83 PAM patients with median follow-up of 86 months. Mean age at diagnosis was 31 years. 80% AJCC V8 stage I, 2.4% stage II, 13% stage III, 4.8% stage IV. Mean Breslow thickness was 0.79 mm and 3.6% exhibited ulceration. The mean mitotic rate was 0.76/mm 2 . In terms of PAM management, 98.6% of ESD patients and 86.7% of LSD patients received standard-of-care therapy per NCCN guidelines for their disease stage. No clinically significant delays in treatment were noted. Time to treatment from diagnosis to systemic therapy for LSD patients was an average of 46 days (95% CI: 34-59 days). Comparing the 83 PAM patients to 309 controls matched on age, stage, and year of diagnosis, similar 5-year overall survival (97% vs. 97%, P  = 0.95) or recurrence-free survival (96% vs. 96%, P  = 0.86) was observed. The outcomes of PAM following SOC treatment at a highly specialized center for melanoma care were comparable to non-PAM when matched by clinical-pathologic features. Specialty center care is encouraged for women with PAM.


Asunto(s)
Melanoma , Neoplasias Cutáneas , Embarazo , Humanos , Femenino , Adulto , Melanoma/terapia , Neoplasias Cutáneas/terapia , Sistema de Registros
3.
EMBO Rep ; 24(10): e55043, 2023 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-37551717

RESUMEN

The cardiac endothelium influences ventricular chamber development by coordinating trabeculation and compaction. However, the endothelial-specific molecular mechanisms mediating this coordination are not fully understood. Here, we identify the Sox7 transcription factor as a critical cue instructing cardiac endothelium identity during ventricular chamber development. Endothelial-specific loss of Sox7 function in mice results in cardiac ventricular defects similar to non-compaction cardiomyopathy, with a change in the proportions of trabecular and compact cardiomyocytes in the mutant hearts. This phenotype is paralleled by abnormal coronary artery formation. Loss of Sox7 function disrupts the transcriptional regulation of the Notch pathway and connexins 37 and 40, which govern coronary arterial specification. Upon Sox7 endothelial-specific deletion, single-nuclei transcriptomics analysis identifies the depletion of a subset of Sox9/Gpc3-positive endocardial progenitor cells and an increase in erythro-myeloid cell lineages. Fate mapping analysis reveals that a subset of Sox7-null endothelial cells transdifferentiate into hematopoietic but not cardiomyocyte lineages. Our findings determine that Sox7 maintains cardiac endothelial cell identity, which is crucial to the cellular cross-talk that drives ventricular compaction and coronary artery development.


Asunto(s)
Vasos Coronarios , Células Endoteliales , Animales , Ratones , Vasos Coronarios/metabolismo , Células Endoteliales/metabolismo , Miocitos Cardíacos/metabolismo , Regulación de la Expresión Génica , Endotelio/metabolismo , Factores de Transcripción SOXF/genética , Factores de Transcripción SOXF/metabolismo
4.
Front Immunol ; 13: 1024039, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36544759

RESUMEN

Introduction: Immune cell infiltration into the tumor microenvironment is generally associated with favorable clinical outcomes in solid tumors. However, the dynamic interplay among distinct immune cell subsets within the tumor-immune microenvironment as it relates to clinical responses to immunotherapy remains unresolved. In this study, we applied multiplex immunofluorescence (MxIF) to spatially characterize tumor-immune interactions within the metastatic melanoma lymph node. Methods: Pretreatment, whole lymph node biopsies were evaluated from 25 patients with regionally metastatic melanoma who underwent subsequent anti-PD1 therapy. Cyclic MxIF was applied to quantitatively and spatially assess expression of 45 pathologist-validated antibodies on a single tissue section. Pixel-based single cell segmentation and a supervised classifier approach resolved 10 distinct tumor, stromal and immune cell phenotypes and functional expression of PD1. Results: Single cell analysis across 416 pathologist-annotated tumor core regions of interest yielded 5.5 million cells for spatial evaluation. Cellular composition of tumor and immune cell subsets did not differ in the tumor core with regards to recurrence outcomes (p>0.05) however spatial patterns significantly differed in regional and paracrine neighborhood evaluations. Specifically, a regional community cluster comprised of primarily tumor and dendritic cells was enriched in patients that did not experience recurrence (p=0.009). By an independent spatial approach, cell-centric neighborhood analyses identified an enrichment for dendritic cells in cytotoxic T cell (CTL) and tumor cell-centric neighborhoods in the no recurrence patient response group (p<0.0001). Further evaluation of these neighborhoods identified an enrichment for CTL-dendritic cell interactions in patients that did not experience recurrence (p<0.0001) whereas CTL-macrophage interactions were more prevalent in CTL-centric neighborhoods of patients who experienced recurrence (p<0.0001). Discussion: Overall, this study offers a more comprehensive evaluation of immune infiltrates and spatial-immune signatures in the metastatic tumor-immune microenvironment as it informs recurrence risk following immunotherapy.


Asunto(s)
Melanoma , Neoplasias Primarias Secundarias , Humanos , Melanoma/tratamiento farmacológico , Linfocitos T Citotóxicos , Inmunoterapia , Ganglios Linfáticos/patología , Microambiente Tumoral
5.
BMJ Open ; 12(5): e050112, 2022 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-35551087

RESUMEN

BACKGROUND: Patients with metastatic melanoma rely on PD-(L)1 immunotherapy, but only one-third of patients experience treatment response and all initial responders eventually develop resistance. Tumour-derived extracellular vesicles expressing Programmed death ligand 1 (evPD-L1) and soluble Programmed death ligand 1 (sPD-L1) in peripheral blood of patients with melanoma limit PD-(L)1 immunotherapy and correlate with poor survival. Therapeutic plasma exchange (TPE) removes immunosuppressive evPD-L1 and sPD-L1. We hypothesise that TPE may rescue and restore antimelanoma immunity. METHODS: In this two-arm study, 60 patients with metastatic melanoma progressing on checkpoint inhibition will be accrued. All patients will undergo radiotherapy on days 1-5 (at least one measurable lesion will not be irradiated) and ongoing checkpoint inhibition on day 8 and every 2-3 weeks per standard of care. Patients with baseline sPD-L1 level of ≥1.7 ng/mL and adequate clinical capacity will be enrolled in the TPE intervention arm and will undergo TPE on days 5-7, in addition to standard of care radiotherapy and immunotherapy. Other patients will remain in the standard of care arm.The primary endpoint of the study is to evaluate safety. Secondary endpoints include kinetics of sPD-L1 and evPD-L1 and clinical response by RECIST (Response Evaluation Criteria in Solid Tumors) criteria. Study registered at ClinicalTrials.gov (NCT04581382). ETHICS AND DISSEMINATION: This trial has been approved by the Mayo Clinic Institutional Review Board. It will assess the safety and feasibility of TPE in improving outcomes for PD-(L)1 inhibitor immunotherapy in melanoma. Data will be maintained on a secure database with deidentified patient information. Data will be shared on publication in a peer-reviewed journal without the aid of professional writers. If successful, this trial will lay the ground for phase II studies that will include cancer treated with PD-(L)1 inhibitors which may benefit from TPE such as renal, bladder and lung cancers. TRIAL REGISTRATION NUMBER: NCT04581382.


Asunto(s)
Neoplasias Pulmonares , Melanoma , Neoplasias Primarias Secundarias , Antígeno B7-H1 , Humanos , Inmunoterapia/métodos , Neoplasias Pulmonares/terapia , Melanoma/terapia , Intercambio Plasmático
7.
Melanoma Res ; 32(1): 67-70, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-34783721

RESUMEN

Currently, there is no known clinical evidence that rituximab increases the rate of subsequent primary malignancies; however, some studies have raised the question of increased melanoma risk following rituximab treatment for non-Hodgkin lymphoma. We report three interesting cases of suspected rituximab-induced melanoma. We hypothesize that this association is secondary to rituximab-driven shifts in the immunologic balance. Based on these cases, it is possible that the number of post-rituximab melanoma cases is underreported. Further mechanistic research into individual cases and population-level studies are required to better define association and risk; however, given the increasing prevalence of oncologic and nononcologic rituximab use, awareness across all fields is essential.


Asunto(s)
Melanoma/inducido químicamente , Rituximab/efectos adversos , Anciano , Humanos , Masculino , Melanoma/patología
8.
J Pathol Inform ; 10: 11, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31057980

RESUMEN

BACKGROUND: To assess reproducibility and accuracy of overall Nottingham grade and component scores using digital whole slide images (WSIs) compared to glass slides. METHODS: Two hundred and eight pathologists were randomized to independently interpret 1 of 4 breast biopsy sets using either glass slides or digital WSI. Each set included 5 or 6 invasive carcinomas (22 total invasive cases). Participants interpreted the same biopsy set approximately 9 months later following a second randomization to WSI or glass slides. Nottingham grade, including component scores, was assessed on each interpretation, providing 2045 independent interpretations of grade. Overall grade and component scores were compared between pathologists (interobserver agreement) and for interpretations by the same pathologist (intraobserver agreement). Grade assessments were compared when the format (WSI vs. glass slides) changed or was the same for the two interpretations. RESULTS: Nottingham grade intraobserver agreement was highest using glass slides for both interpretations (73%, 95% confidence interval [CI]: 68%, 78%) and slightly lower but not statistically different using digital WSI for both interpretations (68%, 95% CI: 61%, 75%; P= 0.22). The agreement was lowest when the format changed between interpretations (63%, 95% CI: 59%, 68%). Interobserver agreement was significantly higher (P < 0.001) using glass slides versus digital WSI (68%, 95% CI: 66%, 70% versus 60%, 95% CI: 57%, 62%, respectively). Nuclear pleomorphism scores had the lowest inter- and intra-observer agreement. Mitotic scores were higher on glass slides in inter- and intra-observer comparisons. CONCLUSIONS: Pathologists' intraobserver agreement (reproducibility) is similar for Nottingham grade using glass slides or WSI. However, slightly lower agreement between pathologists suggests that verification of grade using digital WSI may be more challenging.

9.
Proc Natl Acad Sci U S A ; 111(32): 11768-73, 2014 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-25074915

RESUMEN

The mammalian sex-determining factor SRY comprises a conserved high-mobility group (HMG) box DNA-binding domain and poorly conserved regions outside the HMG box. Mouse Sry is unusual in that it includes a C-terminal polyglutamine (polyQ) tract that is absent in nonrodent SRY proteins, and yet, paradoxically, is essential for male sex determination. To dissect the molecular functions of this domain, we generated a series of Sry mutants, and studied their biochemical properties in cell lines and transgenic mouse embryos. Sry protein lacking the polyQ domain was unstable, due to proteasomal degradation. Replacing this domain with irrelevant sequences stabilized the protein but failed to restore Sry's ability to up-regulate its key target gene SRY-box 9 (Sox9) and its sex-determining function in vivo. These functions were restored only when a VP16 transactivation domain was substituted. We conclude that the polyQ domain has important roles in protein stabilization and transcriptional activation, both of which are essential for male sex determination in mice. Our data disprove the hypothesis that the conserved HMG box domain is the only functional domain of Sry, and highlight an evolutionary paradox whereby mouse Sry has evolved a novel bifunctional module to activate Sox9 directly, whereas SRY proteins in other taxa, including humans, seem to lack this ability, presumably making them dependent on partner proteins(s) to provide this function.


Asunto(s)
Genes sry , Procesos de Determinación del Sexo , Proteína de la Región Y Determinante del Sexo/genética , Proteína de la Región Y Determinante del Sexo/metabolismo , Animales , Evolución Molecular , Femenino , Genes Reporteros , Masculino , Ratones , Ratones Transgénicos , Mutagénesis , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Péptidos/química , Embarazo , Complejo de la Endopetidasa Proteasomal/metabolismo , Desnaturalización Proteica , Estabilidad Proteica , Estructura Terciaria de Proteína , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Eliminación de Secuencia , Proteína de la Región Y Determinante del Sexo/química , Activación Transcripcional
10.
Biol Reprod ; 90(5): 92, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24671879

RESUMEN

Spermatogenesis is a complex developmental process whereby diploid spermatogenic stem cells become haploid and undergo a series of morphological changes to produce physically mature spermatozoa. Crucial to this process are a number of RNA-binding proteins, responsible for the posttranscriptional control of essential mRNAs and particularly pertinent to the two periods of inactive transcription that occur in spermatogenesis. One such group of RNA-binding proteins is the Musashi family, specifically Musashi-1 (MSI1) and Musashi-2 (MSI2), which act as key translational regulators in various stem cell populations and have been linked with the induction of tumorigenesis. In the present study, we examined the differential expression of mammalian MSI1 and MSI2 during germ cell development in the mouse testis. MSI1 was found to be predominately localized in mitotic gonocytes and spermatogonia, whereas MSI2 was detected in meiotic spermatocytes and differentiating spermatids. Extensive examination of the function of Musashi in spermatogenesis was achieved through the use of two transgenic mouse models with germ cell-specific overexpression of full-length isoforms of Msi1 or Msi2. These models demonstrated that aberrant expression of either Msi1 or Msi2 has deleterious effects on normal spermatogenesis, with Msi2 overexpression resulting in male sterility. Studies undertaken on human testicular seminoma tumors provide further insights into the relevance of MSI1 and MSI2 overexpression as diagnostic markers to human stem cell cancers. Overall this study provides further evidence for the unique functions that RNA-binding protein isoforms occupy within spermatogenesis, and introduces the potential manipulation of the Musashi family proteins to elucidate the mechanisms of posttranscriptional gene expression during germ cell development.


Asunto(s)
Proteínas de Unión al ARN/fisiología , Espermatocitos/fisiología , Espermatogénesis/fisiología , Espermatogonias/fisiología , Testículo/fisiología , Animales , Western Blotting , Regulación del Desarrollo de la Expresión Génica , Humanos , Inmunohistoquímica , Masculino , Meiosis/genética , Meiosis/fisiología , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Isoformas de Proteínas , ARN/química , ARN/genética , Proteínas de Unión al ARN/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Espermatocitos/ultraestructura , Espermatogonias/ultraestructura , Estadísticas no Paramétricas , Testículo/citología , Testículo/metabolismo
11.
Dev Biol ; 364(2): 89-98, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22230615

RESUMEN

During lymphangiogenesis in the mammalian embryo, a subset of vascular endothelial cells in the cardinal veins is reprogrammed to adopt a lymphatic endothelial fate. The prevailing model of lymphangiogenesis contends that these lymphatic precursor cells migrate away from the cardinal veins and reassemble peripherally as lymph sacs from which a lymphatic vasculature is generated. However, this model fails to account for a number of observations that, as a result, have remained anecdotal. Here, we use optical projection tomography, confocal microscopy and in vivo live imaging to uncover three key stages of lymphatic vascular morphogenesis in the mouse embryo at high resolution. First, we define territories or "pre-lymphatic clusters" of Prox1-positive lymphatic endothelial progenitor cells along the antero-posterior axis of the cardinal veins. Second, these pre-lymphatic clusters undergo progressive extrusion ("ballooning") to generate primitive lymph sacs. Third, lymphatic vessels emerge by a combination of mechanisms including sprouting from the lymph sacs and direct delamination of streams of cells from the cardinal veins. Our data support a new model for lymphatic vascular patterning and morphogenesis, as a basis for identifying the molecular cues governing these processes.


Asunto(s)
Linfangiogénesis , Vasos Linfáticos/embriología , Venas/embriología , Animales , Proteínas de Homeodominio/análisis , Ratones , Proteínas Supresoras de Tumor/análisis
12.
Dev Cell ; 19(3): 440-9, 2010 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-20833365

RESUMEN

Sex determination of mammalian germ cells occurs during fetal development and depends on signals from gonadal somatic cells. Previous studies have established that retinoic acid (RA) triggers ovarian germ cells to enter meiosis and thereby commit to oogenesis, whereas in the developing testis, the enzyme CYP26B1 degrades RA and germ cells are not induced to enter meiosis. Using in vitro and in vivo models, we demonstrate that fibroblast growth factor 9 (FGF9) produced in the fetal testis acts directly on germ cells to inhibit meiosis; in addition, FGF9 maintains expression of pluripotency-related genes and upregulates markers associated with male germ cell fate. We conclude that two independent and mutually antagonistic pathways involving RA and FGF9 act in concert to determine mammalian germ cell sexual fate commitment and support a model in which the mitosis/meiosis switch is robustly controlled by both positive and negative regulatory factors.


Asunto(s)
Factor 9 de Crecimiento de Fibroblastos/fisiología , Células Germinativas/fisiología , Meiosis/fisiología , Células Madre Pluripotentes/metabolismo , Animales , Antineoplásicos/farmacología , Western Blotting , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Sistema Enzimático del Citocromo P-450/metabolismo , Femenino , Feto/citología , Feto/efectos de los fármacos , Feto/metabolismo , Técnica del Anticuerpo Fluorescente , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oogénesis/efectos de los fármacos , Oogénesis/fisiología , Ovario/embriología , Ovario/metabolismo , ARN Mensajero/genética , Ácido Retinoico 4-Hidroxilasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Testículo/embriología , Testículo/metabolismo , Tretinoina/farmacología
13.
Development ; 136(14): 2385-91, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19515696

RESUMEN

Developmental defects caused by targeted gene inactivation in mice are commonly subject to strain-specific modifiers that modulate the severity of the phenotype. Although several genetic modifier loci have been mapped in mice, the gene(s) residing at these loci are mostly unidentified, and the molecular mechanisms of modifier action remain poorly understood. Mutations in Sox18 cause a variable phenotype in the human congenital syndrome hypotrichosis-lymphedema-telangiectasia, and the phenotype of Sox18-null mice varies from essentially normal to completely devoid of lymphatic vasculature and lethal, depending on the strain of the mice, suggesting a crucial role for strain-specific modifiers in this system. Here we show that two closely related Group F Sox factors, SOX7 and SOX17, are able to functionally substitute for SOX18 in vitro and in vivo. SOX7 and SOX17 are not normally expressed during lymphatic development, excluding a conventional redundancy mechanism. Instead, these genes are activated specifically in the absence of SOX18 function, and only in certain strains. Our studies identify Sox7 and Sox17 as modifiers of the Sox18 mutant phenotype, and reveal their mechanism of action as a novel mode of strain-specific compensatory upregulation.


Asunto(s)
Proteínas HMGB/fisiología , Linfangiogénesis/fisiología , Factores de Transcripción SOXF/fisiología , Animales , Secuencia de Bases , Cartilla de ADN/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas HMGB/genética , Proteínas de Homeodominio/genética , Humanos , Hipotricosis/genética , Linfangiogénesis/genética , Linfedema/genética , Ratones , Ratones Endogámicos CBA , Ratones Noqueados , Ratones Transgénicos , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Transcripción SOXF/deficiencia , Factores de Transcripción SOXF/genética , Especificidad de la Especie , Síndrome , Telangiectasia/genética , Proteínas Supresoras de Tumor/genética , Regulación hacia Arriba
14.
Nature ; 456(7222): 643-7, 2008 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-18931657

RESUMEN

The lymphatic system plays a key role in tissue fluid regulation and tumour metastasis, and lymphatic defects underlie many pathological states including lymphoedema, lymphangiectasia, lymphangioma and lymphatic dysplasia. However, the origins of the lymphatic system in the embryo, and the mechanisms that direct growth of the network of lymphatic vessels, remain unclear. Lymphatic vessels are thought to arise from endothelial precursor cells budding from the cardinal vein under the influence of the lymphatic hallmark gene Prox1 (prospero homeobox 1; ref. 4). Defects in the transcription factor gene SOX18 (SRY (sex determining region Y) box 18) cause lymphatic dysfunction in the human syndrome hypotrichosis-lymphoedema-telangiectasia, suggesting that Sox18 may also play a role in lymphatic development or function. Here we use molecular, cellular and genetic assays in mice to show that Sox18 acts as a molecular switch to induce differentiation of lymphatic endothelial cells. Sox18 is expressed in a subset of cardinal vein cells that later co-express Prox1 and migrate to form lymphatic vessels. Sox18 directly activates Prox1 transcription by binding to its proximal promoter. Overexpression of Sox18 in blood vascular endothelial cells induces them to express Prox1 and other lymphatic endothelial markers, while Sox18-null embryos show a complete blockade of lymphatic endothelial cell differentiation from the cardinal vein. Our findings demonstrate a critical role for Sox18 in developmental lymphangiogenesis, and suggest new avenues to investigate for therapeutic management of human lymphangiopathies.


Asunto(s)
Diferenciación Celular , Vasos Linfáticos/citología , Vasos Linfáticos/embriología , Factores de Transcripción SOXF/metabolismo , Animales , Biomarcadores/análisis , Movimiento Celular , Células Cultivadas , Edema/genética , Células Endoteliales/citología , Células Endoteliales/metabolismo , Efrina-B2/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Hipotricosis/genética , Linfangiogénesis , Vasos Linfáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Regiones Promotoras Genéticas/genética , Factores de Transcripción SOXF/deficiencia , Factores de Transcripción SOXF/genética , Telangiectasia/genética , Proteínas Supresoras de Tumor/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética , Venas/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA