Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Cell Sci ; 125(Pt 18): 4253-63, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22718346

RESUMEN

The Forkhead transcription factor, FoxO3a, is a known suppressor of primary tumor growth through transcriptional regulation of key genes regulating cell cycle arrest and apoptosis. In many types of cancer, in response to growth factor signaling, FoxO3a is phosphorylated by Akt, resulting in its exclusion from the nucleus. Here we show that FoxO3a remains nuclear in anaplastic thyroid carcinoma (ATC). This correlates with lack of Akt phosphorylation at serine473 in ATC cell lines and tissues of ATC patients, providing a potential explanation for nuclear FoxO3a. Mechanistically, nuclear FoxO3a promotes cell cycle progression by transcriptional upregulation of cyclin A1, promoting proliferation of human ATC cells. Silencing FoxO3a with a reverse genetics approach leads to downregulation of CCNA1 mRNA and protein. These combined data suggest an entirely novel function for FoxO3a in ATC promotion by enhancing cell cycle progression and tumor growth through transcriptional upregulation of cyclin A1. This is clinically relevant since we detected highly elevated CCNA1 mRNA and protein levels in tumor tissues of ATC patients. Our data indicate therapeutic inactivation of FoxO3a may lead to attenuation of tumor expansion in ATC. This new paradigm also suggests caution in relation to current dogma focused upon reactivation of FoxO3a as a therapeutic strategy against cancers harboring active PI3-K and Akt signaling pathways.


Asunto(s)
Ciclina A1/genética , Factores de Transcripción Forkhead/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología , Transcripción Genética , Secuencia de Bases , Línea Celular Tumoral , Núcleo Celular/metabolismo , Proliferación Celular , Ciclina A1/metabolismo , Proteína Forkhead Box O3 , Silenciador del Gen , Células HEK293 , Humanos , Datos de Secuencia Molecular , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Carcinoma Anaplásico de Tiroides , Neoplasias de la Tiroides/enzimología , Neoplasias de la Tiroides/terapia
2.
Nat Commun ; 15(1): 5871, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-38997283

RESUMEN

There are numerous mechanisms by which glioblastoma cells evade immunological detection, underscoring the need for strategic combinatorial treatments to achieve appreciable therapeutic effects. However, developing combination therapies is difficult due to dose-limiting toxicities, blood-brain-barrier, and suppressive tumor microenvironment. Glioblastoma is notoriously devoid of lymphocytes driven in part by a paucity of lymphocyte trafficking factors necessary to prompt their recruitment and activation. Herein, we develop a recombinant adeno-associated virus (AAV) gene therapy that enables focal and stable reconstitution of the tumor microenvironment with C-X-C motif ligand 9 (CXCL9), a powerful call-and-receive chemokine for lymphocytes. By manipulating local chemokine directional guidance, AAV-CXCL9 increases tumor infiltration by cytotoxic lymphocytes, sensitizing glioblastoma to anti-PD-1 immune checkpoint blockade in female preclinical tumor models. These effects are accompanied by immunologic signatures evocative of an inflamed tumor microenvironment. These findings support AAV gene therapy as an adjuvant for reconditioning glioblastoma immunogenicity given its safety profile, tropism, modularity, and off-the-shelf capability.


Asunto(s)
Quimiocina CXCL9 , Dependovirus , Terapia Genética , Glioblastoma , Inhibidores de Puntos de Control Inmunológico , Receptor de Muerte Celular Programada 1 , Microambiente Tumoral , Glioblastoma/terapia , Glioblastoma/inmunología , Dependovirus/genética , Microambiente Tumoral/inmunología , Animales , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Quimiocina CXCL9/genética , Quimiocina CXCL9/inmunología , Ratones , Terapia Genética/métodos , Femenino , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Línea Celular Tumoral , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/terapia , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética
3.
Clin Cancer Res ; 29(9): 1751-1762, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36749885

RESUMEN

PURPOSE: An ongoing challenge in cancer is the management of primary and metastatic brain malignancies. This is partly due to restrictions of the blood-brain barrier and their unique microenvironment. These challenges are most evident in cancers such as lymphoma and melanoma, which are typically responsive to treatment in systemic locations but resistant when established in the brain. We propose interleukin-1 receptor-associated kinase-4 (IRAK-4) as a potential target across these diseases and describe the activity and mechanism of oral IRAK-4 inhibitor CA-4948. EXPERIMENTAL DESIGN: Human primary central nervous system lymphoma (PCNSL) and melanoma brain metastases (MBM) samples were analyzed for expression of IRAK-4 and downstream transcription pathways. We next determined the central nervous system (CNS) applicability of CA-4948 in naïve and tumor-bearing mice using models of PCNSL and MBM. The mechanistic effect on tumors and the tumor microenvironment was then analyzed. RESULTS: Human PCNSL and MBM have high expression of IRAK-4, IRAK-1, and nuclear factor kappa B (NF-κB). This increase in inflammation results in reflexive inhibitory signaling. Similar profiles are observed in immunocompetent murine models. Treatment of tumor-bearing animals with CA-4948 results in the downregulation of mitogen-activated protein kinase (MAPK) signaling in addition to decreased NF-κB. These intracellular changes are associated with a survival advantage. CONCLUSIONS: IRAK-4 is an attractive target in PCNSL and MBM. The inhibition of IRAK-4 with CA-4948 downregulates the expression of important transcription factors involved in tumor growth and proliferation. CA-4948 is currently being investigated in clinical trials for relapsed and refractory lymphoma and warrants further translation into PCNSL and MBM.


Asunto(s)
Neoplasias Encefálicas , Melanoma , Animales , Humanos , Ratones , Barrera Hematoencefálica/metabolismo , Neoplasias Encefálicas/tratamiento farmacológico , Factores Inmunológicos , Melanoma/tratamiento farmacológico , Melanoma/genética , FN-kappa B/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal , Microambiente Tumoral
4.
Nat Commun ; 11(1): 1508, 2020 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-32198351

RESUMEN

Tumour cell phagocytosis by antigen presenting cells (APCs) is critical to the generation of antitumour immunity. However, cancer cells can evade phagocytosis by upregulating anti-phagocytosis molecule CD47. Here, we show that CD47 blockade alone is inefficient in stimulating glioma cell phagocytosis. However, combining CD47 blockade with temozolomide results in a significant pro-phagocytosis effect due to the latter's ability to induce endoplasmic reticulum stress response. Increased tumour cell phagocytosis subsequently enhances antigen cross-presentation and activation of cyclic GMP-AMP synthase-stimulator of interferon genes (cGAS-STING) in APCs, resulting in more efficient T cell priming. This bridging of innate and adaptive responses inhibits glioma growth, but also activates immune checkpoint. Sequential administration of an anti-PD1 antibody overcomes this potential adaptive resistance. Together, these findings reveal a dynamic relationship between innate and adaptive immune regulation in tumours and support further investigation of phagocytosis modulation as a strategy to enhance cancer immunotherapy responses.


Asunto(s)
Inmunidad Adaptativa , Glioblastoma/inmunología , Glioma/inmunología , Inmunidad Innata , Fagocitosis/inmunología , Animales , Presentación de Antígeno , Apoptosis , Antígeno CD47/efectos de los fármacos , Antígeno CD47/metabolismo , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Retículo Endoplásmico/metabolismo , Glioblastoma/patología , Humanos , Inmunoterapia/métodos , Ratones , Ratones Endogámicos C57BL , Monitorización Inmunológica , Nucleotidiltransferasas/metabolismo , Linfocitos T/inmunología , Temozolomida/farmacología
5.
Cancer Res ; 79(7): 1535-1548, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30696657

RESUMEN

During development of pancreatic cancer, alternatively activated macrophages contribute to fibrogenesis, pancreatic intraepithelial neoplasia (PanIN) lesion growth, and generation of an immunosuppressive environment. Here, we show that the immunomodulatory agent pomalidomide depletes pancreatic lesion areas of alternatively activated macrophage populations. Pomalidomide treatment resulted in downregulation of interferon regulatory factor 4, a transcription factor for M2 macrophage polarization. Pomalidomide-induced absence of alternatively activated macrophages led to a decrease in fibrosis at PanIN lesions and in syngeneic tumors; this was due to generation of an inflammatory, immune-responsive environment with increased expression of IL1α and presence of activated (IFNγ-positive) CD4+ and CD8+ T-cell populations. Our results indicate that pomalidomide could be used to decrease fibrogenesis in pancreatic cancer and may be ideal as a combination treatment with chemotherapeutic drugs or other immunotherapies. SIGNIFICANCE: These findings reveal new insights into how macrophage populations within the pancreatic cancer microenvironment can be modulated, providing the means to turn the microenvironment from immunosuppressive to immune-responsive.


Asunto(s)
Factores Inmunológicos/farmacología , Macrófagos/inmunología , Neoplasias Pancreáticas/inmunología , Lesiones Precancerosas/inmunología , Talidomida/análogos & derivados , Animales , Humanos , Factores Reguladores del Interferón/metabolismo , Ratones , Neoplasias Pancreáticas/metabolismo , Lesiones Precancerosas/metabolismo , Talidomida/farmacología , Microambiente Tumoral , Células U937
6.
Adv Sci (Weinh) ; 6(5): 1802070, 2019 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-30886813

RESUMEN

A major challenge in the development of cancer nanomedicine is the inability for nanomaterials to efficiently penetrate and deliver therapeutic agents into solid tumors. Previous studies have shown that tumor vasculature and extracellular matrix regulate the transvascular and interstitial transport of nanoparticles, both critical for successfully delivering nanomedicine into solid tumors. Within the malignant tumor microenvironment, blood vessels are morphologically abnormal and functionally exhibit substantial permeability. Furthermore, the tumor extracellular matrix (ECM), unlike that of the normal tissue parenchyma, is densely packed with collagen. These pathophysiological properties greatly impede intratumoral delivery of nanomaterials. By using an antivascular endothelial growth factor receptor antibody, DC101, and an antitransforming growth factor ß1 (TGF-ß1) antibody, normalization of the tumor vasculature and ECM is achieved, respectively, in a syngeneic murine glioma model. This normalization effect results in a more organized vascular network, improves tissue perfusion, and reduces collagen density, all of which contribute to enhanced nanoparticle delivery and distribution within tumors. These findings suggest that combined vascular and ECM normalization strategies can be used to remodel the tumor microenvironment and improve nanomedicine delivery into solid tumors, which has significant implications for developing more effective combinational therapeutic strategies using cancer nanomedicine.

7.
Oncotarget ; 9(1): 3-20, 2018 Jan 02.
Artículo en Inglés | MEDLINE | ID: mdl-29416592

RESUMEN

Here we present an innovative computational-based drug discovery strategy, coupled with machine-based learning and functional assessment, for the rational design of novel small molecule inhibitors of the lipogenic enzyme stearoyl-CoA desaturase 1 (SCD1). Our methods resulted in the discovery of several unique molecules, of which our lead compound SSI-4 demonstrates potent anti-tumor activity, with an excellent pharmacokinetic and toxicology profile. We improve upon key characteristics, including chemoinformatics and absorption/distribution/metabolism/excretion (ADME) toxicity, while driving the IC50 to 0.6 nM in some instances. This approach to drug design can be executed in smaller research settings, applied to a wealth of other targets, and paves a path forward for bringing small-batch based drug programs into the Clinic.

8.
Nat Rev Drug Discov ; 16(6): 369-370, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28303024

RESUMEN

Despite a decade of intensive preclinical research, the translation of cancer nanomedicine to the clinic has been slow. Here, we discuss how recent lessons learned from the successes with immuno-oncology therapies could be applied to cancer nanomedicine and how this may help to overcome some of the key technical challenges in this field.


Asunto(s)
Inmunoterapia/métodos , Nanomedicina/métodos , Neoplasias/terapia , Animales , Diseño de Fármacos , Humanos , Neoplasias/inmunología , Investigación Biomédica Traslacional/métodos
9.
Nat Nanotechnol ; 12(8): 763-769, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28459470

RESUMEN

Tumour-targeted immunotherapy offers the unique advantage of specific tumouricidal effects with reduced immune-associated toxicity. However, existing platforms suffer from low potency, inability to generate long-term immune memory and decreased activities against tumour-cell subpopulations with low targeting receptor levels. Here we adopted a modular design approach that uses colloidal nanoparticles as substrates to create a multivalent bi-specific nanobioconjugate engager (mBiNE) to promote selective, immune-mediated eradication of cancer cells. By simultaneously targeting the human epidermal growth factor receptor 2 (HER2) expressed by cancer cells and pro-phagocytosis signalling mediated by calreticulin, the mBiNE stimulated HER2-targeted phagocytosis and produced durable antitumour immune responses against HER2-expressing tumours. Interestingly, although the initial immune activation mediated by the mBiNE was receptor dependent, the subsequent antitumour immunity also generated protective effects against tumour-cell populations that lacked the HER2 receptor. Thus, the mBiNE represents a new targeted, nanomaterial-immunotherapy platform to stimulate innate and adaptive immunity and promote a universal antitumour response.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Inmunoterapia/métodos , Nanoconjugados/química , Neoplasias/terapia , Receptor ErbB-2/inmunología , Inmunidad Adaptativa , Animales , Coloides , Humanos , Inmunidad Innata , Ratones Endogámicos BALB C , Neoplasias/inmunología , Neoplasias/patología , Células THP-1
10.
Expert Opin Ther Targets ; 20(2): 159-66, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26414044

RESUMEN

INTRODUCTION: Anaplastic thyroid carcinoma (ATC) is the rarest subtype of thyroid cancer; however, it disproportionately accounts for a large percentage of all thyroid cancer-related deaths and is considered one of the most lethal solid tumors in humans, having a median survival of only a few months upon diagnosis. Although a variety of treatment options are available including surgery, radiation and targeted therapies, response rates are low, due in part to the drug-resistant nature of this disease; therefore, new avenues for therapeutic intervention are surely needed. Recent investigation into the metabolic profile of ATC has revealed a tumor-specific dependency for increased de novo lipogenesis, offering new insight into the molecular mechanisms that govern disease initiation and progression. AREAS COVERED: Herein we summarize known oncogenic signaling pathways and current therapeutic strategies for the treatment of ATC. We further discuss the unique expression pattern of lipid metabolism constituents in this disease. Additionally, the current literature correlating aberrant lipogenesis with carcinogenesis is reviewed, and the implications of targeting this pathway as an innovative approach for treating ATC and other malignancies are discussed. As stearoyl-CoA desaturase (SCD) is the most differentially expressed constituent of lipid metabolism in ATC, an additional focus on this enzyme as a novel therapeutic target is applied. EXPERT OPINION: This section is used to summarize the current research efforts underway in defining the role of lipid metabolism specifically in thyroid carcinoma. Included is a brief summary of lipid metabolism factors for which inhibitors have been generated and are under current investigation as anti-cancer agents. Finally, research limitations regarding the use of these inhibitors against components of this pathway are discussed.


Asunto(s)
Metabolismo de los Lípidos/fisiología , Carcinoma Anaplásico de Tiroides/tratamiento farmacológico , Neoplasias de la Tiroides/tratamiento farmacológico , Antineoplásicos/farmacología , Progresión de la Enfermedad , Resistencia a Antineoplásicos , Humanos , Terapia Molecular Dirigida , Carcinoma Anaplásico de Tiroides/patología , Neoplasias de la Tiroides/patología
11.
Sci Rep ; 6: 26269, 2016 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-27197045

RESUMEN

Nanomedicine is a burgeoning industry but an understanding of the interaction of nanomaterials with the immune system is critical for clinical translation. Macrophages play a fundamental role in the immune system by engulfing foreign particulates such as nanoparticles. When activated, macrophages form distinct phenotypic populations with unique immune functions, however the mechanism by which these polarized macrophages react to nanoparticles is unclear. Furthermore, strategies to selectively evade activated macrophage subpopulations are lacking. Here we demonstrate that stimulated macrophages possess higher phagocytic activities and that classically activated (M1) macrophages exhibit greater phagocytic capacity than alternatively activated (M2) macrophages. We show that modification of nanoparticles with polyethylene-glycol results in decreased clearance by all macrophage phenotypes, but importantly, coating nanoparticles with CD47 preferentially lowers phagocytic activity by the M1 phenotype. These results suggest that bio-inspired nanoparticle surface design may enable evasion of specific components of the immune system and provide a rational approach for developing immune tolerant nanomedicines.


Asunto(s)
Macrófagos/fisiología , Nanopartículas/química , Animales , Antígeno CD47/química , Activación de Macrófagos , Macrófagos/inmunología , Ratones Endogámicos C57BL , Fagocitosis , Fenotipo , Polietilenglicoles/química , Poliestirenos
12.
Oncotarget ; 7(22): 32156-71, 2016 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-27050077

RESUMEN

Osteopontin (OPN) is the most upregulated gene in primary central nervous system lymphoma (PCNSL) compared to non-CNS diffuse large B cell lymphoma (DLBCL). We show here that OPN is a key mediator of intracerebral tumor growth, invasion, and dissemination in CNS lymphoma, and that these effects depend upon activation of NF-κB. We further show that activation of NF-κB by OPN occurs through a unique mechanism in which intracellular OPN (iOPN) causes transcriptional downregulation of the NF-κB inhibitors, A20/TNFAIP3 and ABIN1/TNIP1, and secretory OPN (sOPN) promotes receptor-mediated activation of NF-κB. We also identify NF-κB-mediated induction of matrix metalloproteinase-8 (MMP-8) as a specific feature of OPN-mediated tissue invasion. These results implicate OPN as a candidate for development of targeted therapy for patients with PCNSL.


Asunto(s)
Movimiento Celular , Proliferación Celular , Neoplasias del Sistema Nervioso Central/metabolismo , Linfoma de Células B/metabolismo , Osteopontina/metabolismo , Animales , Comunicación Autocrina , Línea Celular Tumoral , Neoplasias del Sistema Nervioso Central/genética , Neoplasias del Sistema Nervioso Central/patología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Linfoma de Células B/genética , Linfoma de Células B/patología , Metaloproteinasa 8 de la Matriz/genética , Metaloproteinasa 8 de la Matriz/metabolismo , Ratones Desnudos , FN-kappa B/genética , FN-kappa B/metabolismo , Invasividad Neoplásica , Metástasis de la Neoplasia , Osteopontina/genética , Interferencia de ARN , Transducción de Señal , Factores de Tiempo , Transcripción Genética , Transfección , Carga Tumoral , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo
13.
J Clin Endocrinol Metab ; 100(5): E697-709, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25675381

RESUMEN

CONTEXT: Currently there are no efficacious therapies for patients with anaplastic thyroid carcinoma (ATC) that result in long-term disease stabilization or regression. OBJECTIVE: We sought to identify pathways critical for ATC cell progression and viability in an effort to develop new therapeutic strategies. We investigated the effects of targeted inhibition of stearoyl-CoA desaturase 1 (SCD1), a constituent of fatty acid metabolism overexpressed in ATC. DESIGN: A gene array of ATC and normal thyroid tissue was performed to identify gene transcripts demonstrating altered expression in tumor samples. Effects of pharmacological and the genetic inhibition of SCD1 on tumor cell viability as well as cell signaling responses to therapy were evaluated in in vitro and in vivo models of this rare, lethal malignancy. RESULTS: The gene array analysis revealed consistent distortion of fatty acid metabolism and overexpression of SCD1 in ATC and well-differentiated thyroid carcinomas. SCD1 is critical for ATC cell survival and proliferation, the inhibition of which induced endoplasmic reticulum stress, activation of the unfolded protein response, and apoptosis. Combined suppression of endoplasmic reticulum-associated degradation, a prosurvival component of the unfolded protein response, using proteasome inhibitors resulted in a synergistic decrease in tumor cell proliferation and increased cell death. CONCLUSIONS: SCD1 is a novel oncogenic factor specifically required for tumor cell viability in ATC. Furthermore, the expression of SCD1 appears to be correlated with thyroid tumor aggressiveness and may serve as a prognostic biomarker. These findings substantiate SCD1 as a novel tumor-specific target for therapy in patients with ATC and should be further investigated in a clinical setting.


Asunto(s)
Metabolismo de los Lípidos/genética , Estearoil-CoA Desaturasa/metabolismo , Carcinoma Anaplásico de Tiroides/metabolismo , Neoplasias de la Tiroides/metabolismo , Apoptosis/genética , Línea Celular Tumoral , Proliferación Celular/genética , Supervivencia Celular/genética , Humanos , Estearoil-CoA Desaturasa/genética , Carcinoma Anaplásico de Tiroides/genética , Carcinoma Anaplásico de Tiroides/patología , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología , Análisis de Matrices Tisulares
14.
Cancer Res ; 74(17): 4796-810, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24962026

RESUMEN

Clear cell renal cell carcinoma (ccRCC) is the most common subtype of kidney cancer and has the highest propensity to manifest as metastatic disease. Recent characterizations of the genetic signature of ccRCC have revealed several factors correlated with tumor cell migration and invasion; however, the specific events driving malignancy are not well defined. Furthermore, there remains a lack of targeted therapies that result in long-term, sustainable response in patients with metastatic disease. We show here that neuronal pentraxin 2 (NPTX2) is overexpressed specifically in ccRCC primary tumors and metastases, and that it contributes to tumor cell viability and promotes cell migration through its interaction with the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor subunit GluR4. We propose NPTX2 as a novel molecular target for therapy for patients with ccRCC diagnosed with or at risk of developing metastatic disease.


Asunto(s)
Proteína C-Reactiva/genética , Carcinoma de Células Renales/genética , Neoplasias Renales/genética , Proteínas del Tejido Nervioso/genética , Receptores AMPA/genética , Proteína C-Reactiva/metabolismo , Carcinoma de Células Renales/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Supervivencia Celular/genética , Humanos , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/patología , Proteínas del Tejido Nervioso/metabolismo , Receptores AMPA/metabolismo , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiónico/metabolismo
15.
Oncotarget ; 5(14): 5320-34, 2014 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-24979721

RESUMEN

Currently there is a lack of targeted therapies that lead to long-term attenuation or regression of disease in patients with advanced clear cell renal cell carcinoma (ccRCC). Our group has implemented a high-throughput genetic analysis coupled with a high-throughput proliferative screen in order to investigate the genetic contributions of a large cohort of overexpressed genes at the functional level in an effort to better understand factors involved in tumor initiation and progression. Patient gene array analysis identified transcripts that are consistently elevated in patient ccRCC as compared to matched normal renal tissues. This was followed by a high-throughput lentivirus screen, independently targeting 195 overexpressed transcripts identified in the gene array in four ccRCC cell lines. This revealed 31 'hits' that contribute to ccRCC cell proliferation. Many of the hits identified are not only presented in the context of ccRCC for the first time, but several have not been previously linked to cancer. We further characterize the function of a group of hits in tumor cell invasion. Taken together these findings reveal pathways that may be critical in ccRCC tumorigenicity, and identifies novel candidate factors that could serve as targets for therapeutic intervention or diagnostic/prognostic biomarkers for patients with advanced ccRCC.


Asunto(s)
Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Movimiento Celular/genética , Genómica/métodos , Neoplasias Renales/genética , Neoplasias Renales/patología , Línea Celular Tumoral , Proliferación Celular/genética , Regulación hacia Abajo , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos
16.
Am J Cancer Res ; 3(4): 390-401, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23977448

RESUMEN

Historically, metastatic renal cell carcinoma (mRCC) is more resistant to conventional cytotoxic chemotherapeutic agents than other solid tumors. Although significant progress has been made over the last decade with several novel therapeutics, these agents invariably go on to fail, largely due to either intrinsic or acquired resistance. To help overcome, or at least delay resistance, combinatorial therapies utilizing agents with disparate, and ideally complementary, mechanisms of actions are needed. In this report, we assess the novel combination of the mTOR inhibitor, temsirolimus, with the microtubule stabilizing drug ixabepilone in RCC. Our results demonstrate synergy in multiple cell lines of RCC and further evaluation of this combination is warranted in the clinical setting. Activation of the endoplasmic reticulum (ER) stress response pathway may in part explain the combinatorial synergy. We further propose that ER stress induced proteins may serve as early response biomarkers to combinatorial therapy in a clinical trial.

17.
Clin Cancer Res ; 19(9): 2368-80, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23633458

RESUMEN

PURPOSE: We set out to identify Stearoyl-CoA desaturase 1 (SCD1) as a novel molecular target in clear cell renal cell carcinoma (ccRCC) and examine its role in tumor cell growth and viability in vitro and in vivo independently as well as in combination with current U.S. Food and Drug Administration (FDA)-approved regimens. EXPERIMENTAL DESIGN: Patient normal and ccRCC tissue samples and cell lines were examined for SCD1 expression. Genetic knockdown models and targeted inhibition of SCD1 through use of a small molecule inhibitor, A939572, were analyzed for growth, apoptosis, and alterations in gene expression using gene array analysis. Therapeutic models of synergy were evaluated utilizing pharmacologic inhibition of SCD1 with the tyrosine kinase inhibitors (TKI) sunitinib and pazopanib, and the mTOR inhibitor temsirolimus. RESULTS: Our studies identify increased SCD1 expression in all stages of ccRCC. Both genetic knockdown and pharmacologic inhibition of SCD1 decreased tumor cell proliferation and induced apoptosis in vitro and in vivo. Upon gene array, quantitative real-time PCR, and protein analysis of A939572-treated or SCD1 lentiviral knockdown samples, induction of endoplasmic reticulum stress response signaling was observed, providing mechanistic insight for SCD1 activity in ccRCC. Furthermore, combinatorial application of A939572 with temsirolimus synergistically inhibited tumor growth in vitro and in vivo. CONCLUSIONS: Increased SCD1 expression supports ccRCC viability and therefore we propose it as a novel molecular target for therapy either independently or in combination with an mTOR inhibitor for patients whose disease cannot be remedied with surgical intervention, such as in cases of advanced or metastatic disease.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Carcinoma de Células Renales/tratamiento farmacológico , Neoplasias Renales/tratamiento farmacológico , Estearoil-CoA Desaturasa/metabolismo , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Células Renales/enzimología , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Sinergismo Farmacológico , Femenino , Expresión Génica , Humanos , Indazoles , Indoles/administración & dosificación , Neoplasias Renales/enzimología , Neoplasias Renales/patología , Ratones , Ratones Desnudos , Terapia Molecular Dirigida , Pirimidinas/administración & dosificación , Pirroles/administración & dosificación , Sirolimus/administración & dosificación , Sirolimus/análogos & derivados , Estearoil-CoA Desaturasa/antagonistas & inhibidores , Estearoil-CoA Desaturasa/genética , Sulfonamidas/administración & dosificación , Sunitinib , Carga Tumoral/efectos de los fármacos , Regulación hacia Arriba , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Mol Cancer Ther ; 11(10): 2105-15, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22826467

RESUMEN

Metastatic solid tumors are aggressive and mostly drug resistant, leading to few treatment options and poor prognosis as seen with clear cell renal cell carcinoma (ccRCC) and triple-negative breast cancer (TNBC). Therefore, the identification of new therapeutic regimes for the treatment of metastatic disease is desirable. ccRCC and TNBC cell lines were treated with the HDAC inhibitor romidepsin and the methyltransferase inhibitor decitabine, two epigenetic modifying drugs approved by the U.S. Food and Drug Administration for the treatment of various hematologic malignancies. Cell proliferation analysis, flow cytometry, quantitative PCR, and immunoblotting techniques were used to evaluate the antitumor synergy of this drug combination and identify the reexpression of epigenetically silenced tumor suppressor genes. Combinatorial treatment of metastatic TNBC and stage IV ccRCC cell lines with romidepsin/decitabine leads to synergistic inhibition of cell growth and induction of apoptosis above levels of individual drug treatments alone. Synergistic reexpression of the tumor suppressor gene secreted frizzled-related protein one (sFRP1) was observed in combinatorial drug-treated groups. Silencing sFRP1 (short hairpin RNA) before combinatorial drug treatment showed that sFRP1 mediates the growth inhibitory and apoptotic activity of combined romidepsin/decitabine. Furthermore, addition of recombinant sFRP1 to ccRCC or TNBC cells inhibits cell growth in a dose-dependent manner through the induction of apoptosis, identifying that epigenetic silencing of sFRP1 contributes to renal and breast cancer cell survival. Combinatorial treatment with romidepsin and decitabine in drug resistant tumors is a promising treatment strategy. Moreover, recombinant sFRP1 may be a novel therapeutic strategy for cancers with suppressed sFRP1 expression.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Resistencia a Antineoplásicos/efectos de los fármacos , Glicoproteínas/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Metiltransferasas/antagonistas & inhibidores , Proteínas Supresoras de Tumor/metabolismo , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Apoptosis/genética , Azacitidina/análogos & derivados , Azacitidina/química , Azacitidina/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Decitabina , Depsipéptidos/química , Depsipéptidos/farmacología , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/genética , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glicoproteínas/genética , Inhibidores de Histona Desacetilasas/química , Humanos , Péptidos y Proteínas de Señalización Intracelular , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/genética , Neoplasias Renales/patología , Metiltransferasas/metabolismo , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Proteínas Supresoras de Tumor/genética
20.
PLoS One ; 5(5): e10696, 2010 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-20502531

RESUMEN

BACKGROUND: Clear cell renal cell carcinoma (ccRCC) is the most common kidney cancer. The purpose of this study is to define a biological pathway signature and a cellular differentiation program in ccRCC. METHODOLOGY: We performed gene expression profiling of early-stage ccRCC and patient-matched normal renal tissue using Affymetrix HG-U133a and HG-U133b GeneChips combined with a comprehensive bioinformatic analyses, including pathway analysis. The results were validated by real time PCR and IHC on two independent sample sets. Cellular differentiation experiments were performed on ccRCC cell lines and their matched normal renal epithelial cells, in differentiation media, to determine their mesenchymal differentiation potential. PRINCIPAL FINDINGS: We identified a unique pathway signature with three major biological alterations-loss of normal renal function, down-regulated metabolism, and immune activation-which revealed an adipogenic gene expression signature linked to the hallmark lipid-laden clear cell morphology of ccRCC. Culturing normal renal and ccRCC cells in differentiation media showed that only ccRCC cells were induced to undergo adipogenic and, surprisingly, osteogenic differentiation. A gene expression signature consistent with epithelial mesenchymal transition (EMT) was identified for ccRCC. We revealed significant down-regulation of four developmental transcription factors (GATA3, TFCP2L1, TFAP2B, DMRT2) that are important for normal renal development. CONCLUSIONS: ccRCC is characterized by a lack of epithelial differentiation, mesenchymal/adipogenic transdifferentiation, and pluripotent mesenchymal stem cell-like differentiation capacity in vitro. We suggest that down-regulation of developmental transcription factors may mediate the aberrant differentiation in ccRCC. We propose a model in which normal renal epithelial cells undergo dedifferentiation, EMT, and adipogenic transdifferentiation, resulting in ccRCC. Because ccRCC cells grown in adipogenic media regain the characteristic ccRCC phenotype, we have identified a new in vitro ccRCC cell model more resembling ccRCC tumor morphology.


Asunto(s)
Carcinoma de Células Renales/genética , Diferenciación Celular/genética , Neoplasias Renales/genética , Transducción de Señal/genética , Adipogénesis/genética , Biomarcadores de Tumor/metabolismo , Carcinoma de Células Renales/patología , Transdiferenciación Celular/genética , Epitelio/metabolismo , Epitelio/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Riñón/metabolismo , Riñón/patología , Neoplasias Renales/patología , Mesodermo/metabolismo , Mesodermo/patología , Modelos Biológicos , Osteogénesis/genética , Reproducibilidad de los Resultados , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA