Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
NPJ Breast Cancer ; 9(1): 50, 2023 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-37268625

RESUMEN

Inflammatory breast cancer (IBC) is a rare, aggressive form of breast cancer that presents as de novo metastatic disease in 20-30% of cases, with one-third of cases demonstrating HER2-positivity. There has been limited investigation into locoregional therapy utilization following HER2-directed systemic therapy for these patients, and their locoregional progression or recurrence (LRPR) and survival outcomes. Patients with de novo HER2-positive metastatic IBC (mIBC) were identified from an IRB-approved IBC registry at Dana-Farber Cancer Institute. Clinical, pathology, and treatment data were abstracted. Rates of LRPR, progression-free survival (PFS), overall survival (OS), and pathologic complete response (pCR) were determined. Seventy-eight patients diagnosed between 1998 and 2019 were identified. First-line systemic therapy comprised chemotherapy for most patients (97.4%) and HER2-directed therapy for all patients (trastuzumab [47.4%]; trastuzumab+pertuzumab [51.3%]; or trastuzumab emtansine [1.3%]). At a median follow-up of 2.7 years, the median PFS was 1.0 year, and the median OS was 4.6 years. The 1- and 2-year cumulative incidence of LRPR was 20.7% and 29.0%, respectively. Mastectomy was performed after systemic therapy in 41/78 patients (52.6%); 10 had a pCR (24.4%) and all were alive at last follow-up (1.3-8.9 years after surgery). Among 56 patients who were alive and LRPR-free at one year, 10 developed LRPR (surgery group = 1; no-surgery group = 9). In conclusion, patients with de novo HER2-positive mIBC who undergo surgery have favorable outcomes. More than half of patients received systemic and local therapy with good locoregional control and prolonged survival, suggesting a potential role for local therapy.

2.
bioRxiv ; 2023 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-37034710

RESUMEN

Tumors acquire an increased ability to obtain and metabolize nutrients. Here, we engineered and implanted adipocytes to outcompete tumors for nutrients and show that they can substantially reduce cancer progression. Growing cells or xenografts from several cancers (breast, colon, pancreas, prostate) alongside engineered human adipocytes or adipose organoids significantly suppresses cancer progression and reduces hypoxia and angiogenesis. Transplanting modulated adipocyte organoids in pancreatic or breast cancer mouse models nearby or distal from the tumor significantly suppresses its growth. To further showcase therapeutic potential, we demonstrate that co-culturing tumor organoids derived from human breast cancers with engineered patient-derived adipocytes significantly reduces cancer growth. Combined, our results introduce a novel cancer therapeutic approach, termed adipose modulation transplantation (AMT), that can be utilized for a broad range of cancers.

3.
Dev Cell ; 57(11): 1400-1420.e7, 2022 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-35617956

RESUMEN

The breast is a dynamic organ whose response to physiological and pathophysiological conditions alters its disease susceptibility, yet the specific effects of these clinical variables on cell state remain poorly annotated. We present a unified, high-resolution breast atlas by integrating single-cell RNA-seq, mass cytometry, and cyclic immunofluorescence, encompassing a myriad of states. We define cell subtypes within the alveolar, hormone-sensing, and basal epithelial lineages, delineating associations of several subtypes with cancer risk factors, including age, parity, and BRCA2 germline mutation. Of particular interest is a subset of alveolar cells termed basal-luminal (BL) cells, which exhibit poor transcriptional lineage fidelity, accumulate with age, and carry a gene signature associated with basal-like breast cancer. We further utilize a medium-depletion approach to identify molecular factors regulating cell-subtype proportion in organoids. Together, these data are a rich resource to elucidate diverse mammary cell states.


Asunto(s)
Neoplasias de la Mama , Transcriptoma , Animales , Mama , Neoplasias de la Mama/genética , Femenino , Humanos , Glándulas Mamarias Animales , Embarazo , Proteómica , Transcriptoma/genética
4.
Cancer Res Commun ; 2(12): 1579-1589, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36970720

RESUMEN

Ductal carcinoma in situ (DCIS) is a biologically heterogenous entity with uncertain risk for invasive ductal carcinoma (IDC) development. Standard treatment is surgical resection often followed by radiation. New approaches are needed to reduce overtreatment. This was an observational study that enrolled patients with DCIS who chose not to pursue surgical resection from 2002 to 2019 at a single academic medical center. All patients underwent breast MRI exams at 3- to 6-month intervals. Patients with hormone receptor-positive disease received endocrine therapy. Surgical resection was strongly recommended if clinical or radiographic evidence of disease progression developed. A recursive partitioning (R-PART) algorithm incorporating breast MRI features and endocrine responsiveness was used retrospectively to stratify risk of IDC. A total of 71 patients were enrolled, 2 with bilateral DCIS (73 lesions). A total of 34 (46.6%) were premenopausal, 68 (93.2%) were hormone-receptor positive, and 60 (82.1%) were intermediate- or high-grade lesions. Mean follow-up time was 8.5 years. Over half (52.1%) remained on active surveillance without evidence of IDC with mean duration of 7.4 years. Twenty patients developed IDC, of which 6 were HER2 positive. DCIS and subsequent IDC had highly concordant tumor biology. Risk of IDC was characterized by MRI features after 6 months of endocrine therapy exposure; low-, intermediate-, and high-risk groups were identified with respective IDC rates of 8.7%, 20.0%, and 68.2%. Thus, active surveillance consisting of neoadjuvant endocrine therapy and serial breast MRI may be an effective tool to risk-stratify patients with DCIS and optimally select medical or surgical management. Significance: A retrospective analysis of 71 patients with DCIS who did not undergo upfront surgery demonstrated that breast MRI features after short-term exposure to endocrine therapy identify those at high (68.2%), intermediate (20.0%), and low risk (8.7%) of IDC. With 7.4 years mean follow-up, 52.1% of patients remain on active surveillance. A period of active surveillance offers the opportunity to risk-stratify DCIS lesions and guide decisions for operative management.


Asunto(s)
Neoplasias de la Mama , Carcinoma Ductal de Mama , Carcinoma Intraductal no Infiltrante , Humanos , Femenino , Carcinoma Intraductal no Infiltrante/diagnóstico por imagen , Estudios Retrospectivos , Carcinoma Ductal de Mama/patología , Terapia Neoadyuvante , Espera Vigilante , Neoplasias de la Mama/diagnóstico por imagen
5.
Am J Clin Oncol ; 44(9): 449-455, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34149037

RESUMEN

OBJECTIVES: Dermal lymphatic invasion (DLI) with tumor emboli is a common pathologic characteristic of inflammatory breast cancer (IBC), although its presence is not required for diagnosis. We examined whether documented DLI on skin biopsy was associated with survival and time to recurrence or progression in IBC. MATERIALS AND METHODS: A total of 340 women enrolled in the IBC Registry at Dana-Farber Cancer Institute between 1997 and 2019 were included in this study. Kaplan-Meier curves and multivariable Cox proportional hazards models were used to estimate hazard ratios and 95% confidence intervals for associations of DLI and overall survival, time to locoregional recurrence/progression, and distant metastasis by stage at presentation. RESULTS: DLI was detected in 215 (63.2%) of IBC cases overall. At disease presentation, IBC with DLI had a higher prevalence of de novo metastases (37.7% vs. 26.4%), breast skin ulceration (6.1% vs. 2.4%), and lymphovascular invasion within the breast parenchyma (52.9% vs. 25.5%) and a lower prevalence of palpable breast mass (48.2% vs. 70.6%) than IBC without DLI. Over a median follow-up of 2.0 years, 147 deaths occurred. DLI was not associated with survival or recurrence in multivariable models (all P ≥0.10). For example, among women with stage III disease, hazard ratios (95% confidence intervals) for DLI presence was 1.29 (0.77-2.15) for overall survival, 1.29 (0.56-3.00) for locoregional recurrence, and 1.71 (0.97-3.02) for distant metastasis. CONCLUSION: Although the extent of tumor emboli in dermal lymphatics may be associated with biological features of IBC, DLI was not an independent prognostic marker of clinical outcomes in this study.


Asunto(s)
Neoplasias Inflamatorias de la Mama/mortalidad , Neoplasias Inflamatorias de la Mama/patología , Biopsia , Terapia Combinada , Femenino , Humanos , Neoplasias Inflamatorias de la Mama/terapia , Metástasis Linfática/patología , Persona de Mediana Edad , Análisis Multivariante , Recurrencia Local de Neoplasia , Modelos de Riesgos Proporcionales , Análisis de Supervivencia
6.
Nat Protoc ; 16(4): 1936-1965, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33692550

RESUMEN

Organoid technology has revolutionized the study of human organ development, disease and therapy response tailored to the individual. Although detailed protocols are available for the generation and long-term propagation of human organoids from various organs, such methods are lacking for breast tissue. Here we provide an optimized, highly versatile protocol for long-term culture of organoids derived from either normal human breast tissues or breast cancer (BC) tissues, as well as culturing conditions for a panel of 45 biobanked samples, including BC organoids covering all major disease subtypes (triple-negative, estrogen receptor-positive/progesterone receptor-positive and human epidermal growth receptor 2-positive). Additionally, we provide methods for genetic manipulation by Lipofectamine 2000, electroporation or lentivirus and subsequent organoid selection and clonal culture. Finally, we introduce an optimized method for orthotopic organoid transplantation in mice, which includes injection of organoids and estrogen pellets without the need for surgery. Organoid derivation from tissue fragments until the first split takes 7-21 d; generation of genetically manipulated clonal organoid cultures takes 14-21 d; and organoid expansion for xenotransplantation takes >4 weeks.


Asunto(s)
Neoplasias de la Mama/patología , Mama/patología , Técnicas de Cultivo de Célula/métodos , Técnicas Genéticas , Organoides/patología , Trasplante Heterólogo , Animales , Bancos de Muestras Biológicas , Células Clonales , Femenino , Humanos , Ratones , Factores de Tiempo
7.
Breast Cancer Res Treat ; 181(2): 383-390, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32318957

RESUMEN

PURPOSE: Optimizing treatment strategies for patients with inflammatory breast cancer (IBC) relies on accurate initial staging. This study compared contrast-enhanced computed tomography (ce-CT) and FDG-PET/CT for initial staging of IBC to determine the frequency of discordance between the two imaging modalities and potential impact on management. METHODS: 81 patients with IBC underwent FDG-PET/CT and ce-CT prior to starting treatment. FDG-PET/CT and ce-CT scans were independently reviewed for locoregional and distant metastases and findings recorded by anatomic site as negative, equivocal, or positive for breast cancer involvement. Each paired ce-CT and FDG-PET/CT case was classified as concordant or discordant for findings. Discordant findings were subclassified as (a) related to the presence or absence of distant metastases; (b) affecting the locoregional radiation therapy plan; or (c) due to incidental findings not related to IBC. RESULTS: There were 47 discordant findings between ce-CT and FDG-PET/CT in 41 of 81 patients (50.6%). Thirty (63.8%) were related to the presence or absence of distant metastases; most commonly disease detection on FDG-PET/CT but not ce-CT (n = 12). FDG-PET/CT suggested alterations of the locoregional radiation therapy plan designed by CT alone in 15 patients. FDG-PET/CT correctly characterized 5 of 7 findings equivocal for metastatic IBC on ce-CT. CONCLUSIONS: This study demonstrates differences between ce-CT and FDG-PET/CT for initial staging of IBC and how these differences potentially affect patient management. Preliminary data suggest that FDG-PET/CT may be the imaging modality of choice for initial staging of IBC. Prospective trials testing initial staging with FDG-PET/CT versus important clinical end-points in IBC are warranted.


Asunto(s)
Carcinoma Ductal de Mama/diagnóstico , Carcinoma Lobular/diagnóstico , Fluorodesoxiglucosa F18/metabolismo , Neoplasias Inflamatorias de la Mama/diagnóstico , Planificación de Atención al Paciente/normas , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Tomografía Computarizada por Rayos X/métodos , Adulto , Anciano , Carcinoma Ductal de Mama/diagnóstico por imagen , Carcinoma Ductal de Mama/metabolismo , Carcinoma Lobular/diagnóstico por imagen , Carcinoma Lobular/metabolismo , Diagnóstico Diferencial , Femenino , Estudios de Seguimiento , Humanos , Neoplasias Inflamatorias de la Mama/diagnóstico por imagen , Neoplasias Inflamatorias de la Mama/metabolismo , Persona de Mediana Edad , Estadificación de Neoplasias , Radiofármacos/metabolismo , Receptor ErbB-2/metabolismo , Receptores de Estrógenos/metabolismo , Receptores de Progesterona/metabolismo , Estudios Retrospectivos
8.
Nat Commun ; 11(1): 1711, 2020 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-32249764

RESUMEN

Recently, organoid technology has been used to generate a large repository of breast cancer organoids. Here we present an extensive evaluation of the ability of organoid culture technology to preserve complex stem/progenitor and differentiated cell types via long-term propagation of normal human mammary tissues. Basal/stem and luminal progenitor cells can differentiate in culture to generate mature basal and luminal cell types, including ER+ cells that have been challenging to maintain in culture. Cells associated with increased cancer risk can also be propagated. Single-cell analyses of matched organoid cultures and native tissues by mass cytometry for 38 markers provide a higher resolution representation of the multiple mammary epithelial cell types in the organoids, and demonstrate that protein expression patterns of the tissue of origin can be preserved in culture. These studies indicate that organoid cultures provide a valuable platform for studies of mammary differentiation, transformation, and breast cancer risk.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Linaje de la Célula , Glándulas Mamarias Humanas/citología , Organoides/citología , Organoides/metabolismo , Células Madre/citología , Adulto , Proteína BRCA1/genética , Neoplasias de la Mama , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Linaje de la Célula/genética , Factor de Crecimiento Epidérmico/farmacología , Receptores ErbB/metabolismo , Femenino , Humanos , Glándulas Mamarias Humanas/química , Glándulas Mamarias Humanas/metabolismo , Persona de Mediana Edad , Organoides/química , Análisis de la Célula Individual , Células Madre/química , Células Madre/metabolismo , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Adulto Joven , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
9.
Nat Biotechnol ; 38(2): 199-209, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31844290

RESUMEN

Prediction of HLA epitopes is important for the development of cancer immunotherapies and vaccines. However, current prediction algorithms have limited predictive power, in part because they were not trained on high-quality epitope datasets covering a broad range of HLA alleles. To enable prediction of endogenous HLA class I-associated peptides across a large fraction of the human population, we used mass spectrometry to profile >185,000 peptides eluted from 95 HLA-A, -B, -C and -G mono-allelic cell lines. We identified canonical peptide motifs per HLA allele, unique and shared binding submotifs across alleles and distinct motifs associated with different peptide lengths. By integrating these data with transcript abundance and peptide processing, we developed HLAthena, providing allele-and-length-specific and pan-allele-pan-length prediction models for endogenous peptide presentation. These models predicted endogenous HLA class I-associated ligands with 1.5-fold improvement in positive predictive value compared with existing tools and correctly identified >75% of HLA-bound peptides that were observed experimentally in 11 patient-derived tumor cell lines.


Asunto(s)
Bases de Datos de Proteínas , Epítopos/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Péptidos/metabolismo , Proteoma/metabolismo , Algoritmos , Alelos , Secuencias de Aminoácidos , Línea Celular , Sitios Genéticos , Humanos , Ligandos , Péptido Hidrolasas/metabolismo , Péptidos/química , Complejo de la Endopetidasa Proteasomal/metabolismo
10.
Curr Oncol Rep ; 21(10): 86, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31414257

RESUMEN

PURPOSE OF REVIEW: Inflammatory breast cancer (IBC) is an uncommon but highly aggressive subtype of breast cancer that contributes significantly to breast cancer-related mortality. In this review, we provide an overview of the clinical and molecular characteristics of IBC, and highlight some areas of need for ongoing research. RECENT FINDINGS: The disease is characterized by florid tumor emboli that obstruct dermal lymphatics, leading to swelling and inflammation of the affected breast. Recent studies have focused on tumor cell intrinsic features, such as signaling through pathways involved in growth and stem-like behavior, as well as extrinsic features, such as the immune system, that can be leveraged to develop new potential therapies. Key efforts have led to an increase in awareness of the disease as well as new insights into IBC pathogenesis. However, there is a strong need for new therapies designed specifically for IBC, and many unanswered questions remain.


Asunto(s)
Neoplasias Inflamatorias de la Mama/patología , Neoplasias Inflamatorias de la Mama/terapia , Femenino , Humanos , Neoplasias Inflamatorias de la Mama/clasificación , Pronóstico
11.
Cell Metab ; 29(5): 1166-1181.e6, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-30799286

RESUMEN

Cells are subjected to oxidative stress during the initiation and progression of tumors, and this imposes selective pressure for cancer cells to adapt mechanisms to tolerate these conditions. Here, we examined the dependency of cancer cells on glutathione (GSH), the most abundant cellular antioxidant. While cancer cell lines displayed a broad range of sensitivities to inhibition of GSH synthesis, the majority were resistant to GSH depletion. To identify cellular pathways required for this resistance, we carried out genetic and pharmacologic screens. Both approaches revealed that inhibition of deubiquitinating enzymes (DUBs) sensitizes cancer cells to GSH depletion. Inhibition of GSH synthesis, in combination with DUB inhibition, led to an accumulation of polyubiquitinated proteins, induction of proteotoxic stress, and cell death. These results indicate that depletion of GSH renders cancer cells dependent on DUB activity to maintain protein homeostasis and cell viability and reveal a potentially exploitable vulnerability for cancer therapy.


Asunto(s)
Antioxidantes/metabolismo , Supervivencia Celular/efectos de los fármacos , Enzimas Desubicuitinizantes/metabolismo , Glutatión/metabolismo , Proteostasis/efectos de los fármacos , Células A549 , Aminopiridinas/farmacología , Animales , Butionina Sulfoximina/farmacología , Dominio Catalítico/efectos de los fármacos , Enzimas Desubicuitinizantes/antagonistas & inhibidores , Femenino , Glutamato-Cisteína Ligasa/antagonistas & inhibidores , Glutamato-Cisteína Ligasa/química , Glutamato-Cisteína Ligasa/metabolismo , Humanos , Células MCF-7 , Glándulas Mamarias Animales/citología , Glándulas Mamarias Humanas/citología , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Organoides/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Tiocianatos/farmacología , Carga Tumoral/efectos de los fármacos , Proteínas Ubiquitinadas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Clin Cancer Res ; 23(15): 4035-4045, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28270498

RESUMEN

Purpose: Because of inherent disease heterogeneity, targeted therapies have eluded triple-negative breast cancer (TNBC), and biomarkers predictive of treatment response have not yet been identified. This study was designed to determine whether the mTOR inhibitor everolimus with cisplatin and paclitaxel would provide synergistic antitumor effects in TNBC.Methods: Patients with stage II/III TNBC were enrolled in a randomized phase II trial of preoperative weekly cisplatin, paclitaxel and daily everolimus or placebo for 12 weeks, until definitive surgery. Tumor specimens were obtained at baseline, cycle 1, and surgery. Primary endpoint was pathologic complete response (pCR); secondary endpoints included clinical responses, breast conservation rate, safety, and discovery of molecular features associated with outcome.Results: Between 2009 and 2013, 145 patients were accrued; 36% of patients in the everolimus arm and 49% of patients in the placebo arm achieved pCR; in each arm, 50% of patients achieved complete responses by imaging. Higher rates of neutropenia, mucositis, and transaminase elevation were seen with everolimus. Clinical response to therapy and long-term outcome correlated with increased frequency of DNA damage response (DDR) gene mutations, Basal-like1 and Mesenchymal TNBC-subtypes, AR-negative status, and high Ki67, but not with tumor-infiltrating lymphocytes.Conclusions: The paclitaxel/cisplatin combination was well tolerated and active, but addition of everolimus was associated with more adverse events without improvement in pCR or clinical response. However, discoveries made from correlative studies could lead to predictive TNBC biomarkers that may impact clinical decision-making and provide new avenues for mechanistic exploration that could lead to clinical utility. Clin Cancer Res; 23(15); 4035-45. ©2017 AACR.


Asunto(s)
Cisplatino/administración & dosificación , Everolimus/administración & dosificación , Paclitaxel/administración & dosificación , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Adulto , Cisplatino/efectos adversos , Daño del ADN/efectos de los fármacos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/genética , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/patología , Everolimus/efectos adversos , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Antígeno Ki-67/genética , Linfocitos Infiltrantes de Tumor/patología , Persona de Mediana Edad , Mutación , Estadificación de Neoplasias , Paclitaxel/efectos adversos , Receptores Androgénicos/genética , Resultado del Tratamiento , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología
13.
Cell Rep ; 14(10): 2289-300, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26947080

RESUMEN

We report that p73 is expressed in multiciliated cells (MCCs), is required for MCC differentiation, and directly regulates transcriptional modulators of multiciliogenesis. Loss of ciliary biogenesis provides a unifying mechanism for many phenotypes observed in p73 knockout mice including hydrocephalus; hippocampal dysgenesis; sterility; and chronic inflammation/infection of lung, middle ear, and sinus. Through p73 and p63 ChIP-seq using murine tracheal cells, we identified over 100 putative p73 target genes that regulate MCC differentiation and homeostasis. We validated Foxj1, a transcriptional regulator of multiciliogenesis, and many other cilia-associated genes as direct target genes of p73 and p63. We show p73 and p63 are co-expressed in a subset of basal cells and suggest that p73 marks these cells for MCC differentiation. In summary, p73 is essential for MCC differentiation, functions as a critical regulator of a transcriptome required for MCC differentiation, and, like p63, has an essential role in development of tissues.


Asunto(s)
Cilios/metabolismo , Factores de Transcripción Forkhead/metabolismo , Redes Reguladoras de Genes , Pulmón/metabolismo , Proteína Tumoral p73/metabolismo , Animales , Bronquiolos/metabolismo , Bronquiolos/patología , Diferenciación Celular , Células Cultivadas , Cilios/patología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Epitelio/metabolismo , Epitelio/patología , Femenino , Factores de Transcripción Forkhead/genética , Pulmón/citología , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Fosfoproteínas/deficiencia , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Interferencia de ARN , Análisis de Secuencia de ARN , Tráquea/metabolismo , Tráquea/patología , Transactivadores/deficiencia , Transactivadores/genética , Transactivadores/metabolismo , Transcriptoma , Proteína Tumoral p73/deficiencia , Proteína Tumoral p73/genética
14.
Proc Natl Acad Sci U S A ; 108(5): 2076-81, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21245298

RESUMEN

The transcription factor p73 plays critical roles during development and tumorigenesis. It exhibits sequence identity and structural homology with p53, and can engage p53-like tumor-suppressive programs. However, different pathways regulate p53 and p73, and p73 is not mutated in human tumors. Therefore, p73 represents a therapeutic target, and there is a critical need to understand genes and noncoding RNAs regulated by p73 and how they change during treatment regimens. Here, we define the p73 genomic binding profile and demonstrate its modulation by rapamycin, an inhibitor of mammalian target of rapamycin (mTOR) and inducer of p73. Rapamycin selectively increased p73 occupancy at a subset of its binding sites. In addition, multiple determinants of p73 binding, activity, and function were evident, and were modulated by mTOR. We generated an mTOR-p73 signature that is enriched for p73 target genes and miRNAs that are involved in mesenchymal differentiation and tumorigenesis, can classify rhabdomyosarcomas by clinical subtype, and can predict patient outcome.


Asunto(s)
Transformación Celular Neoplásica , Proteínas de Unión al ADN/genética , Mesodermo/patología , Proteínas Nucleares/genética , Serina-Treonina Quinasas TOR/genética , Transcripción Genética , Proteínas Supresoras de Tumor/genética , Proteínas de Unión al ADN/metabolismo , Genoma Humano , Humanos , MicroARNs/genética , Proteínas Nucleares/metabolismo , Unión Proteica , Rabdomiosarcoma/clasificación , Rabdomiosarcoma/genética , Rabdomiosarcoma/patología , Proteína Tumoral p73 , Proteínas Supresoras de Tumor/metabolismo
15.
Mol Cancer ; 9: 95, 2010 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-20429933

RESUMEN

BACKGROUND: Autophagy is characterized by the sequestration of cytoplasm and organelles into multimembrane vesicles and subsequent degradation by the cell's lysosomal system. It is linked to many physiological functions in human cells including stress response, protein degradation, organelle turnover, caspase-independent cell death and tumor suppression. Malignant transformation is frequently associated with deregulation of autophagy and several tumor suppressors can modulate autophagic processes. The tumor suppressor p53 can induce autophagy after metabolic or genotoxic stress through transcriptionally-dependent and -independent mechanisms. In this study we expand on the former mechanism by functionally characterizing a p53 family target gene, ISG20L1 under conditions of genotoxic stress. RESULTS: We identified a p53 target gene, ISG20L1, and show that transcription of the gene can be regulated by all three p53 family members (p53, p63, and p73). We generated an antibody to ISG20L1 and found that it localizes to the nucleolar and perinucleolar regions of the nucleus and its protein levels increase in a p53- and p73-dependent manner after various forms of genotoxic stress. When ectopically expressed in epithelial cancer-derived cell lines, ISG20L1 expression decreased clonogenic survival without a concomitant elevation in apoptosis and this effect was partially rescued in cells that were ATG5 deficient. Knockdown of ISG20L1 did not alter 5-FU induced apoptosis as assessed by PARP and caspase-3 cleavage, sub-G1 content, and DNA laddering. Thus, we investigated the role of ISG20L1 in autophagy, a process commonly associated with type II cell death, and found that ISG20L1 knockdown decreased levels of autophagic vacuoles and LC3-II after genotoxic stress as assessed by electron microscopy, biochemical, and immunohistochemical measurements of LC3-II. CONCLUSIONS: Our identification of ISG20L1 as a p53 family target and discovery that modulation of this target can regulate autophagic processes further strengthens the connection between p53 signaling and autophagy. Given the keen interest in targeting autophagy as an anticancer therapeutic approach in tumor cells that are defective in apoptosis, investigation of genes and signaling pathways involved in cell death associated with autophagy is critical.


Asunto(s)
Autofagia/fisiología , Daño del ADN/fisiología , Exodesoxirribonucleasas/metabolismo , Regulación de la Expresión Génica , Proteína p53 Supresora de Tumor/metabolismo , Western Blotting , Línea Celular , Separación Celular , Inmunoprecipitación de Cromatina , Exodesoxirribonucleasas/genética , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Microscopía Electrónica de Transmisión , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transfección , Proteína p53 Supresora de Tumor/genética
16.
Clin Cancer Res ; 16(2): 681-90, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20068102

RESUMEN

PURPOSE: To identify molecular markers of pathologic response to neoadjuvant paclitaxel/radiation treatment, protein and gene expression profiling were done on pretreatment biopsies. EXPERIMENTAL DESIGN: Patients with high-risk, operable breast cancer were treated with three cycles of paclitaxel followed by concurrent paclitaxel/radiation. Tumor tissue from pretreatment biopsies was obtained from 19 of the 38 patients enrolled in the study. Protein and gene expression profiling were done on serial sections of the biopsies from patients that achieved a pathologic complete response (pCR) and compared to those with residual disease, non-pCR (NR). RESULTS: Proteomic and validation immunohistochemical analyses revealed that alpha-defensins (DEFA) were overexpressed in tumors from patients with a pCR. Gene expression analysis revealed that MAP2, a microtubule-associated protein, had significantly higher levels of expression in patients achieving a pCR. Elevation of MAP2 in breast cancer cell lines led to increased paclitaxel sensitivity. Furthermore, expression of genes that are associated with the basal-like, triple-negative phenotype were enriched in tumors from patients with a pCR. Analysis of a larger panel of tumors from patients receiving presurgical taxane-based treatment showed that DEFA and MAP2 expression as well as histologic features of inflammation were all statistically associated with response to therapy at the time of surgery. CONCLUSION: We show the utility of molecular profiling of pretreatment biopsies to discover markers of response. Our results suggest the potential use of immune signaling molecules such as DEFA as well as MAP2, a microtubule-associated protein, as tumor markers that associate with response to neoadjuvant taxane-based therapy.


Asunto(s)
Biomarcadores Farmacológicos/análisis , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Carcinoma/genética , Carcinoma/metabolismo , Paclitaxel/uso terapéutico , Taxoides/uso terapéutico , Biomarcadores Farmacológicos/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/radioterapia , Carcinoma/tratamiento farmacológico , Carcinoma/radioterapia , Terapia Combinada , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genómica , Humanos , Metaboloma/efectos de los fármacos , Terapia Neoadyuvante , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteómica , Taxoides/farmacología , Células Tumorales Cultivadas
17.
Cell Cycle ; 8(22): 3702-6, 2009 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19855172

RESUMEN

The tumor suppressor p53 is commonly mutated in human cancers. However, two homologs of p53, p63 and p73, are frequently overexpressed in tumors and are associated with tumor subtypes, clinical outcomes, and responses to therapy. There are many isoforms of p53, p63 and p73 (the p53 family). Proper detection of and discrimination between the members of this tumor suppressor family in human tissues is of critical importance to cancer research and clinical care. In this study, we assessed the specificity of several commercially available and newly generated p73 antibodies, focusing on antibodies that distinguish between the TA p73 and DeltaNp73 isoforms by western analysis, immunohistochemistry, and immunofluorescence. In addition, we found that the pan-p63 and pan-p73 antibodies tested cross-react with p73 and p63 respectively. The results of this study have important implications for analysis of p63 and p73 expression and co-expression in human tumors, and for potential use of these reagents in molecular diagnostics and therapeutic decision-making.


Asunto(s)
Anticuerpos/metabolismo , Especificidad de Anticuerpos/inmunología , Reacciones Cruzadas/inmunología , Proteínas de Unión al ADN/metabolismo , Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Transactivadores/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Western Blotting , Línea Celular Tumoral , Proteínas de Unión al ADN/inmunología , Técnica del Anticuerpo Fluorescente , Humanos , Inmunohistoquímica , Proteínas Nucleares/inmunología , Transactivadores/inmunología , Factores de Transcripción , Proteína Tumoral p73 , Proteínas Supresoras de Tumor/inmunología
18.
Autophagy ; 5(1): 114-6, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19001857

RESUMEN

The p53 family consists of three transcription factors, p53, p63 and p73 that share domain architecture and sequence identity. The mTOR (mammalian target of rapamycin) kinase responds to growth factors and nutrient levels to regulate cellular growth and autophagy. Whereas p53 acts both upstream and downstream of mTOR, gene signature-based analyses have revealed that p73 is inhibited by mTOR activity. p53 can both activate and repress autophagy levels depending on cellular context. While less is known about p73, recent studies have shown that it induces cellular autophagy and multiple autophagy-associated genes downstream of mTOR. Chromatin immunoprecipitation analyses demonstrate that endogenous p73 binds the regulatory regions of genes such as ATG5, ATG7 and UVRAG. How p73 regulates the expression levels of these genes in response to different cellular stresses remains unknown. Because p53 family members play key roles in tumor suppression, development, aging and neurodegeneration, the context and manner by which these transcription factors regulate autophagy may have implications for a wide range of human diseases.


Asunto(s)
Autofagia/genética , Regulación de la Expresión Génica , Proteínas Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Secuencia de Bases , Humanos , Ratones , Datos de Secuencia Molecular , Serina-Treonina Quinasas TOR
19.
Genes Dev ; 22(19): 2591-5, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18832062

RESUMEN

While p53 has been extensively characterized as a tumor suppressor, it has been more difficult to determine whether p63 and/or p73 play a similar role. Every system in which these family members have been studied, from cells to animal models to human tissues, seems to create more questions than answers. Tomasini and colleagues (2677-2691) demonstrate that one isoform of p73 is responsible for preventing tumor formation in vivo, providing critical validation of an isoform-based model of p73 function.


Asunto(s)
Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Genes Supresores de Tumor , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/fisiología , Animales , Proteínas de Unión al ADN/deficiencia , Femenino , Inestabilidad Genómica , Humanos , Infertilidad/genética , Masculino , Ratones , Ratones Noqueados , Modelos Biológicos , Mutación , Neoplasias/etiología , Neoplasias/genética , Neoplasias/prevención & control , Proteínas Nucleares/deficiencia , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología , Transducción de Señal , Proteína Tumoral p73 , Proteínas Supresoras de Tumor/deficiencia
20.
Mol Cell Biol ; 28(19): 5951-64, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18678646

RESUMEN

Although genomic technologies have advanced the characterization of gene regulatory networks downstream of transcription factors, the identification of pathways upstream of these transcription factors has been more challenging. In this study we present a gene signature-based approach for connecting signaling pathways to transcription factors, as exemplified by p73. We generated a p73 gene signature by integrating whole-genome chromatin immunoprecipitation and expression profiling. The p73 signature was linked to corresponding signatures produced by drug candidates, using the in silico Connectivity Map resource, to identify drugs that would induce p73 activity. Of the pharmaceutical agents identified, there was enrichment for direct or indirect inhibitors of mammalian Target of Rapamycin (mTOR) signaling. Treatment of both primary cells and cancer cell lines with rapamycin, metformin, and pyrvinium resulted in an increase in p73 levels, as did RNA interference-mediated knockdown of mTOR. Further, a subset of genes associated with insulin response or autophagy exhibited mTOR-mediated, p73-dependent expression. Thus, downstream gene signatures can be used to identify upstream regulators of transcription factor activity, and in doing so, we identified a new link between mTOR, p73, and p73-regulated genes associated with autophagy and metabolic pathways.


Asunto(s)
Proteínas de Unión al ADN/genética , Genómica/métodos , Proteínas Nucleares/genética , Proteínas Quinasas/metabolismo , Transducción de Señal , Proteínas Supresoras de Tumor/genética , Línea Celular , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/metabolismo , Humanos , Proteínas Nucleares/metabolismo , Serina-Treonina Quinasas TOR , Proteína Tumoral p73 , Proteínas Supresoras de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA