Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Cell Commun Signal ; 17(1): 120, 2019 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-31530281

RESUMEN

BACKGROUND: Prostate cancer development involves various mechanisms, which are poorly understood but pointing to epithelial mesenchymal transition (EMT) as the key mechanism in progression to metastatic disease. ABI1, a member of WAVE complex and actin cytoskeleton regulator and adaptor protein, acts as tumor suppressor in prostate cancer but the role of ABI1 in EMT is not clear. METHODS: To investigate the molecular mechanism by which loss of ABI1 contributes to tumor progression, we disrupted the ABI1 gene in the benign prostate epithelial RWPE-1 cell line and determined its phenotype. Levels of ABI1 expression in prostate organoid tumor cell lines was evaluated by Western blotting and RNA sequencing. ABI1 expression and its association with prostate tumor grade was evaluated in a TMA cohort of 505 patients and metastatic cell lines. RESULTS: Low ABI1 expression is associated with biochemical recurrence, metastasis and death (p = 0.038). Moreover, ABI1 expression was significantly decreased in Gleason pattern 5 vs. pattern 4 (p = 0.0025) and 3 (p = 0.0012), indicating an association between low ABI1 expression and highly invasive prostate tumors. Disruption of ABI1 gene in RWPE-1 cell line resulted in gain of an invasive phenotype, which was characterized by a loss of cell-cell adhesion markers and increased migratory ability of RWPE-1 spheroids. Through RNA sequencing and protein expression analysis, we discovered that ABI1 loss leads to activation of non-canonical WNT signaling and EMT pathways, which are rescued by re-expression of ABI1. Furthermore, an increase in STAT3 phosphorylation upon ABI1 inactivation and the evidence of a high-affinity interaction between the FYN SH2 domain and ABI1 pY421 support a model in which ABI1 acts as a gatekeeper of non-canonical WNT-EMT pathway activation downstream of the FZD2 receptor. CONCLUSIONS: ABI1 controls prostate tumor progression and epithelial plasticity through regulation of EMT-WNT pathway. Here we discovered that ABI1 inhibits EMT through suppressing FYN-STAT3 activation downstream from non-canonical WNT signaling thus providing a novel mechanism of prostate tumor suppression.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Carcinogénesis/genética , Proteínas del Citoesqueleto/deficiencia , Proteínas del Citoesqueleto/genética , Transición Epitelial-Mesenquimal/genética , Técnicas de Inactivación de Genes , Neoplasias de la Próstata/patología , Vía de Señalización Wnt/genética , Cadherinas/metabolismo , Adhesión Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Receptores Frizzled/metabolismo , Humanos , Masculino , Clasificación del Tumor , Fenotipo , Recurrencia , Factor de Transcripción STAT3/metabolismo , Regulación hacia Arriba/genética , beta Catenina/metabolismo
2.
Invest New Drugs ; 30(6): 2161-72, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22203214

RESUMEN

The epidermal growth factor receptor (EGFR) pathway is aberrantly activated in tumors and plays a key role in promoting tumor growth. Small molecule inhibitors which bind reversibly to EGFR have demonstrated limited clinical activity. Thus, there is a continued need to develop novel EGFR inhibitors with improved anti-tumor activity. Bay846 is a newly developed small molecule inhibitor that binds irreversibly to the tyrosine kinase domains of EGFR and Her2. The in vitro and in vivo efficacy of Bay846 was tested using a panel of nine human malignant brain tumor (glioma) models. Lapatinib, a reversible inhibitor of EGFR and Her2, was included for comparison. Six glioma cell lines were sensitive to Bay846 treatment. Bay846 strongly suppressed tumor cell growth in vitro by inducing cell lysis/death rather than cell cycle arrest. Consistent with this, Bay846 had potent anti-tumor activity which led to regressions in tumor size. The active, phosphorylated form of EGFR was reduced by Bay846 treatment in vitro and in tumors. Importantly, the efficacy of Bay846 was significantly greater than lapatinib in all assays. Bay846-sensitivity was associated with expression of a wild-type PTEN in conjunction with high levels of an oncogenic EGFR variant (A289V or EGFRvIII). These studies demonstrate that targeting the EGFR pathway with the irreversible inhibitor Bay846 has great potential to increase the efficacy of this cancer therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Receptores ErbB/antagonistas & inhibidores , Glioma/tratamiento farmacológico , Receptor ErbB-2/antagonistas & inhibidores , Animales , Antineoplásicos/farmacología , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Receptores ErbB/genética , Receptores ErbB/metabolismo , Glioma/metabolismo , Glioma/patología , Humanos , Ratones , Ratones Desnudos , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Receptor ErbB-2/metabolismo , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Cancer Res ; 67(14): 6872-81, 2007 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-17638898

RESUMEN

There is a need for novel therapies targeting hypoxic cells in tumors. These cells are associated with tumor resistance to therapy and express hypoxia inducible factor-1 (HIF-1), a transcription factor that mediates metabolic adaptation to hypoxia and activates tumor angiogenesis. We previously developed an oncolytic adenovirus (HYPR-Ad) for the specific killing of hypoxic/HIF-active tumor cells, which we now armed with an interleukin-4 gene (HYPR-Ad-IL4). We designed HYPR-Ad-IL4 by cloning the Ad E1A viral replication and IL-4 genes under the regulation of a bidirectional hypoxia/HIF-responsive promoter. The IL-4 cytokine was chosen for its ability to induce a strong host antitumor immune response and its potential antiangiogenic activity. HYPR-Ad-IL4 induced hypoxia-dependent IL-4 expression, viral replication, and conditional cytolysis of hypoxic, but not normoxic cells. The treatment of established human tumor xenografts with HYPR-Ad-IL4 resulted in rapid and maintained tumor regression with the same potency as that of wild-type dl309-Ad. HYPR-Ad-IL4-treated tumors displayed extensive necrosis, fibrosis, and widespread viral replication. Additionally, these tumors contained a distinctive leukocyte infiltrate and prominent hypoxia. The use of an oncolytic Ad that locally delivers IL-4 to tumors is novel, and we expect that HYPR-Ad-IL4 will have broad therapeutic use for all solid tumors that have hypoxia or active HIF, regardless of tissue origin or genetic alterations.


Asunto(s)
Adenoviridae/metabolismo , Terapia Genética/métodos , Factor 1 Inducible por Hipoxia/metabolismo , Interleucina-4/genética , Interleucina-4/metabolismo , Viroterapia Oncolítica/métodos , Virus Oncolíticos/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular , Clonación Molecular , Humanos , Hipoxia , Ratones , Modelos Genéticos , Trasplante de Neoplasias
4.
Mol Biol Cell ; 29(5): 532-541, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29282278

RESUMEN

We propose to understand how the mitotic kinase PLK1 drives chromosome segregation errors, with a specific focus on Gravin, a PLK1 scaffold. In both three-dimensional primary prostate cancer cell cultures that are prone to Gravin depletion and Gravin short hairpin RNA (shRNA)-treated cells, an increase in cells containing micronuclei was noted in comparison with controls. To examine whether the loss of Gravin affected PLK1 distribution and activity, we utilized photokinetics and a PLK1 activity biosensor. Gravin depletion resulted in an increased PLK1 mobile fraction, causing the redistribution of active PLK1, which leads to increased defocusing and phosphorylation of the mitotic centrosome protein CEP215 at serine-613. Gravin depletion further led to defects in microtubule renucleation from mitotic centrosomes, decreased kinetochore-fiber integrity, increased incidence of chromosome misalignment, and subsequent formation of micronuclei following mitosis completion. Murine Gravin rescued chromosome misalignment and micronuclei formation, but a mutant Gravin that cannot bind PLK1 did not. These findings suggest that disruption of a Gravin-PLK1 interface leads to inappropriate PLK1 activity contributing to chromosome segregation errors, formation of micronuclei, and subsequent DNA damage.


Asunto(s)
Proteínas de Anclaje a la Quinasa A/metabolismo , Proteínas de Ciclo Celular/metabolismo , Centrosoma/metabolismo , Mitosis , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas de Anclaje a la Quinasa A/genética , Animales , Proteínas de Ciclo Celular/genética , Segregación Cromosómica , Daño del ADN , Fibroblastos , Células HEK293 , Células HeLa , Humanos , Ratones , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Huso Acromático/metabolismo , Quinasa Tipo Polo 1
5.
Cancer Res ; 65(4): 1406-13, 2005 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-15735028

RESUMEN

We have previously proposed that intravascular thrombosis and subsequent vasoocclusion contribute to the development of pseudopalisading necrosis, a pathologic hallmark that distinguishes glioblastoma (WHO grade 4) from lower grade astrocytomas. To better understand the potential prothrombotic mechanisms underlying the formation of these structures that drive tumor angiogenesis, we investigated tissue factor (TF), a potent procoagulant protein known to be overexpressed in astrocytomas. We hypothesized that PTEN loss and tumor hypoxia, which characterize glioblastoma but not lower grade astrocytomas, could up-regulate TF expression and cause intravascular thrombotic occlusion. We examined the effect of PTEN restoration and hypoxia on TF expression and plasma coagulation using a human glioma cell line containing an inducible wt-PTEN cDNA. Cell exposure to hypoxia (1% O(2)) markedly increased TF expression, whereas restoration of wt-PTEN caused decreased cellular TF. The latter effect was at least partially dependent on PTEN's protein phosphatase activity. Hypoxic cells accelerated plasma clotting in tilt tube assays and this effect was prevented by both inhibitory antibodies to TF and plasma lacking factor VII, implicating TF-dependent mechanisms. To further examine the genetic events leading to TF up-regulation during progression of astrocytomas, we investigated its expression in a series of human astrocytes sequentially infected with E6/E7/human telomerase, Ras, and Akt. Cells transformed with Akt showed the greatest incremental increase in hypoxia-induced TF expression and secretion. Together, our results show that PTEN loss and hypoxia up-regulate TF expression and promote plasma clotting by glioma cells, suggesting that these mechanisms may underlie intravascular thrombosis and pseudopalisading necrosis in glioblastoma.


Asunto(s)
Coagulación Sanguínea/fisiología , Glioblastoma/metabolismo , Monoéster Fosfórico Hidrolasas/fisiología , Tromboplastina/biosíntesis , Proteínas Supresoras de Tumor/fisiología , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Glioblastoma/sangre , Glioblastoma/genética , Humanos , Fosfohidrolasa PTEN , Fosfatidato Fosfatasa/metabolismo , Monoéster Fosfórico Hidrolasas/biosíntesis , Monoéster Fosfórico Hidrolasas/deficiencia , Monoéster Fosfórico Hidrolasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Tromboplastina/genética , Proteínas Supresoras de Tumor/biosíntesis , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética , Regulación hacia Arriba , Proteínas ras/metabolismo
6.
Cancer Res ; 62(9): 2478-82, 2002 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-11980636

RESUMEN

Hypoxia-inducible factor 1 (HIF-1) is a heterodimeric transcription factor composed of alpha and beta subunits. HIF-1 is critically involved in cellular responses to hypoxia, glycolysis, and angiogenesis. Here, we show that treatment of prostate cancer PC-3 and LNCaP cells with the benzoquinone ansamycin geldanamycin, an Hsp90-specific inhibitor, induced degradation of HIF-1alpha protein in a dose- and time-dependent manner under both normoxia and hypoxia. This inhibition was also shown in other common cancer types tested. Rapid degradation of nuclear HIF-1alpha protein levels was accompanied by respective inhibition in HIF-1alpha functional transcription activity of VEGF. No difference between HIF-1alpha mRNA levels before or after geldanamycin treatment was found. Moreover, [35S]methionine pulse-chase analysis revealed that HIF-1alpha protein half-life was markedly decreased in the presence of geldanamycin compared with that in control. The geldanamycin-induced degradation of HIF-1alpha was reversed by proteosome inhibitors lactacystin and MG-132. We conclude that geldanamycin induces reduction of HIF-1alpha levels and its downstream transcriptional activity by accelerating protein degradation independent of O2 tension. Thus, benzoquinone ansamycin drugs and their derivatives, such as 17-allyl-aminogeldanamycin, are excellent candidates as small molecule drug inhibitors of HIF-1 overexpression in cancer cells.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Neoplasias de la Próstata/metabolismo , Quinonas/farmacología , Factores de Transcripción/metabolismo , Antibióticos Antineoplásicos/farmacocinética , Benzoquinonas , Relación Dosis-Respuesta a Droga , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia , Lactamas Macrocíclicas , Masculino , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Quinonas/farmacocinética , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Transcripción Genética/efectos de los fármacos , Células Tumorales Cultivadas
7.
Cell Rep ; 14(4): 872-884, 2016 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-26804907

RESUMEN

The molecular chaperone Hsp90 protects deregulated signaling proteins that are vital for tumor growth and survival. Tumors generally display sensitivity and selectivity toward Hsp90 inhibitors; however, the molecular mechanism underlying this phenotype remains undefined. We report that the mitotic checkpoint kinase Mps1 phosphorylates a conserved threonine residue in the amino-domain of Hsp90. This, in turn, regulates chaperone function by reducing Hsp90 ATPase activity while fostering Hsp90 association with kinase clients, including Mps1. Phosphorylation of Hsp90 is also essential for the mitotic checkpoint because it confers Mps1 stability and activity. We identified Cdc14 as the phosphatase that dephosphorylates Hsp90 and disrupts its interaction with Mps1. This causes Mps1 degradation, thus providing a mechanism for its inactivation. Finally, Hsp90 phosphorylation sensitizes cells to its inhibitors, and elevated Mps1 levels confer renal cell carcinoma selectivity to Hsp90 drugs. Mps1 expression level can potentially serve as a predictive indicator of tumor response to Hsp90 inhibitors.


Asunto(s)
Carcinoma de Células Renales/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Neoplasias Renales/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Secuencia de Aminoácidos , Antineoplásicos/farmacología , Proteínas de Ciclo Celular/metabolismo , Inhibidores Enzimáticos/farmacología , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Humanos , Datos de Secuencia Molecular , Fosforilación , Unión Proteica , Proteolisis , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/metabolismo
8.
Oncogene ; 22(14): 2065-72, 2003 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-12687009

RESUMEN

New therapy targeting the hypoxic fraction of tumors needs to be designed as this population of cells is the most resistant to radio- and chemotherapies. Hypoxia-inducible factor (HIF) mediates transcriptional responses to hypoxia by binding to hypoxia-responsive elements (HRE) in target genes. We developed a hypoxia/HIF-dependent replicative adenovirus (HYPR-Ad) to target hypoxic cells. HYPR-Ad displays hypoxia-dependent E1A expression and conditional cytolysis of hypoxic but not normoxic cells. This work provides proof-of-principle evidence that an attenuated oncolytic adenovirus that selectively lyses cells under hypoxia can be generated. This therapeutic approach can be used to treat all solid tumors that develop hypoxia, regardless of their tissue origin or genetic alterations.


Asunto(s)
Adenoviridae/genética , Neoplasias Encefálicas/terapia , Hipoxia de la Célula/genética , Proteínas de Unión al ADN/farmacología , Terapia Genética , Glioma/terapia , Proteínas Nucleares/farmacología , Factores de Transcripción , Neoplasias Encefálicas/metabolismo , Vectores Genéticos , Glioma/metabolismo , Humanos , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Células Tumorales Cultivadas
9.
Clin Cancer Res ; 10(16): 5299-312, 2004 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-15328165

RESUMEN

Oncolytic virotherapy is the use of genetically engineered viruses that specifically target and destroy tumor cells via their cytolytic replication cycle. Viral-mediated tumor destruction is propagated through infection of nearby tumor cells by the newly released progeny. Each cycle should amplify the number of oncolytic viruses available for infection. Our understanding of the life cycles of cytolytic viruses has allowed manipulation of their genome to selectively kill tumor cells over normal tissue. Because the mechanism of tumor destruction is different, oncolytic virotherapy should work synergistically with current modes of treatment such as chemotherapy and radiation therapy. This article focuses on oncolytic adenoviruses that have been created and tested in preclinical and clinical trials in combination with chemotherapy, radiation therapy, and gene therapy.


Asunto(s)
Adenoviridae/fisiología , Neoplasias/patología , Neoplasias/terapia , Replicación Viral/fisiología , Animales , Ensayos Clínicos como Asunto , Terapia Combinada , Humanos
10.
Clin Cancer Res ; 10(24): 8603-12, 2004 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-15623644

RESUMEN

Hypoxia plays a critical role in driving tumor malignancy and is associated with poor patient survival in many human cancers. Novel therapies targeting hypoxic tumor cells are urgently needed, because these cells hinder tumor eradication. Here we demonstrate than an anticancer strategy based on intratumoral delivery of a novel type of oncolytic adenovirus targeting tumor hypoxia is therapeutically efficient and can augment standard chemotherapy. We used a conditionally replicative adenovirus (HYPR-Ad) to specifically kill hypoxic tumor cells. Viral infection and conditional replication occurred efficiently in hypoxic/hypoxia-inducible factor-active cells in culture and in vivo, prevented tumor formation, and reduced the growth of established tumors. Combining HYPR-Ad with chemotherapy effective against normoxic cells resulted in strongly enhanced antitumor efficacy. These studies demonstrate that targeting the hypoxic microenvironment of tumors rather than an intrinsic gene expression defect is a viable and novel antitumor therapeutic strategy that can be used in combination with existing treatment regimens. The replication and oncolytic potential of this virus was made dependent on hypoxic/hypoxia-inducible factor, a transcription factor activated in the tumor hypoxic microenvironment, broadening its therapeutic use to solid tumors of any genetic make-up or tissue of origin.


Asunto(s)
Adenoviridae/genética , Proteínas E1A de Adenovirus/genética , Neoplasias Encefálicas/terapia , Hipoxia de la Célula/genética , Glioma/terapia , Replicación Viral/fisiología , Animales , Antineoplásicos Alquilantes/uso terapéutico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/virología , Carmustina/uso terapéutico , Terapia Combinada , Proteínas de Unión al ADN/metabolismo , Glioma/genética , Glioma/virología , Humanos , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Ratones , Ratones Desnudos , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Hum Gene Ther ; 14(10): 933-46, 2003 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-12869212

RESUMEN

The use of replication-competent viruses that have a cytolytic cycle has emerged as a viable strategy (oncolytic virotherapy) to specifically kill tumor cells and the field has advanced to the point of clinical trials. A theoretical advantage of replicative oncolytic viruses is that their numbers should increase via viral replication within infected tumor cells and resulting viral progeny can then infect additional cells within the tumor mass. The life cycle of a virus involves multiple interactions between viral and cellular proteins/genes, which maximize the ability of the virus to infect and replicate within cells. Understanding such interactions has led to the design of numerous genetically engineered adenovirus (Ad) vectors that selectively kill tumor cells while sparing normal cells. These viruses have also been modified to function as therapeutic gene delivery vehicles, thus augmenting their anticancer capacity. In addition, the oncolytic mode of tumor killing differs from that of standard anticancer therapies, providing the possibility for synergistic interactions with other therapies in a multimodal antitumor approach. In this review, we describe the oncolytic Ad vectors tested in preclinical and clinical models and their use in combination with chemo-, radio-, and gene therapies.


Asunto(s)
Adenoviridae/genética , Vectores Genéticos/uso terapéutico , Neoplasias/terapia , Adenoviridae/fisiología , Terapia Combinada , Humanos , Regiones Promotoras Genéticas , Replicación Viral
12.
Curr Gene Ther ; 4(1): 41-51, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15032613

RESUMEN

Viruses that kill the host cell during their replication cycle have attracted much interest for the specific killing of tumor cells and this oncolytic virotherapy is being evaluated in clinical trials. The rationale for using replicative oncolytic viruses is that viral replication in infected tumor cells will permit in situ viral multiplication and spread of viral infection throughout the tumor mass thus overcoming the delivery problems of gene therapy. Improved understanding of the life cycle of viruses has evidenced multiple interactions between viral and cellular gene products, which have evolved to maximize the ability of viruses to infect and multiply within cells. Differences in viral-cell interactions between normal and tumor cells have emerged that have led to the design of a number of genetically engineered viral vectors that selectively kill tumor cells while sparing normal cells. These viruses have undergone further modifications to carry adjunct therapy genes to increase their anti-cancer abilities. Since these viruses kill cells by oncolytic mechanisms differing from standard anticancer therapies, there is an opportunity that synergistic interactions with other therapies might be found with the use of combination therapy. In this review, we focus on the oncolytic Herpes Simplex Virus-1 (HSV-1) vectors that have been examined in preclinical and clinical cancer models and their use in combination with chemo-, radio-, and gene therapies.


Asunto(s)
Terapia Genética , Vectores Genéticos , Herpesvirus Humano 1/genética , Neoplasias/terapia , Quimioterapia Adyuvante , Genes Transgénicos Suicidas , Vectores Genéticos/administración & dosificación , Humanos , Mutación , Radioterapia Adyuvante , Replicación Viral
13.
Future Oncol ; 1(2): 247-58, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16555996

RESUMEN

Oncolytic therapy is a novel anticancer treatment with attenuated lytic viruses such as adenovirus (Ad). These viruses kill the host cells through their lytic replication cycle and are thus distinct from classical gene therapy viruses, which serve as gene delivery agents and do not replicate. Oncolytic Ads are genetically engineered so as to replicate only in cancer cells. Their replication cycle leads to viral multiplication, the killing of the host cells and spreading of the infection throughout the tumor. Following success in preclinical studies, their anti-tumor potential is now being evaluated in the clinic. Three oncolytic Ads (dl1520, Ad5-CD/TKrep, and CV706) have completed Phase I and II clinical trials in cancer patients where their administration via multiple routes and in combination with chemo- or radiotherapies, has demonstrated overall safety. These viruses are being re-engineered to arm them with additional therapeutic genes, bolstering their oncolytic activity with a bystander effect. For example, Ad5-CD/TKrep delivers a therapeutic prodrug-activating (suicide) gene. These data indicate that oncolytic Ads are a promising novel cancer treatment approach that can be combined with other modalities, such as gene therapy and classical chemo- and radiotherapies. Further improvements to enhance their specificity, targeting and oncolytic activity are needed however, as these first-generation viruses showed modest anti-tumor activity. To improve their efficacy in the clinic, it will be important to devise and incorporate novel monitoring techniques in the clinical trials, such as analysis of viral replication in biopsies and through the use of creative noninvasive imaging technologies.


Asunto(s)
Neoplasias/terapia , Viroterapia Oncolítica , Virus Oncolíticos/fisiología , Replicación Viral , Predicción , Humanos , Neoplasias/virología
14.
J Neurooncol ; 70(2): 229-43, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15674480

RESUMEN

Infiltrative astrocytic neoplasms are by far the most common malignant brain tumors in adults. Clinically, they are highly problematic due to their widely invasive nature which makes a complete resection almost impossible. Biologic progression of these tumors is inevitable and adjuvant therapies are only moderately effective in prolonging survival. Glioblastoma multiforme (GBM; WHO grade IV), the most malignant form of infiltrating astrocytoma, can evolve from a lower grade precursor tumor (secondary GBM) or can present as high grade lesion from the outset, so-called de novo GBM. Molecular genetic investigations suggest that GBMs are comprised of multiple molecular genetic subsets. Notwithstanding the diversity of genetic alterations leading to the GBM phenotype, the vascular changes that evolve in this disease, presumably favoring further growth, are remarkably similar. Underlying genetic alterations in GBM may tilt the balance in favor of an angiogenic phenotype by upregulation of pro-angiogenic factors and down-regulation of angiogenesis inhibitors. Increased vascularity and endothelial cell proliferation in GBMs are also driven by hypoxia-induced expression of pro-angiogenic cytokines, such vascular endothelial growth factor (VEGF). Understanding the contribution of genetic alterations and hypoxia in angiogenic dysregulation in astrocytic neoplasms will lead to the development of better anti-angiogenic therapies for this disease. This review will summarize the properties of angiogenic dysregulation that lead to the highly vascularized nature of these tumors.


Asunto(s)
Neoplasias Encefálicas/irrigación sanguínea , Glioma/irrigación sanguínea , Hipoxia/fisiopatología , Neovascularización Patológica/genética , Transducción de Señal/fisiología , Animales , Neoplasias Encefálicas/genética , Genotipo , Glioma/genética , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA