Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Molecules ; 26(23)2021 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-34885848

RESUMEN

Phospholipase A2 (PLA2) enzymes were first recognized as an enzyme activity class in 1961. The secreted (sPLA2) enzymes were the first of the five major classes of human PLA2s to be identified and now number nine catalytically-active structurally homologous proteins. The best-studied of these, group IIA sPLA2, has a clear role in the physiological response to infection and minor injury and acts as an amplifier of pathological inflammation. The enzyme has been a target for anti-inflammatory drug development in multiple disorders where chronic inflammation is a driver of pathology since its cloning in 1989. Despite intensive effort, no clinically approved medicines targeting the enzyme activity have yet been developed. This review catalogues the major discoveries in the human group IIA sPLA2 field, focusing on features of enzyme function that may explain this lack of success and discusses future research that may assist in realizing the potential benefit of targeting this enzyme. Functionally-selective inhibitors together with isoform-selective inhibitors are necessary to limit the apparent toxicity of previous drugs. There is also a need to define the relevance of the catalytic function of hGIIA to human inflammatory pathology relative to its recently-discovered catalysis-independent function.


Asunto(s)
Fosfolipasas A2 Grupo II/metabolismo , Desarrollo de Medicamentos , Fosfolipasas A2 Grupo II/antagonistas & inhibidores , Fosfolipasas A2 Grupo II/farmacología , Humanos , Neoplasias/diagnóstico , Neoplasias/enzimología , Pronóstico
2.
Int J Mol Sci ; 20(5)2019 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-30866453

RESUMEN

Emerging evidence suggests that gamma-tocotrienol (γ-T3), a vitamin E isomer, has potent anti-cancer properties against a wide-range of cancers. γ-T3 not only inhibited the growth and survival of cancer cells in vitro, but also suppressed angiogenesis and tumour metastasis under in vivo conditions. Recently, γ-T3 was found to target cancer stem cells (CSCs), leading to suppression of tumour formation and chemosensitisation. Despite its promising anti-cancer potential, the exact mechanisms responsible for the effects of γ-T3 are still largely unknown. Here, we report the identification of Ang-1 (Angiopoietin-1)/Tie-2 as a novel γ-T3 downstream target. In prostate cancer cells, γ-T3 treatment leads to the suppression of Ang-1 at both the mRNA transcript and protein levels. Supplementing the cells with Ang-1 was found to protect them against the anti-CSC effect of γ-T3. Intriguingly, inactivation of Tie-2, a member receptor that mediates the effect of Ang-1, was found to significantly enhance the cytotoxic effect of γ-T3 through activation of AMP-activated protein kinase (AMPK) and subsequent interruption of autophagy. Our results highlighted the therapeutic potential of using γ-T3 in combination with a Tie-2 inhibitor to treat advanced prostate cancer.


Asunto(s)
Cromanos/farmacología , Neoplasias de la Próstata/metabolismo , Transducción de Señal/efectos de los fármacos , Vitamina E/análogos & derivados , Angiopoyetina 1/genética , Angiopoyetina 1/metabolismo , Autofagia , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Masculino , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Receptor TIE-2/genética , Receptor TIE-2/metabolismo , Vitamina E/farmacología
3.
Proteomics ; 17(23-24)2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29105980

RESUMEN

Current treatments for advanced prostate cancer focus on inhibition of the androgen receptor (AR) by androgen deprivation therapy (ADT). However, complex interactions mediated by tumor suppressors, oncogenes, aberrations of AR expression, or de novo androgen production have been shown to induce the adaptive response of prostate cancer, leading to the development of castration resistant prostate cancer. In this study, we report the effects of AR antagonist, enzalutamide on the protein contents of extracellular vesicles (EVs). EVs mediate cell-to-cell communication and increasing evidence shows the role of EVs in promoting cancer survival and metastasis. We found that treatment with enzalutamide alters the secretion of EVs, one of which is a plasma membrane calcium pump, ATP2B1/PMCA ATPase, as an AR-regulated EV protein. We highlight the networks of interactions between AR, Ca2+ , and ATP2B1, where the extracellular proteins thrombospondin-1, gelsolin, and integrinß1 were previously reported as regulators for cancer progression and metastasis, indicating the potential role of EV-derived proteins in mediating calcium homoeostasis under AR inhibition by enzalutamide. Our data further highlight the cross-talk between AR signaling and EV pathways in mediating resistance toward ADT.


Asunto(s)
Adenocarcinoma/metabolismo , Vesículas Extracelulares/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Feniltiohidantoína/análogos & derivados , Neoplasias de la Próstata/metabolismo , Receptores Androgénicos/química , Transducción de Señal/efectos de los fármacos , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/patología , Benzamidas , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Vesículas Extracelulares/efectos de los fármacos , Gelsolina/metabolismo , Humanos , Integrina beta1/metabolismo , Masculino , Nitrilos , Feniltiohidantoína/farmacología , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/patología , Trombospondina 1/metabolismo , Células Tumorales Cultivadas
4.
Prostate ; 77(14): 1416-1423, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28856701

RESUMEN

The use of circulating tumor cells (CTCs) and circulating extracellular vesicles (EVs), such as exosomes, as liquid biopsy-derived biomarkers for cancers have been investigated. CTC enumeration using the CellSearch based platform provides an accurate insight on overall survival where higher CTC counts indicate poor prognosis for patients with advanced metastatic cancer. EVs provide information based on their lipid, protein, and nucleic acid content and can be isolated from biofluids and analyzed from a relatively small volume, providing a routine and non-invasive modality to monitor disease progression. Our pilot experiment by assessing the level of two subpopulations of small EVs, the CD9 positive and CD63 positive EVs, showed that the CD9 positive EV level is higher in plasma from patients with advanced metastatic prostate cancer with detectable CTCs. These data show the potential utility of a particular EV subpopulation to serve as biomarkers for advanced metastatic prostate cancer. EVs can potentially be utilized as biomarkers to provide accurate genotypic and phenotypic information for advanced prostate cancer, where new strategies to design a more personalized therapy is currently the focus of considerable investigation.


Asunto(s)
Vesículas Extracelulares , Células Neoplásicas Circulantes , Medicina de Precisión/métodos , Neoplasias de la Próstata , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/sangre , Técnicas de Apoyo para la Decisión , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/patología , Humanos , Masculino , Estadificación de Neoplasias , Células Neoplásicas Circulantes/patología , Selección de Paciente , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/terapia
5.
Anal Biochem ; 496: 30-4, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26717899

RESUMEN

The mass spectrometry technique of multiple reaction monitoring (MRM) was used to quantify and compare the expression level of lactoferrin in tear films among control, prostate cancer (CaP), and benign prostate hyperplasia (BPH) groups. Tear samples from 14 men with CaP, 15 men with BPH, and 14 controls were analyzed in the study. Collected tears (2 µl) of each sample were digested with trypsin overnight at 37 °C without any pretreatment, and tear lactoferrin was quantified using a lactoferrin-specific peptide, VPSHAVVAR, both using natural/light and isotopic-labeled/heavy peptides with MRM. The average tear lactoferrin concentration was 1.01 ± 0.07 µg/µl in control samples, 0.96 ± 0.07 µg/µl in the BPH group, and 0.98 ± 0.07 µg/µl in the CaP group. Our study is the first to quantify tear proteins using a total of 43 individual (non-pooled) tear samples and showed that direct digestion of tear samples is suitable for MRM studies. The calculated average lactoferrin concentration in the control group matched that in the published range of human tear lactoferrin concentration measured by enzyme-linked immunosorbent assay (ELISA). Moreover, the lactoferrin was stably expressed across all of the samples, with no significant differences being observed among the control, BPH, and CaP groups.


Asunto(s)
Lactoferrina/análisis , Lágrimas/química , Anciano , Anciano de 80 o más Años , Secuencia de Aminoácidos , Calibración , Estudios de Casos y Controles , Humanos , Marcaje Isotópico , Lactoferrina/química , Límite de Detección , Masculino , Persona de Mediana Edad , Hiperplasia Prostática/metabolismo , Reproducibilidad de los Resultados
6.
Prostate ; 75(6): 628-36, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25560784

RESUMEN

BACKGROUND: Understanding the progression of prostate cancer to androgen-independence/castrate resistance and development of preclinical testing models are important for developing new prostate cancer therapies. This report describes studies performed 30 years ago, which demonstrate utility and shortfalls of xenografting to preclinical modeling. METHODS: We subcutaneously implanted male nude mice with small prostate cancer fragments from transurethral resection of the prostate (TURP) from 29 patients. Successful xenografts were passaged into new host mice. They were characterized using histology, immunohistochemistry for marker expression, flow cytometry for ploidy status, and in some cases by electron microscopy and response to testosterone. Two xenografts were karyotyped by G-banding. RESULTS: Tissues from 3/29 donors (10%) gave rise to xenografts that were successfully serially passaged in vivo. Two, (UCRU-PR-1, which subsequently was replaced by a mouse fibrosarcoma, and UCRU-PR-2, which combined epithelial and neuroendocrine features) have been described. UCRU-PR-4 line was a poorly differentiated prostatic adenocarcinoma derived from a patient who had undergone estrogen therapy and bilateral castration after his cancer relapsed. Histologically, this comprised diffusely infiltrating small acinar cell carcinoma with more solid aggregates of poorly differentiated adenocarcinoma. The xenografted line showed histology consistent with a poorly differentiated adenocarcinoma and stained positively for prostatic acid phosphatase (PAcP), epithelial membrane antigen (EMA) and the cytokeratin cocktail, CAM5.2, with weak staining for prostate specific antigen (PSA). The line failed to grow in female nude mice. Castration of three male nude mice after xenograft establishment resulted in cessation of growth in one, growth regression in another and transient growth in another, suggesting that some cells had retained androgen sensitivity. The karyotype (from passage 1) was 43-46, XY, dic(1;12)(p11;p11), der(3)t(3:?5)(q13;q13), -5, inv(7)(p15q35) x2, +add(7)(p13), add(8)(p22), add(11)(p14), add(13)(p11), add(20)(p12), -22, +r4[cp8]. CONCLUSIONS: Xenografts provide a clinically relevant model of prostate cancer, although establishing serially transplantable prostate cancer patient derived xenografts is challenging and requires rigorous characterization and high quality starting material. Xenografting from advanced prostate cancer is more likely to succeed, as xenografting from well differentiated, localized disease has not been achieved in our experience. Strong translational correlations can be demonstrated between the clinical disease state and the xenograft model.


Asunto(s)
Neoplasias de la Próstata/patología , Animales , Línea Celular Tumoral , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Tumores Neuroendocrinos/patología , Neoplasias de la Próstata/genética , Testosterona/farmacología , Trasplante Heterólogo
7.
Biomacromolecules ; 16(10): 3235-47, 2015 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-26335533

RESUMEN

Targeted nanomedicines offer a strategy for greatly enhancing accumulation of a therapeutic within a specific tissue in animals. In this study, we report on the comparative targeting efficiency toward prostate-specific membrane antigen (PSMA) of a number of different ligands that are covalently attached by the same chemistry to a polymeric nanocarrier. The targeting ligands included a small molecule (glutamate urea), a peptide ligand, and a monoclonal antibody (J591). A hyperbranched polymer (HBP) was utilized as the nanocarrier and contained a fluorophore for tracking/analysis, whereas the pendant functional chain-ends provided a handle for ligand conjugation. Targeting efficiency of each ligand was assessed in vitro using flow cytometry and confocal microscopy to compare degree of binding and internalization of the HBPs by human prostate cancer (PCa) cell lines with different PSMA expression status (PC3-PIP (PSMA+) and PC3-FLU (PSMA-). The peptide ligand was further investigated in vivo, in which BALB/c nude mice bearing subcutaneous PC3-PIP and PC3-FLU PCa tumors were injected intravenously with the HBP-peptide conjugate and assessed by fluorescence imaging. Enhanced accumulation in the tumor tissue of PC3-PIP compared to PC3-FLU highlighted the applicability of this system as a future imaging and therapeutic delivery vehicle.


Asunto(s)
Antígenos de Superficie/efectos de los fármacos , Glutamato Carboxipeptidasa II/efectos de los fármacos , Nanomedicina , Polímeros/química , Espectroscopía de Resonancia Magnética con Carbono-13 , Línea Celular Tumoral , Humanos , Ligandos , Masculino , Espectroscopía de Protones por Resonancia Magnética
8.
Cancer Metastasis Rev ; 29(1): 123-42, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20143131

RESUMEN

Despite considerable success in treatment of early stage localized prostate cancer (PC), acute inadequacy of late stage PC treatment and its inherent heterogeneity poses a formidable challenge. Clearly, an improved understanding of PC genesis and progression along with the development of new targeted therapies are warranted. Animal models, especially, transgenic immunocompetent mouse models, have proven to be the best ally in this respect. A series of models have been developed by modulation of expression of genes implicated in cancer-genesis and progression; mainly, modulation of expression of oncogenes, steroid hormone receptors, growth factors and their receptors, cell cycle and apoptosis regulators, and tumor suppressor genes have been used. Such models have contributed significantly to our understanding of the molecular and pathological aspects of PC initiation and progression. In particular, the transgenic mouse models based on multiple genetic alterations can more accurately address the inherent complexity of PC, not only in revealing the mechanisms of tumorigenesis and progression but also for clinically relevant evaluation of new therapies. Further, with advances in conditional knockout technologies, otherwise embryonically lethal gene changes can be incorporated leading to the development of new generation transgenics, thus adding significantly to our existing knowledge base. Different models and their relevance to PC research are discussed.


Asunto(s)
Adenocarcinoma/patología , Modelos Animales de Enfermedad , Ratones Transgénicos , Neoplasias de la Próstata/patología , Adenocarcinoma/genética , Adenocarcinoma/terapia , Animales , Antineoplásicos/uso terapéutico , Perros , Evaluación Preclínica de Medicamentos , Estudios de Evaluación como Asunto , Humanos , Inmunoterapia/métodos , Masculino , Ratones , Modelos Biológicos , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/terapia , Ratas
9.
Stem Cells ; 28(9): 1686-702, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20629172

RESUMEN

Ineffective treatment and poor patient management continue to plague the arena of clinical oncology. The crucial issues include inadequate treatment efficacy due to ineffective targeting of cancer deposits, systemic toxicities, suboptimal cancer detection and disease monitoring. This has led to the quest for clinically relevant, innovative multifaceted solutions such as development of targeted and traceable therapies. Mesenchymal stem cells (MSCs) have the intrinsic ability to "home" to growing tumors and are hypoimmunogenic. Therefore, these can be used as (a) "Trojan Horses" to deliver gene therapy directly into the tumors and (b) carriers of nanoparticles to allow cell tracking and simultaneous cancer detection. The camouflage of MSC carriers can potentially tackle the issues of safety, vector, and/or transgene immunogenicity as well as nanoparticle clearance and toxicity. The versatility of the nanotechnology platform could allow cellular tracking using single or multimodal imaging modalities. Toward that end, noninvasive magnetic resonance imaging (MRI) is fast becoming a clinical favorite, though there is scope for improvement in its accuracy and sensitivity. In that, use of superparamagnetic iron-oxide nanoparticles (SPION) as MRI contrast enhancers may be the best option for tracking therapeutic MSC. The prospects and consequences of synergistic approaches using MSC carriers, gene therapy, and SPION in developing cancer diagnostics and therapeutics are discussed.


Asunto(s)
Terapia Genética , Trasplante de Células Madre Mesenquimatosas , Imagen Molecular , Nanopartículas , Neoplasias/terapia , Animales , Medios de Contraste , Dextranos , Óxido Ferrosoférrico , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Humanos , Imagen por Resonancia Magnética , Nanopartículas de Magnetita , Imagen Molecular/métodos , Neoplasias/diagnóstico , Neoplasias/patología , Valor Predictivo de las Pruebas , Resultado del Tratamiento
10.
BMC Cancer ; 11: 368, 2011 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-21861932

RESUMEN

BACKGROUND: Late stage Ovarian Cancer is essentially incurable primarily due to late diagnosis and its inherent heterogeneity. Single agent treatments are inadequate and generally lead to severe side effects at therapeutic doses. It is crucial to develop clinically relevant novel combination regimens involving synergistic modalities that target a wider repertoire of cells and lead to lowered individual doses. Stemming from this premise, this is the first report of two- and three-way synergies between Adenovirus-mediated Purine Nucleoside Phosphorylase based gene directed enzyme prodrug therapy (PNP-GDEPT), docetaxel and/or carboplatin in multidrug-resistant ovarian cancer cells. METHODS: The effects of PNP-GDEPT on different cellular processes were determined using Shotgun Proteomics analyses. The in vitro cell growth inhibition in differentially treated drug resistant human ovarian cancer cell lines was established using a cell-viability assay. The extent of synergy, additivity, or antagonism between treatments was evaluated using CalcuSyn statistical analyses. The involvement of apoptosis and implicated proteins in effects of different treatments was established using flow cytometry based detection of M30 (an early marker of apoptosis), cell cycle analyses and finally western blot based analyses. RESULTS: Efficacy of the trimodal treatment was significantly greater than that achieved with bimodal- or individual treatments with potential for 10-50 fold dose reduction compared to that required for individual treatments. Of note was the marked enhancement in apoptosis that specifically accompanied the combinations that included PNP-GDEPT and accordingly correlated with a shift in the expression of anti- and pro-apoptotic proteins. PNP-GDEPT mediated enhancement of apoptosis was reinforced by cell cycle analyses. Proteomic analyses of PNP-GDEPT treated cells indicated a dowregulation of proteins involved in oncogenesis or cancer drug resistance in treated cells with accompanying upregulation of apoptotic- and tumour- suppressor proteins. CONCLUSION: Inclusion of PNP-GDEPT in regular chemotherapy regimens can lead to significant enhancement of the cancer cell susceptibility to the combined treatment. Overall, these data will underpin the development of regimens that can benefit patients with late stage ovarian cancer leading to significantly improved efficacy and increased quality of life.


Asunto(s)
Adenoviridae/genética , Antineoplásicos/farmacología , Resistencia a Antineoplásicos , Terapia Genética , Vectores Genéticos , Neoplasias Ováricas/terapia , Purina-Nucleósido Fosforilasa/genética , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Efecto Espectador , Carboplatino/farmacología , Carboplatino/uso terapéutico , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Docetaxel , Sinergismo Farmacológico , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Profármacos/metabolismo , Proteómica , Purina-Nucleósido Fosforilasa/metabolismo , Taxoides/farmacología , Taxoides/uso terapéutico , Transducción Genética
11.
Commun Biol ; 4(1): 1014, 2021 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-34462519

RESUMEN

Prostate cancer (PCa) is the second most commonly diagnosed cancer in men, and bone is the most frequent site of metastasis. The tumor microenvironment (TME) impacts tumor growth and metastasis, yet the role of the TME in PCa metastasis to bone is not fully understood. We used a tissue-engineered xenograft approach in NOD-scid IL2Rγnull (NSG) mice to incorporate two levels of humanization; the primary tumor and TME, and the secondary metastatic bone organ. Bioluminescent imaging, histology, and immunohistochemistry were used to study metastasis of human PC-3 and LNCaP PCa cells from the prostate to tissue-engineered bone. Here we show pre-seeding scaffolds with human osteoblasts increases the human cellular and extracellular matrix content of bone constructs, compared to unseeded scaffolds. The humanized prostate TME showed a trend to decrease metastasis of PC-3 PCa cells to the tissue-engineered bone, but did not affect the metastatic potential of PCa cells to the endogenous murine bones or organs. On the other hand, the humanized TME enhanced LNCaP tumor growth and metastasis to humanized and murine bone. Together this demonstrates the importance of the TME in PCa bone tropism, although further investigations are needed to delineate specific roles of the TME components in this context.


Asunto(s)
Neoplasias Óseas/secundario , Neoplasias de la Próstata/patología , Ingeniería de Tejidos , Microambiente Tumoral , Animales , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Metástasis de la Neoplasia
12.
Med Res Rev ; 30(1): 67-101, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19536865

RESUMEN

Prostate cancer (CaP) is one of the most prevalent malignant diseases among men in Western countries. There is currently no cure for metastatic castrate-resistant CaP, and median survival for these patients is about 18 months; the high mortality rate seen is associated with widespread metastases. Progression of CaP from primary to metastatic disease is associated with several molecular and genetic changes that can affect the expression of specific tumor-associated antigens (TAAs) or receptors on the cell surface. Targeting TAAs is emerging as an area of promise for controlling late-stage and recurrent CaP. Several reviews have summarized the progress made in targeting signaling pathways for CaP but will not be discussed here. We describe some important CaP TAAs. These include prostate stem-cell antigen, prostate-specific membrane antigen, MUC1, epidermal growth factor receptor, platelet-derived growth factor and its receptor, urokinase plasminogen activator and its receptor, and extracellular matrix metalloproteinase inducer. We summarize recent advancements in our understanding of their role in CaP metastasis, as well as potential therapeutic options for targeting CaP TAAs. We also discuss the origin, identification, and characterization of prostate cancer stem cells (CSCs) and the potential benefits of targeting prostate CSCs to overcome chemoresistance and CaP recurrence.


Asunto(s)
Antígenos de Neoplasias/sangre , Neoplasias de la Próstata/terapia , Humanos , Masculino , Metástasis de la Neoplasia , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/patología
13.
Electrophoresis ; 31(11): 1853-61, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20506419

RESUMEN

This is the first 2-DE study using sequential dyes to analyse phospho-, glyco- and total tear protein profiles (Pro-Q Diamond for phosphoprotein, Pro-Q Emerald for glycoprotein and Sypro Ruby for total protein). This method minimised the gel-gel variations, allowing better comparisons among the three profiles and generated a whole map of PTM profiles of tear protein. A novel tear protein, dermcidin, was identified for the first time in this study. The identification of this antimicrobial protein suggests a new model of defence in tears. In addition, we are able to present the first experimental evidence of the presence of glycosylated lipocalin 1 and cystatin S. Nucleobindin 2 was only detected using phospho staining, suggesting it is only phosphorylated in tears. This study provides the groundwork for understanding the PTM of tear proteins and consequently these methods could be useful in the search for biomarkers in tears.


Asunto(s)
Proteínas del Ojo/metabolismo , Procesamiento Proteico-Postraduccional , Espectrometría de Masas en Tándem/métodos , Lágrimas/metabolismo , Anciano , Anciano de 80 o más Años , Electroforesis en Gel Bidimensional , Proteínas del Ojo/análisis , Glicoproteínas/análisis , Glicoproteínas/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Péptidos/análisis , Péptidos/metabolismo , Fosforilación , Reproducibilidad de los Resultados , Lágrimas/química
14.
Clin Cancer Res ; 15(7): 2323-34, 2009 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-19318483

RESUMEN

PURPOSE: To test the effects of a new combination, cytosine deaminase (CD) + uracil phosphoribosyltransferase (UPRT)-mediated gene-directed enzyme prodrug therapy (GDEPT) with interleukin (IL)-12 and IL-18, on (a) growth of murine prostate and remote tumor deposits, (b) mouse survival, and (c) T helper (Th) 1/Th2 serum cytokine balance with a special focus to assess correlation with tumor burden/survival. EXPERIMENTAL DESIGN: Efficacy of intraprostatic administration of adenovirally delivered murine IL-12 and IL-18 against orthotopic RM1 tumors and lung pseudometastases was assessed in C57BL/6 mice. At necropsy, tumor growth, lung colony counts, effects on immune cell infiltration, vasculature, apoptosis, and proliferation were estimated. Next, CDUPRT-GDEPT + cytokines were tested at suboptimal doses in mice with RM1CDUPRT prostate tumors/RM1 lung deposits and analyzed as above. Effects on mouse survival were also assessed. Host immune responses to different treatments were assessed by monitoring 11 serum cytokines using Luminex technology. RESULTS: Our data show that IL-12 and IL-18, when combined with CDUPRT-GDEPT, caused significant reduction in local RM1 tumors and lung colonies with enhanced long-term survival versus individual treatments. A dramatic enhancement of tumor infiltration by a wider repertoire of immune cells and disruption of vasculature implied the combination to be more immunostimulatory and antiangiogenic. Remarkably, lowering of serum IL-4 and monocyte chemoattractant protein-1 (MCP-1) was consistently associated with lower tumor burden (local and systemic), and this, rather than an increase in Th1 cytokines, better predicted treatment efficacy. In addition, mouse survival correlated with substantially higher cytokine (Th1/Th2) levels after treatment. CONCLUSION: Locoregional application of CDUPRT-GDEPT and IL-12/IL-18 was effective against local and systemic prostate cancer and improved survival. Monitoring serum levels of IL-4 and MCP-1 may accurately reflect tumor burden and, hence, host response to therapy.


Asunto(s)
Citocinas/sangre , Citosina Desaminasa/genética , Interleucina-12/genética , Interleucina-18/genética , Pentosiltransferasa/genética , Neoplasias de la Próstata/terapia , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Terapia Combinada , Flucitosina/uso terapéutico , Neoplasias Pulmonares/secundario , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Profármacos/uso terapéutico , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología , Análisis de Supervivencia , Células Th2/inmunología
15.
Clin Cancer Res ; 15(3): 865-75, 2009 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-19188157

RESUMEN

PURPOSE: To investigate the therapeutic potential of 213Bilabeled multiple targeted alpha-radioimmunoconjugates for treating prostate cancer (CaP) micrometastases in mouse models. EXPERIMENTAL DESIGN: PC-3 CaP cells were implanted s.c., in the prostate, and intratibially in NODSCID mice. The expression of multiple tumor-associated antigens on tumor xenografts and micrometastases was detected by immunohistochemistry. Targeting vectors were two monoclonal antibodies, and a plasminogen activator inhibitor type 2 that binds to cell surface urokinase plasminogen activator, labeled with 213Bi using standard methodology. In vivo efficacy of multiple alpha conjugates (MTAT) at different activities was evaluated in these mouse models. Tumor growth was monitored during observations and local regional lymph node metastases were assessed at the end of experiments. RESULTS: The take rate of PC-3 cells was 100% for each route of injection. The tumor-associated antigens (MUC1, urokinase plasminogen activator, and BLCA-38) were heterogeneously expressed on primary tumors and metastatic cancer clusters at transit. A single i.p. injection of MTAT (test) at high and low doses caused regression of the growth of primary tumors and prevented local lymph node metastases in a concentration-dependent fashion; it also caused cancer cells to undergo necrosis and apoptosis. CONCLUSIONS: Our results suggest that MTAT can impede primary PC-3 CaP growth at three different sites in vivo through induction of apoptosis, and can prevent the spread of cancer cells and target lymph node micrometastases in a concentration-dependent manner. MTAT, by targeting multiple antigens, can overcome heterogeneous antigen expression to kill small CaP cell clusters, thus providing a potent therapy for micrometastases.


Asunto(s)
Bismuto , Inmunoconjugados/uso terapéutico , Neoplasias de la Próstata/terapia , Animales , Anticuerpos Monoclonales/uso terapéutico , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Masculino , Ratones , Ratones Desnudos , Ratones SCID , Metástasis de la Neoplasia/prevención & control , Trasplante de Neoplasias , Neoplasias de la Próstata/patología , Radioisótopos/uso terapéutico , Activador de Plasminógeno de Tipo Uroquinasa/antagonistas & inhibidores
16.
Cancers (Basel) ; 12(12)2020 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-33255452

RESUMEN

Recent reports have suggested the role of kallikrein-related peptidase 4 (KLK4) to be that of remodeling the tumor microenvironment in many cancers, including prostate cancer. Notably, these studies have suggested a pro-tumorigenic role for KLK4, especially in prostate cancer. However, these have been primarily in vitro studies, with limited in vivo studies performed to date. Herein, we employed an orthotopic inoculation xenograft model to mimic the growth of primary tumors, and an intracardiac injection to induce metastatic dissemination to determine the in vivo tumorigenic effects of KLK4 overexpressed in PC3 prostate cancer cells. Notably, we found that these KLK4-expressing cells gave rise to smaller localized tumors and decreased metastases than the parent PC-3 cells. To our knowledge, this is the first report of an anti-tumorigenic effect of KLK4, particularly in prostate cancer. These findings also provide a cautionary tale of the need for in vivo analyses to substantiate in vitro experimental data.

17.
EJNMMI Res ; 10(1): 46, 2020 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-32382920

RESUMEN

PURPOSE: Chimeric antibody Miltuximab®, a human IgG1 engineered from the parent antibody MIL-38, is in clinical development for solid tumour therapy. Miltuximab® targets glypican-1 (GPC-1), a cell surface protein involved in tumour growth, which is overexpressed in solid tumours, including prostate cancer (PCa). This study investigated the potential of 89Zr-labelled Miltuximab® as an imaging agent, and 177Lu-labelled Miltuximab® as a targeted beta therapy, in a mouse xenograft model of human prostate cancer. METHODS: Male BALB/c nude mice were inoculated subcutaneously with GPC-1-positive DU-145 PCa cells. In imaging and biodistribution studies, mice bearing palpable tumours received (a) 2.62 MBq [89Zr]Zr-DFO-Miltuximab® followed by PET-CT imaging, or (b) 6 MBq [177Lu]Lu-DOTA-Miltuximab® by Cerenkov imaging, and ex vivo assessment of biodistribution. In an initial tumour efficacy study, mice bearing DU-145 tumours were administered intravenously with 6 MBq [177Lu]Lu-DOTA-Miltuximab® or control DOTA-Miltuximab® then euthanised after 27 days. In a subsequent survival efficacy study, tumour-bearing mice were given 3 or 10 MBq of [177Lu]Lu-DOTA-Miltuximab®, or control, and followed up to 120 days. RESULTS: Antibody accumulation in DU-145 xenografts was detected by PET-CT imaging using [89Zr]Zr-DFO-Miltuximab® and confirmed by Cerenkov luminescence imaging post injection of [177Lu]Lu-DOTA-Miltuximab®. Antibody accumulation was higher (% IA/g) in tumours than other organs across multiple time points. A single injection with 6 MBq of [177Lu]Lu-DOTA-Miltuximab® significantly inhibited tumour growth as compared with DOTA-Miltuximab® (control). In the survival study, mice treated with 10 MBq [177Lu]Lu-DOTA-Miltuximab® had significantly prolonged survival (mean 85 days) versus control (45 days), an effect associated with increased cancer cell apoptosis. Tissue histopathology assessment showed no abnormalities associated with [177Lu]Lu-DOTA-Miltuximab®, in line with other observations of tolerability, including body weight stability. CONCLUSION: These findings demonstrate the potential utility of Miltuximab® as a PET imaging agent ([89Zr]Zr-DFO-Miltuximab®) and a beta therapy ([177Lu]Lu-DOTA-Miltuximab®) in patients with PCa or other GPC-1 expressing tumours.

18.
Proteomics ; 9(7): 1883-92, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19294694

RESUMEN

Bladder cancer (BLCa) is a severe urological cancer of both men and women that commonly recurs and once invasive, is difficult to treat. MINA-05 (CK Life Sciences Int'l, Hong Kong) is a derivative of complex botanical extracts, shown to reduce cellular proliferation of bladder and prostate carcinomas. We tested the effects of MINA-05 against human BLCa cell sublines, B8, B8-RSP-GCK, B8-RSP-LN and C3, from a transitional cell carcinoma, grade IV, to determine the molecular targets of treatment by observing the cellular protein profile. Cells were acclimatised for 48 h then treated for 72 h with concentrations of MINA-05 reflecting 1/2 IC(50), IC(50) and 2 x IC(50) (n = 3) or with vehicle, (0.5% DMSO). Dose-dependant changes in protein abundance were detected and characterised using 2-dimensional electrophoresis and MS. We identified 10 proteins that underwent changes in abundance, pI and/or molecular mass in response to treatment. MINA-05 was shown to influence proteins across numerous functional classes including cytoskeletal proteins, energy metabolism proteins, protein degradation proteins and tumour suppressors, suggesting a global impact on these cell lines. This study implies that the ability of MINA-05 to retard cellular proliferation is attributed to its ability to alter cell cycling, metabolism, protein degradation and the cancer cell environment.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias de la Vejiga Urinaria/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Electroforesis en Gel Bidimensional , Humanos , Proteínas de Neoplasias/metabolismo , Fosfopiruvato Hidratasa/metabolismo , Schisandra , Glycine max , Yucca
19.
Prostate ; 69(15): 1613-23, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19585491

RESUMEN

BACKGROUND: Bone metastasis is a frequent and catastrophic consequence of prostate cancer for which only palliative treatment is available. Animal models of bone metastatic prostate cancer are necessary for understanding disease mechanisms but few models exist. METHODS: We have used the murine prostate carcinoma cell line RM1 to generate a bone metastatic model of prostate cancer. Repeated intracardiac injection of RM1 cells followed by isolation of cells from bone tumors has yielded a cell line with strong bone-metastatic potential, RM1 bone metastatic (BM). RESULTS: This cell line metastasizes to multiple bony sites in over 95% of injected C57BL/6 mice and is far less tropic to soft tissues. Bone tumors produced by the RM1(BM) cell line show no preference for particular skeletal sites as most bones are affected. Histology, and micro-computed tomography show that RM1(BM) cells form osteolytic tumors, but with evidence of osteoblastic changes. In vitro the RM1 cells express E-cadherin but not vimentin, do not form colonies in soft agar, are non-invasive but are more motile than the parent cell line. CONCLUSIONS: This model provides a novel means for identifying cellular and molecular mechanisms that contribute to bone metastasis and allow for preclinical testing of therapies to prevent and treat tumor metastasis to bone. Finally as the syngeneic tumor cells are injected into immunocompetent mice, this model will provide a means to study interactions between the immune system, tumors and bone, and therapies that target such interactions.


Asunto(s)
Neoplasias Óseas/secundario , Neoplasias de la Próstata/patología , Fosfatasa Ácida/sangre , Animales , Neoplasias Óseas/sangre , Neoplasias Óseas/inmunología , Adhesión Celular/fisiología , Línea Celular Tumoral , Movimiento Celular/fisiología , Modelos Animales de Enfermedad , Histocitoquímica , Inmunocompetencia , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Osteocalcina/sangre , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/inmunología , Tomografía Computarizada por Rayos X
20.
Ann Surg Oncol ; 16(10): 2888-94, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19669840

RESUMEN

BACKGROUND: Field cancerization is a feature of HNSCCs. No biological marker in the index tumor has correlated with second primary tumor (SPTs) development. Changes in MDM-2 and epidermal growth factor receptor (EGFR) expression are known to be early neoplastic changes in HNSCC. EGFR expression is correlated with clinical outcomes. This study has assessed the predictive correlation of MDM-2 and EGFR protein expression with clinicopathological parameters and occurrence of SPTs in HNSCC. METHODS: Using immunohistochemistry, 106 patients who were treated for primary laryngeal squamous cell carcinoma were investigated for expression of MDM-2 and EGFR. RESULTS: Positive expression of MDM-2 and EGFR was found in 51 of 106 (48.1%) and 82 of 106 (77.4%) cases, respectively. EGFR expression was found to correlate with diagnosis of new primary tumors (P = 0.003), disease-free survival (P = 0.008), as well as overall survival (P = 0.003). MDM-2 expression correlated with nodal relapse (P = 0.03). CONCLUSIONS: SPTs relate to poor prognosis in HNSCC, indicating that closer clinical surveillance of this patient group would be beneficial. Examination of the expression of EGFR by the primary tumor could have potential clinical benefits because this study suggests that it may become a vital biomarker for patients who are most at risk of developing SPTs.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma de Células Escamosas/metabolismo , Receptores ErbB/metabolismo , Neoplasias Laríngeas/metabolismo , Neoplasias Primarias Secundarias/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Anciano , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/terapia , Estudios de Cohortes , Femenino , Estudios de Seguimiento , Humanos , Técnicas para Inmunoenzimas , Neoplasias Laríngeas/patología , Neoplasias Laríngeas/terapia , Masculino , Estadificación de Neoplasias , Neoplasias Primarias Secundarias/diagnóstico , Neoplasias Primarias Secundarias/terapia , Pronóstico , Tasa de Supervivencia , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA