Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.781
Filtrar
1.
Pol J Pathol ; 75(2): 97-104, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39166518

RESUMEN

Leukaemia-related protein 16 (LRP16) has been found to be highly expressed in various tumours and to be related to poor prognosis. However, the role of LRP16 in endometrial carcinoma remains to be explored. We aimed to investigate the prognosis and role of LRP16 in endometrial carcinoma. Overall, 160 endometrial carcinoma (EC) tissues and 60 benign samples were collected. The expression of LRP16 protein in EC tissues was significantly increased compared with that in normal endometrial tissues, and high LRP16 expression was related to poor patient prognosis. Reduced LRP16 expression markedly inhibited cancer cell growth. The proliferation rates in the prophylactic bilateral salpingectomy (PBS) group and the shNon group were 0.727 ±0.015 and 0.743 ±0.009, respectively, while the proliferation rate in the shLRP16 group was only 0.373 ±0.012. The migration experiment showed that the number of cells passing through the basement membrane in the shLRP16 group was 34.2 ±5.1, which was significantly different to the shNon (161.6 ±7.8) and PBS groups (138.0 ±7.2). The results of the invasion experiment showed that the number of cells was 39.2 ±6.2 in the shLRP16 group, 146.7 ±8.2 in the shNon group, and 141.2 ±8.1 in the PBS group ( p < 0.05). Leukaemia-related protein 16 is highly expressed in oestrogen-dependent EC and may promote cancer cell growth.


Asunto(s)
Biomarcadores de Tumor , Proliferación Celular , Neoplasias Endometriales , Humanos , Femenino , Neoplasias Endometriales/patología , Neoplasias Endometriales/metabolismo , Persona de Mediana Edad , Biomarcadores de Tumor/metabolismo , Biomarcadores de Tumor/análisis , Anciano , Línea Celular Tumoral , Movimiento Celular , Adulto , Neoplasias Hormono-Dependientes/patología , Neoplasias Hormono-Dependientes/metabolismo , Estrógenos/metabolismo , Hidrolasas de Éster Carboxílico
2.
J Steroid Biochem Mol Biol ; 243: 106559, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38823459

RESUMEN

Steroid hormone receptors are key mediators in the execution of hormone action through a combination of genomic and non-genomic action. Since their isolation and characterisation in the early 20th Century much of our understanding of the biological actions of steroid hormones are underpinned by their activated receptor activity. Over the past two decades there has been an acceleration of more omics-based research which has resulted in a major uptick in our comprehension of genomic steroid action. However, it is well understood that steroid hormones can induce very rapid signalling events in tandem with their genomic actions wherein they exert their influence through alterations in gene expression. Thus the totality of genomic and non-genomic steroid action occurs in a simultaneous and reciprocal manner and a greater appreciation of whole cell action is required to fully evaluate steroid hormone activity in vivo. In this mini-review we outline the most recent developments in non-genomic steroid action and cytoplasmic steroid hormone receptor biology in endocrine-related cancers with a focus on the 3-keto steroid receptors, in particular the androgen receptor.


Asunto(s)
Receptores de Esteroides , Transducción de Señal , Humanos , Receptores de Esteroides/metabolismo , Receptores de Esteroides/genética , Animales , Receptores Androgénicos/metabolismo , Receptores Androgénicos/genética , Neoplasias Hormono-Dependientes/metabolismo , Neoplasias Hormono-Dependientes/patología , Neoplasias Hormono-Dependientes/genética , Citoplasma/metabolismo
3.
Cancer Sci ; 114(7): 2729-2738, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37026526

RESUMEN

FK506 binding protein 52 (FKBP52) (gene name FKBP4) is a 52 kDa protein that belongs to the FKBP family; it binds to the immunosuppressant FK506 and has proline isomerase activity. In addition to its FK domain-containing peptidylprolyl isomerase activity, FKBP52 also acts as a cochaperone through the tetratricopeptide repeat domain that mediates binding to heat shock protein 90. Previous studies have reported that FKBP52 is associated with hormone-dependent, stress-related, and neurodegenerative diseases, revealing its diverse functions. In particular, the effects of FKBP52 on cancer have attracted considerable attention. FKBP52 promotes the growth of hormone-dependent cancers by activating steroid hormone receptors. Recent studies have shown that the expression of FKBP52 is increased not only in steroid hormone-dependent cancer cells but also in colorectal, lung, and liver cancers, revealing its diverse functions that contribute to cancer growth. This review summarizes reports related to hormone-dependent cancer and cell proliferation in terms of the structure of FKBP52 and its function on interacting molecules.


Asunto(s)
Neoplasias Hormono-Dependientes , Proteínas de Unión a Tacrolimus , Humanos , Proliferación Celular/genética , Proteínas HSP90 de Choque Térmico/metabolismo , Neoplasias Hormono-Dependientes/metabolismo , Neoplasias Hormono-Dependientes/patología , Unión Proteica , Proteínas de Unión a Tacrolimus/genética , Proteínas de Unión a Tacrolimus/química , Proteínas de Unión a Tacrolimus/metabolismo
4.
Chin Med J (Engl) ; 136(21): 2609-2620, 2023 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-37027287

RESUMEN

BACKGROUND: Observational research has reported that systemic lupus erythematosus (SLE) is related to common female hormone-dependent cancers, but the underlying causal effect remains undefined. This study aimed to explore the causal association of these conditions by Mendelian randomization (MR) analysis. METHODS: We selected instrumental variables for SLE from genome-wide association studies (GWASs) conducted in European and East Asian populations. The genetic variants for female malignant neoplasms were obtained from corresponding ancestry GWASs. We utilized inverse variance weighted (IVW) as the primary analysis, followed by sensitivity analysis. Furthermore, we conducted multivariable MR (MVMR) to estimate direct effects by adjusting for the body mass index and estradiol. Finally, we implemented reverse direction MR analysis and gave a negative example to test the reliability of MR results. RESULTS: We found SLE was significantly negatively associated with overall endometrial cancer risk (odds ratio [OR] = 0.961, 95% confidence interval [CI] = 0.935-0.987, P  = 3.57E-03) and moderately inversely related to endometrioid endometrial cancer (ENEC) (OR = 0.965, 95% CI = 0.936-0.995, P  = 0.024) risk in the European population by IVW. We replicated these results using other MR models and detected a direct effect by MVMR (overall endometrial cancer, OR = 0.962, 95% CI = 0.941-0.983, P  = 5.11E-04; ENEC, OR = 0.964, 95% CI = 0.940-0.989, P  = 0.005). Moreover, we revealed that SLE was correlated with decreased breast cancer risk (OR = 0.951, 95% CI = 0.918-0.986, P  = 0.006) in the East Asian population by IVW, and the effect was still significant in MVMR (OR = 0.934, 95% CI = 0.859-0.976, P  = 0.002). The statistical powers of positive MR results were all >0.9. CONCLUSION: This finding suggests a possible causal effect of SLE on the risk of overall endometrial cancer and breast cancer in European and East Asian populations, respectively, by MR analysis, which compensates for inherent limitations of observational research.


Asunto(s)
Neoplasias de la Mama , Carcinoma Endometrioide , Neoplasias Endometriales , Lupus Eritematoso Sistémico , Neoplasias Hormono-Dependientes , Femenino , Humanos , Estudio de Asociación del Genoma Completo , Análisis de la Aleatorización Mendeliana , Reproducibilidad de los Resultados , Lupus Eritematoso Sistémico/genética , Polimorfismo de Nucleótido Simple
5.
Mastology (Online) ; 332023. tab, graf, ilus
Artículo en Inglés | LILACS | ID: biblio-1443729

RESUMEN

Hormone-dependent breast cancer has growth factors that respond positively to the hormones estrogen and progesterone. Thus, adjuvant endocrine therapy causes decreased or undetectable serum levels of these hormones. However, this treatment can have side effects that compromise the sexual health of patients, such as dyspareunia, vaginal dryness and decreased libido. In this scenario, the objective of this work was to document the main outcomes in sexuality in women after treatment for hormonepositive breast cancer. Thus, this is an integrative literature review, in which the following databases were used: U.S. National Library of Medicine (PubMed), Virtual Health Library (BVS), SCOPUS and Scientific Electronic Library Online (SCIELO), using the descriptors: "sexuality", "antineoplastic agents, hormonal" and "breast neoplasms", joined by the Boolean operator "AND". Full articles published in the last 5 years (2017-2022) were included; written in Portuguese or English. Articles dealing with non-hormone-dependent or metastatic breast cancer, or with patients younger than 18 years, or articles that did not answer the research question were excluded. In total, 26 articles were identified, of which 7 comprised the final sample of this review. A total of 3,850 women participated in the included studies. The main sexual dysfunctions found were: dyspareunia, hot flashes, decreased libido, vaginal dryness, breast tenderness, self-image concerns and hair loss. The symptom vaginal dryness was the most prevalent, mentioned in 71.4% of the articles included. In view of the adverse effects listed in this review, there is a need to carry out more studies on this topic, since the diagnosis of this comorbidity brings clinical, psychological, emotional, sociocultural and economic outcomes for the patient. Thus, a multidisciplinary team must assertively address these complaints to improve the overall quality of life of these women (AU)


Asunto(s)
Humanos , Femenino , Disfunciones Sexuales Fisiológicas/inducido químicamente , Neoplasias de la Mama/tratamiento farmacológico , Antineoplásicos Hormonales/uso terapéutico , Sexualidad/efectos de los fármacos , Neoplasias Hormono-Dependientes/tratamiento farmacológico
6.
Med Oncol ; 40(1): 41, 2022 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-36471176

RESUMEN

Coumarin is a bicyclic oxygen bearing heterocyclic scaffold formed by fusion of benzene with the pyrone ring. Because of its unique physicochemical characteristics and the ease with which it may be transformed into a wide range of functionalized coumarins during synthesis, coumarin provides a privileged scaffold for medicinal chemists. As a result, many coumarin derivatives have been developed, synthesized, and evaluated to target a variety of therapeutic domains, thereby making it an attractive template for designing novel anti-breast cancer compounds. The main culprit in estrogen overproduction in the estrogen-dependent breast cancer (EDBC), is the enzyme aromatase (AR), and it is thought to be a significant target for the effective treatment of EDBC. Considering coumarins versatility, this review presents a detailed overview of diverse study of aromatase as a target for coumarins. An overview of structure-activity relationship analysis of coumarin core is also included so as to summarize the desired pharmacophoric features essential for design and development of aromatase inhibitors (AIs) using coumarin core. Identification of key synthesis techniques that could aid researchers in designing and developing novel analogues with significant anti-breast cancer properties along with their mechanism of action have also been covered in the current review.


Asunto(s)
Antineoplásicos , Inhibidores de la Aromatasa , Neoplasias de la Mama , Cumarinas , Estrógenos , Neoplasias Hormono-Dependientes , Femenino , Humanos , Antineoplásicos/uso terapéutico , Aromatasa/metabolismo , Inhibidores de la Aromatasa/farmacología , Inhibidores de la Aromatasa/química , Inhibidores de la Aromatasa/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Química Farmacéutica , Cumarinas/farmacología , Cumarinas/química , Cumarinas/uso terapéutico , Estrógenos/metabolismo , Neoplasias Hormono-Dependientes/tratamiento farmacológico
7.
Mol Genet Genomics ; 297(4): 947-963, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35532795

RESUMEN

Malignancies dependent on hormone homeostasis include breast, ovary, cervical, prostate, testis and uterine tumors. Hormones are involved in signal transduction which orchestrate processes, such as apoptosis, proliferation, cell cycle or cytoskeleton organization. Currently, there is a need for novel biomarkers which would help to diagnose cancers efficiently. In this study, the genes implicated in signaling that is important in hormone-sensitive carcinogenesis were investigated regarding their prognostic significance. Data of seven cancer cohorts were collected from FireBrowse. 54 gene sets implicated in specific pathways were browsed through MSig database. Profiling was assessed via Monocle3, while gene ontology through PANTHER. For confirmation, correlation analysis was performed using WGCNA. Protein-protein networks were visualized via Cytoscape and impact of genes on survival, as well as cell cycle or cytoskeleton-related prognostic signatures, was tested. Several differences in expression profile were identified, some of them allowed to distinguish histology. Functional annotation revealed that various regulation of cell cycle, adhesion, migration, apoptosis and angiogenesis underlie these differences. Clinical traits, such as histological type or cancer staging, were found during evaluation of module-trait relationships. Of modules, the TopHubs (COL6A3, TNR, GTF2A1, NKX3-1) interacted directly with, e.g., PDGFB, ITGA10, SP1 or AKT3. Among TopHubs and interacting proteins, many showed an impact on hazard ratio and affected the cell cycle or cytoskeleton-related prognostic signatures, e.g., COL1A1 or PDGFB. In conclusion, this study laid the foundation for further hormone-sensitive carcinogenesis research through identification of genes which prove that crosstalk between cell cycle and cytoskeleton exists, opening avenues for future therapeutic strategies.


Asunto(s)
Neoplasias Hormono-Dependientes , Neoplasias , Biomarcadores , Carcinogénesis/genética , Ciclo Celular/genética , Biología Computacional , Citoesqueleto/genética , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Hormonas , Humanos , Neoplasias/diagnóstico , Neoplasias/genética , Neoplasias Hormono-Dependientes/genética , Proteínas Proto-Oncogénicas c-sis/genética , Transducción de Señal/genética
8.
Int J Mol Sci ; 23(3)2022 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-35163166

RESUMEN

It is known that estrogen stimulates growth and inhibits apoptosis through estrogen receptor(ER)-mediated mechanisms in many cancer cell types. Interestingly, there is strong evidence that estrogens can also induce apoptosis, activating different ER isoforms in cancer cells. It has been observed that E2/ERα complex activates multiple pathways involved in both cell cycle progression and apoptotic cascade prevention, while E2/ERß complex in many cases directs the cells to apoptosis. However, the exact mechanism of estrogen-induced tumor regression is not completely known. Nevertheless, ERs expression levels of specific splice variants and their cellular localization differentially affect outcome of estrogen-dependent tumors. The goal of this review is to provide a general overview of current knowledge on ERs-mediated apoptosis that occurs in main hormone dependent-cancers. Understanding the molecular mechanisms underlying the induction of ER-mediated cell death will be useful for the development of specific ligands capable of triggering apoptosis to counteract estrogen-dependent tumor growth.


Asunto(s)
Apoptosis , Neoplasias Hormono-Dependientes/patología , Receptores de Estrógenos/metabolismo , Animales , Humanos , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/metabolismo , Receptores de Estrógenos/genética , Transducción de Señal
9.
Genes (Basel) ; 12(8)2021 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-34440460

RESUMEN

Endocrine tumors are neoplasms originating from specialized hormone-secreting cells. They can develop as sporadic tumors, caused by somatic mutations, or in the context of familial Mendelian inherited diseases. Congenital forms, manifesting as syndromic or non-syndromic diseases, are caused by germinal heterozygote autosomal dominant mutations in oncogenes or tumor suppressor genes. The genetic defect leads to a loss of cell growth control in target endocrine tissues and to tumor development. In addition to the classical cancer manifestations, some affected patients can manifest alterations of bone and mineral metabolism, presenting both as pathognomonic and/or non-specific skeletal clinical features, which can be either secondary complications of endocrine functioning primary tumors and/or a direct consequence of the gene mutation. Here, we specifically review the current knowledge on possible direct roles of the genes that cause inherited endocrine tumors in the regulation of bone modeling and remodeling by exploring functional in vitro and in vivo studies highlighting how some of these genes participate in the regulation of molecular pathways involved in bone and mineral metabolism homeostasis, and by describing the potential direct effects of gene mutations on the development of skeletal and mineral metabolism clinical features in patients.


Asunto(s)
Neoplasias de las Glándulas Endocrinas/genética , Genes Supresores de Tumor , Neoplasias Hormono-Dependientes/genética , Osteoporosis/genética , Huesos/metabolismo , Huesos/patología , Neoplasias de las Glándulas Endocrinas/complicaciones , Neoplasias de las Glándulas Endocrinas/patología , Heterocigoto , Humanos , Mutación , Neoplasias Hormono-Dependientes/complicaciones , Neoplasias Hormono-Dependientes/patología , Oncogenes/genética , Osteoporosis/complicaciones , Osteoporosis/patología
10.
Curr Oncol Rep ; 23(11): 127, 2021 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-34453233

RESUMEN

PURPOSE OF REVIEW: While females make up almost 60% of all brain and spinal cord tumors in adults, guidelines that address women's issues in neuro-oncology are lacking. This review sheds light on two common women's issues in neuro-oncology. RECENT FINDINGS: Neuro-oncology providers are often faced with patient questions about fertility and pregnancy maintenance or prevention and typically respond with generic cancer chemotherapy recommendations, based on the paucity of evidence on the use of common neuro-oncology chemotherapies and pregnancy. While these remain important gap issues, there are several other poorly researched issues in the Neuro-Oncology of Women (N.O.W.) including recommendations around endogenous and iatrogenic hormone exposure and female sexuality in cancer. As a significant percentage of cancers are hormone-dependent, it is important to understand how changes in hormone levels impact tumor biology over the course of a woman's lifespan. Furthermore, greater attention should be given to the impact of tumors and tumor treatments on female sexuality. This article is intended to serve as an introduction to these two specific subjects within the vast expanse of N.O.W. subject matter.


Asunto(s)
Neoplasias Encefálicas/etiología , Neoplasias Hormono-Dependientes/etiología , Calidad de Vida , Neoplasias Encefálicas/psicología , Neoplasias Encefálicas/secundario , Medicina Basada en la Evidencia , Femenino , Glioma/etiología , Humanos , Meningioma/etiología , Neoplasias Hormono-Dependientes/psicología , Neoplasias Hipofisarias/etiología , Neoplasias Hipofisarias/psicología , Guías de Práctica Clínica como Asunto , Autoimagen , Disfunciones Sexuales Fisiológicas/etiología , Disfunciones Sexuales Fisiológicas/terapia , Sexualidad
11.
Int J Mol Sci ; 22(11)2021 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-34199930

RESUMEN

Endometriosis is a common gynecological disorder that has been associated with endometrial, breast and epithelial ovarian cancers in epidemiological studies. Since complex diseases are a result of multiple environmental and genetic factors, we hypothesized that the biological mechanism underlying their comorbidity might be explained, at least in part, by shared genetics. To assess their potential genetic relationship, we performed a two-sample mendelian randomization (2SMR) analysis on results from public genome-wide association studies (GWAS). This analysis confirmed previously reported genetic pleiotropy between endometriosis and endometrial cancer. We present robust evidence supporting a causal genetic association between endometriosis and ovarian cancer, particularly with the clear cell and endometrioid subtypes. Our study also identified genetic variants that could explain those associations, opening the door to further functional experiments. Overall, this work demonstrates the value of genomic analyses to support epidemiological data, and to identify targets of relevance in multiple disorders.


Asunto(s)
Neoplasias Endometriales/epidemiología , Endometriosis/epidemiología , Endometrio/patología , Predisposición Genética a la Enfermedad , Neoplasias Hormono-Dependientes/epidemiología , Neoplasias Ováricas/epidemiología , Polimorfismo de Nucleótido Simple , Neoplasias Endometriales/genética , Neoplasias Endometriales/patología , Endometriosis/genética , Endometriosis/patología , Femenino , Estudio de Asociación del Genoma Completo , Humanos , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/patología , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Factores de Riesgo , España/epidemiología
12.
Cancer Lett ; 520: 48-56, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34229060

RESUMEN

The deregulation of PI3K/Akt signaling is among the most causes in inducing the acquisition of a metastatic phenotype in breast cancer cells, leading to Epithelial-Mesenchymal Transition (EMT). Inhibition of the PI3K/Akt pathway is known to be beneficial in the clinical setting. However, the activation of secondary pathways and toxicity profiles of available inhibitors, hindering optimal therapeutic results. Preliminary studies showed that myo-Inositol inhibits the PI3K/Akt pathway by exerting a pleiotropic anti-tumor action. Herein, we demonstrate that myo-Inositol triggers a prompt and profound remodeling of delineated expression pattern in triple-negative breast cancer cells (MDA-MB-231). Consequently, it inhibits metastasis and tumor progression through miR-125a-5p transcription and the subsequent inhibition of IP6K1. In contrast, hormone-responsive breast cancer cells (MCF-7) are insensitive to myo-Inositol. This is due to the persistence of MDM2 synthesis promoted by estrogen-dependent pathways. Conversely, the counteraction of estrogen effects recovered the sensitivity to myo-Inositol in the hormone-responsive model. Overall, these results identify a novel axis primed by miR-125a-5p to downregulate IP6K1 gene that inhibits metastasis. Thus, administration of myo-Inositol can activate this axis as a molecular target therapy in breast cancer.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , MicroARNs/genética , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Fosfotransferasas (Aceptor del Grupo Fosfato)/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inositol/farmacología , Células MCF-7 , Metástasis de la Neoplasia , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/patología , Transducción de Señal/efectos de los fármacos
13.
Chest ; 160(5): 1915-1924, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34023321

RESUMEN

BACKGROUND: Malignant pleural effusions (MPEs) often cause symptoms, and guidelines recommend early definitive intervention. However, observational data suggest that systemic anticancer treatment (SACT) may control MPE caused by certain pharmacologically sensitive tumors. RESEARCH QUESTION: Is SACT associated with higher rates of MPE resolution in people with pharmacologically sensitive tumors? STUDY DESIGN AND METHODS: This was a retrospective analysis of prospectively collected data from an observational cohort study of people diagnosed with MPE from lung, breast, ovarian, and hematologic malignancy between May 11, 2008, and August 6, 2017. MPE resolution (defined as radiologic resolution with removal of drain or catheter and cessation of interventions) was compared in pharmacologically sensitive (high-grade lymphoma, small cell or target-mutation-positive lung cancer, and hormone-receptor-positive breast or ovarian cancer) and nonsensitive (remainder of cohort) tumors, with and without SACT. Secondary outcomes included time to resolution, 3-month resolution rates, and total pleural interventions. RESULTS: Of 280 patients, 127 had sensitive and 153 had nonsensitive tumors. One hundred seventy-one received SACT, and 109 did not. More patients with sensitive tumors achieved MPE resolution than those with nonsensitive tumors (53/127 [41.7%] vs 42/153 [27.5%]; P = .01), and this occurred predominantly after receipt of SACT. However, hematologic malignancies were overrepresented in the sensitive group, with high rates of SACT use and MPE resolution. After adjustment for this and other confounders, no relationship was found among pharmacologic sensitivity, SACT, and MPE resolution (adjusted OR, 1.4; 95% CI, 0.5-4.1). The strongest predictor of MPE resolution was administration of chemical pleurodesis (adjusted OR, 6.2; 95% CI, 3.3-11.7). In sensitive tumors, MPE resolution occurred without chemical pleurodesis in 14 of 52 patients (26.9%; 95% CI, 15.6%-41.1%) after SACT and in 5 of 22 patients (22.7%; 95% CI, 8.2%-47.2%) without SACT. INTERPRETATION: In this observational study, SACT was not associated independently on MPE resolution in pharmacologically sensitive tumors. Randomized trials are required, but with current data, patients with symptomatic MPE should receive early definitive pleural intervention regardless of underlying tumor or intended treatment.


Asunto(s)
Terapia Molecular Dirigida/métodos , Neoplasias Hormono-Dependientes , Neoplasias , Derrame Pleural Maligno , Pleurodesia , Anciano , Antineoplásicos Inmunológicos/farmacología , Catéteres de Permanencia/estadística & datos numéricos , Correlación de Datos , Intervención Médica Temprana/métodos , Femenino , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inmunoterapia/métodos , Masculino , Neoplasias/clasificación , Neoplasias/complicaciones , Neoplasias/genética , Neoplasias/terapia , Neoplasias Hormono-Dependientes/complicaciones , Neoplasias Hormono-Dependientes/terapia , Derrame Pleural Maligno/diagnóstico por imagen , Derrame Pleural Maligno/epidemiología , Derrame Pleural Maligno/etiología , Derrame Pleural Maligno/terapia , Pleurodesia/métodos , Pleurodesia/estadística & datos numéricos , Estudios Retrospectivos , Reino Unido/epidemiología
14.
Anticancer Res ; 41(5): 2669-2680, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33952498

RESUMEN

BACKGROUND/AIM: The histopathological variability of each type of pituitary adenoma (PA) that causes growth hormone (GH) excess influences the phenotype, radiological characteristics and therapy response of acromegaly patients. We correlated the immunohistochemical (IHC) features of GH-secreting PAs with their clinical, laboratory and imaging data. PATIENTS AND METHODS: We included 32 patients with documented acromegaly; tumour specimens were histologically and IHC examined: anterior pituitary hormones, pituitary-specific transcription factor-1 (PIT-1), Ki-67 labelling index were evaluated. RESULTS: Macroadenomas represented 93.75%. Post-surgery disease control negatively correlated with the maximum initial tumour diameter (p=0.04). Ki-67 did not predict remission. No correlation was found between GH serum levels and IHC expression (p=0.45). PIT-1 was positive in all specimens, two had a weak expression. Four were considered PIT-1 positive plurihormonal adenomas and several had unusual IHC combinations. CONCLUSION: PIT-1 accurately classifies GH-secreting PAs. The IHC classification as well as radiological dimensions and extent influence disease control, probably being the best prognosis factors.


Asunto(s)
Acromegalia/sangre , Hormona del Crecimiento/sangre , Neoplasias Hormono-Dependientes/sangre , Neoplasias Hipofisarias/sangre , Acromegalia/complicaciones , Acromegalia/genética , Acromegalia/patología , Adulto , Anciano , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Antígeno Ki-67/sangre , Masculino , Persona de Mediana Edad , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/patología , Hormonas Adenohipofisarias/sangre , Neoplasias Hipofisarias/complicaciones , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/patología , Periodo Preoperatorio , Factor de Transcripción Pit-1/sangre
15.
Prostate ; 81(9): 572-579, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33955569

RESUMEN

BACKGROUND: Clinical outcomes may differ among patients presenting with primary (de novo) metastatic hormone-sensitive prostate cancer (mHSPC) versus secondary (metachronous) mHSPC occurring after local therapy. It is unknown what molecular features distinguish these potentially distinct presentations. METHODS: A single-center retrospective study of mHSPC patients classified as primary mHSPC (n = 121) or secondary mHSPC (n = 106). A targeted set of genes was analyzed: BRCA2, PTEN, RB1, TP53, SPOP, CDK12, any two out of PTEN/RB1/TP53 alterations, and homologous recombination deficiency mutations. TP53 mutations were categorized as loss-of-function (LOF) versus dominant-negative (DN). The impacts of genetic features on progression-free survival (PFS) and overall survival (OS) were assessed using univariate and multivariate Cox proportional hazards regression. RESULTS: Median PFS was 15 and 30 months for men with primary and secondary mHSPC, respectively (hazard ratio: 0.57, 95% confidence interval: 0.41-0.78; p < .01). OS did not show a significant difference between groups. There were more men with Gleason 8-10 disease in the primary versus secondary mHSPC groups (83% vs. 68%; p < .01). In univariate and multivariate analyses, TP53 DN mutations showed a statistically significant association with OS for the entire mHSPC population. Conversely, SPOP mutations were associated with improved OS. Additionally, TP53 mutations (DN and LOF) were associated with worse OS for secondary mHSPC. A combination of PTEN/RB1/TP53 mutations was associated with worse OS and PFS for secondary mHSPC, while no genomic alteration affected outcomes for primary mHSPC. CONCLUSIONS: TP53 DN mutations, but not all TP53 alterations, were the strongest predictor of negative outcomes in men with mHSPC, while SPOP mutations were associated with improved outcomes. In subgroup analyses, specific alterations were prognostic of outcomes in secondary, but not primary, mHSPC.


Asunto(s)
Genes p53/genética , Neoplasias Hormono-Dependientes , Neoplasias Primarias Secundarias , Proteínas Nucleares/genética , Neoplasias de la Próstata , Proteínas Represoras/genética , Antagonistas de Andrógenos/uso terapéutico , Humanos , Masculino , Persona de Mediana Edad , Mutación , Clasificación del Tumor , Metástasis de la Neoplasia/genética , Estadificación de Neoplasias , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Neoplasias Hormono-Dependientes/genética , Neoplasias Hormono-Dependientes/patología , Neoplasias Primarias Secundarias/tratamiento farmacológico , Neoplasias Primarias Secundarias/genética , Neoplasias Primarias Secundarias/patología , Pronóstico , Supervivencia sin Progresión , Modelos de Riesgos Proporcionales , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Estudios Retrospectivos
16.
Br J Cancer ; 125(4): 495-509, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33911195

RESUMEN

Obesity is a risk factor for at least 13 different types of cancer, many of which are hormonally driven, and is associated with increased cancer incidence and morbidity. Adult obesity rates are steadily increasing and a subsequent increase in cancer burden is anticipated. Obesity-related dysfunction can contribute to cancer pathogenesis and treatment resistance through various mechanisms, including those mediated by insulin, leptin, adipokine, and aromatase signalling pathways, particularly in women. Furthermore, adiposity-related changes can influence tumour vascularity and inflammation in the tumour microenvironment, which can support tumour development and growth. Trials investigating non-pharmacological approaches to target the mechanisms driving obesity-mediated cancer pathogenesis are emerging and are necessary to better appreciate the interplay between malignancy, adiposity, diet and exercise. Diet, exercise and bariatric surgery are potential strategies to reverse the cancer-promoting effects of obesity; trials of these interventions should be conducted in a scientifically rigorous manner with dose escalation and appropriate selection of tumour phenotypes and have cancer-related clinical and mechanistic endpoints. We are only beginning to understand the mechanisms by which obesity effects cell signalling and systemic factors that contribute to oncogenesis. As the rates of obesity and cancer increase, we must promote the development of non-pharmacological lifestyle trials for the treatment and prevention of malignancy.


Asunto(s)
Neoplasias Hormono-Dependientes/prevención & control , Obesidad/terapia , Cirugía Bariátrica , Ensayos Clínicos como Asunto , Dietoterapia , Terapia por Ejercicio , Femenino , Humanos , Masculino , Neoplasias Hormono-Dependientes/etiología , Neoplasias Hormono-Dependientes/inmunología , Obesidad/complicaciones , Obesidad/inmunología
17.
Clin Cancer Res ; 27(10): 2706-2711, 2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-33648989

RESUMEN

The dramatic impact of the COVID-19 pandemic has resulted in an "all hands on deck" approach to find new therapies to improve outcomes in this disease. In addition to causing significant respiratory pathology, infection with SARS-CoV-2 (like infection with other respiratory viruses) directly or indirectly results in abnormal vasculature, which may contribute to hypoxemia. These vascular effects cause significant morbidity and may contribute to mortality from the disease. Given that abnormal vasculature and poor oxygenation are also hallmarks of solid tumors, lessons from the treatment of cancer may help identify drugs that can be repurposed to treat COVID-19. Although the mechanisms that result in vascular abnormalities in COVID-19 are not fully understood, it is possible that there is dysregulation of many of the same angiogenic and thrombotic pathways as seen in patients with cancer. Many anticancer therapeutics, including androgen deprivation therapy (ADT) and immune checkpoint blockers (ICB), result in vascular normalization in addition to their direct effects on tumor cells. Therefore, these therapies, which have been extensively explored in clinical trials of patients with cancer, may have beneficial effects on the vasculature of patients with COVID-19. Furthermore, these drugs may have additional effects on the disease course, as some ADTs may impact viral entry, and ICBs may accelerate T-cell-mediated viral clearance. These insights from the treatment of cancer may be leveraged to abrogate the vascular pathologies found in COVID-19 and other forms of hypoxemic respiratory failure.


Asunto(s)
Antagonistas de Andrógenos/uso terapéutico , Vasos Sanguíneos/efectos de los fármacos , COVID-19/prevención & control , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Neovascularización Patológica/tratamiento farmacológico , Neoplasias de la Próstata/tratamiento farmacológico , Vasos Sanguíneos/patología , Vasos Sanguíneos/fisiopatología , COVID-19/epidemiología , COVID-19/virología , Ensayos Clínicos como Asunto , Progresión de la Enfermedad , Humanos , Masculino , Neoplasias Hormono-Dependientes/irrigación sanguínea , Evaluación de Resultado en la Atención de Salud , Pandemias , Neoplasias de la Próstata/irrigación sanguínea , Factores de Riesgo , SARS-CoV-2/fisiología
18.
Prostate Cancer Prostatic Dis ; 24(3): 623-637, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33568748

RESUMEN

BACKGROUND: Many systemic therapies for advanced prostate cancer work by disrupting androgen receptor signaling. Androgen indifferent prostate cancer (AIPC) variants, including aggressive variant prostate cancer (AVPC), neuroendocrine prostate cancer (NEPC), and double-negative prostate cancer (DNPC), are increasingly common and often overlapping resistance phenotypes following treatment with androgen receptor signaling inhibitors in men with metastatic castration-resistant prostate cancer and are associated with poor outcomes. Understanding the underlying biology and identifying effective therapies for AIPC is paramount for improving survival for men with prostate cancer. METHODS: In this review, we summarize the current knowledge on AIPC variants, including our current understanding of the clinical, morphologic, and molecular features as well as current therapeutic approaches. We also explore emerging therapies and biomarkers aimed at improving outcomes for men with AIPC. RESULTS AND CONCLUSIONS: Establishing consensus definitions, developing novel biomarkers for early and accurate detection, further characterization of molecular drivers of each phenotype, and developing effective therapies will be critical to improving outcomes for men with AIPC. Significant progress has been made toward defining the clinical and molecular characteristics of AVPC, NEPC, and DNPC. Novel diagnostic approaches, including cell-free DNA, circulating tumor cells, and molecular imaging are promising tools for detecting AIPC in clinical practice. Building on previous treatment advances, several clinical trials are underway evaluating novel therapeutic approaches in patients with AIPC informed by an understanding of variant-specific biology. In this review, we discuss how these recent and ongoing studies will help to improve diagnosis, prognosis, and therapy for men with AIPC.


Asunto(s)
Antineoplásicos Hormonales/uso terapéutico , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Células Neoplásicas Circulantes/patología , Neoplasias de la Próstata/tratamiento farmacológico , Humanos , Masculino , Neoplasias Hormono-Dependientes/patología , Pronóstico , Neoplasias de la Próstata/patología
19.
Prostate Cancer Prostatic Dis ; 24(3): 725-732, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33495569

RESUMEN

BACKGROUND: Androgen deprivation therapy (ADT) for prostate cancer (PC) has detrimental effects on physical function and quality of life (QoL), but the addition of androgen receptor signalling inhibitors (ARSI) on these outcomes is unclear. PURPOSE: To compare body composition, physical function, and QoL across progressive stages of PC and non-cancer controls (CON). METHODS: In men with hormone sensitive PC (HSPC, n = 43) or metastatic castration-resistant PC (mCRPC, n = 22) or CON (n = 37), relative and absolute lean and fat mass, physical function (6 m walk, chair stands, timed up and go [TUG], stair climb), and QoL were determined. RESULTS: Relative body composition differed amongst all groups, along with ~39% greater absolute fat mass in mCRPC vs. CON. TUG and chair stands were ~71% and ~33% slower in mCRPC compared to both CON and HSPC, whereas stair climb was ~29% and 6 m walk was ~18% slower in mCRPC vs. CON. Relative body composition was correlated with physical function (r = 0.259-0.385). Clinically relevant differences for mCRPC were observed for overall QoL and several subscales vs. CON, although body composition and physical function did not influence QoL. CONCLUSIONS: PC progression is associated with deteriorations in body composition and physical function. As ADT length was similar between groups, ARSI use for mCRPC likely contributed in part to these changes. Given the difficulties of improving lean mass during ADT, interventions that reduce adiposity may lessen the side effects of hormone therapy.


Asunto(s)
Antagonistas de Andrógenos/uso terapéutico , Composición Corporal , Ejercicio Físico , Neoplasias Hormono-Dependientes/patología , Neoplasias de la Próstata/patología , Calidad de Vida , Caminata , Anciano , Estudios de Casos y Controles , Estudios Transversales , Estudios de Seguimiento , Humanos , Masculino , Neoplasias Hormono-Dependientes/tratamiento farmacológico , Neoplasias Hormono-Dependientes/psicología , Pronóstico , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/psicología
20.
Prog Urol ; 31(5): 243-244, 2021 04.
Artículo en Francés | MEDLINE | ID: mdl-33468416
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...