Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Clin Invest ; 133(22)2023 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-37966111

RESUMEN

Prostate cancer is generally considered an immunologically "cold" tumor type that is insensitive to immunotherapy. Targeting surface antigens on tumors through cellular therapy can induce a potent antitumor immune response to "heat up" the tumor microenvironment. However, many antigens expressed on prostate tumor cells are also found on normal tissues, potentially causing on-target, off-tumor toxicities and a suboptimal therapeutic index. Our studies revealed that six-transmembrane epithelial antigen of prostate-2 (STEAP2) was a prevalent prostate cancer antigen that displayed high, homogeneous cell surface expression across all stages of disease with limited distal normal tissue expression, making it ideal for therapeutic targeting. A multifaceted lead generation approach enabled development of an armored STEAP2 chimeric antigen receptor T cell (CAR-T) therapeutic candidate, AZD0754. This CAR-T product was armored with a dominant-negative TGF-ß type II receptor, bolstering its activity in the TGF-ß-rich immunosuppressive environment of prostate cancer. AZD0754 demonstrated potent and specific cytotoxicity against antigen-expressing cells in vitro despite TGF-ß-rich conditions. Further, AZD0754 enforced robust, dose-dependent in vivo efficacy in STEAP2-expressing cancer cell line-derived and patient-derived xenograft mouse models, and exhibited encouraging preclinical safety. Together, these data underscore the therapeutic tractability of STEAP2 in prostate cancer as well as build confidence in the specificity, potency, and tolerability of this potentially first-in-class CAR-T therapy.


Asunto(s)
Neoplasias de la Próstata , Receptores Quiméricos de Antígenos , Masculino , Humanos , Ratones , Animales , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/metabolismo , Inmunoterapia Adoptiva , Neoplasias de la Próstata/patología , Linfocitos T , Factor de Crecimiento Transformador beta/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Línea Celular Tumoral , Microambiente Tumoral , Oxidorreductasas/metabolismo
2.
Oncotarget ; 9(33): 22960-22975, 2018 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-29796165

RESUMEN

Despite recent advances in treatment, breast cancer remains the second-most common cause of cancer death among American women. A greater understanding of the molecular characteristics of breast tumors could ultimately lead to improved tumor-targeted treatment options, particularly for subsets of breast cancer patients with unmet needs. Using an unbiased genomics approach to uncover membrane-localized tumor-associated antigens (TAAs), we have identified glial cell line derived neurotrophic factor (GDNF) family receptor α 1 (GFRA1) as a breast cancer TAA. Immunohistochemistry (IHC) revealed that GFRA1 displays a limited normal tissue expression profile coupled with overexpression in specific breast cancer subsets. The cell surface localization as determined by fluorescence-activated cell sorting (FACS) and the rapid internalization kinetics of GFRA1 makes it an ideal target for therapeutic exploitation as an antibody-drug conjugate (ADC). Here, we describe the development of a pyrrolobenzodiazepine (PBD)-armed, GFRA1-targeted ADC that demonstrates cytotoxicity in GFRA1-positive cell lines and patient-derived xenograft (PDX) models. The safety profile of the rat cross-reactive GFRA1-PBD was assessed in a rat toxicology study to find transient cellularity reductions in the bone marrow and peripheral blood, consistent with known off-target effects of PBD ADC's. These studies reveal no evidence of on-target toxicity and support further evaluation of GFRA1-PBD in GFRA1-positive tumors.

3.
Chem Biol ; 19(2): 228-42, 2012 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-22365606

RESUMEN

The NADPH oxidase enzyme complex, NOX2, is responsible for reactive oxygen species production in neutrophils and has been recognized as a key mediator of inflammation. Here, we have performed rational design and in silico screen to identify a small molecule inhibitor, Phox-I1, targeting the interactive site of p67(phox) with Rac GTPase, which is a necessary step of the signaling leading to NOX2 activation. Phox-I1 binds to p67(phox) with a submicromolar affinity and abrogates Rac1 binding and is effective in inhibiting NOX2-mediated superoxide production dose-dependently in human and murine neutrophils without detectable toxicity. Medicinal chemistry characterizations have yielded promising analogs and initial information of the structure-activity relationship of Phox-I1. Our studies suggest the potential utility of Phox-I class inhibitors in NOX2 oxidase inhibition and present an application of rational targeting of a small GTPase-effector interface.


Asunto(s)
Benzoxazinas/farmacología , Diseño de Fármacos , Inflamación/metabolismo , Fosfoproteínas/antagonistas & inhibidores , Pirazoles/farmacología , Transducción de Señal/efectos de los fármacos , Proteínas de Unión al GTP rac/antagonistas & inhibidores , Sitios de Unión , Simulación por Computador , Células HL-60 , Humanos , Inflamación/patología , Neutrófilos/metabolismo , Fosfoproteínas/metabolismo , Estructura Terciaria de Proteína , Especies Reactivas de Oxígeno/metabolismo , Relación Estructura-Actividad , Proteínas de Unión al GTP rac/metabolismo
4.
Blood ; 115(16): 3320-8, 2010 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-20179179

RESUMEN

Mutation of the p53 tumor suppressor is associated with disease progression, therapeutic resistance, and poor prognosis in patients with lymphoid malignancies and can occur in approximately 50% of Burkitt lymphomas. Thus, new therapies are needed to specifically target p53-deficient lymphomas with increased efficacy. In the current study, the specific impact of inhibition of the small GTPase Rac1 on p53-deficient B- and T-lymphoma cells was investigated. p53 deficiency resulted in increased Rac1 activity in both B-cell and T-cell lines, and its suppression was able to abrogate p53 deficiency-mediated lymphoma cell proliferation. Further, Rac targeting resulted in increased apoptosis via a p53-independent mechanism. By probing multiple signaling axes and performing rescue studies, we show that the antiproliferative effect of Rac1 targeting in lymphoma cells may involve the PAK and Akt signaling pathway, but not the mitogen-activated protein (MAP) kinase pathway. The effects of inhibition of Rac1 were extended in vivo where Rac1 targeting was able to specifically impair p53-deficient lymphoma cell growth in mouse xenografts and postpone lymphomagenesis onset in murine transplantation models. Because the Rac1 signaling axis is a critical determinant of apoptosis and tumorigenesis, it may represent an important basis for therapy in the treatment of p53-deficient lymphomas.


Asunto(s)
Linfoma/metabolismo , Transducción de Señal/fisiología , Proteína p53 Supresora de Tumor/deficiencia , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Animales , Apoptosis/fisiología , Línea Celular Tumoral , Separación Celular , Citometría de Flujo , Humanos , Immunoblotting , Linfoma/genética , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Cell Cycle ; 6(6): 667-71, 2007 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-17361100

RESUMEN

Cancer is a highly heterogeneous disease, wherein specific determinants modulate disease severity and therapeutic outcomes. In breast cancer, significant effort has been channeled into defining critical genetic effectors of disease behavior. One key molecular determinant is the retinoblastoma tumor suppressor (RB), which is functionally inactivated in the majority of human cancers, and aberrant in nearly half of breast cancers. Deficiency in RB function compromises cell cycle checkpoints, contributes to aggressive tumor proliferation, and is associated with advanced disease. Recent investigation indicates that RB-deficiency has dramatic and disparate effects on the response to therapeutic modalities utilized in the treatment of breast cancer. Loss of RB function promotes inappropriate cell cycle progression during therapeutic challenge. In the context of cytotoxic therapies, this lack of checkpoint function leads to increased sensitivity to the agent. However, RB-deficiency efficiently bypasses the anti-mitogenic function of hormonal therapies and is associated with early disease recurrence following tamoxifen therapy. Thus, RB-pathway status has powerful effects on both tumorigenic proliferation and therapeutic response, and may represent a critical basis for informing breast cancer therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/metabolismo , Proteína de Retinoblastoma/fisiología , Animales , Antineoplásicos/farmacología , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Femenino , Humanos , Proteína de Retinoblastoma/antagonistas & inhibidores , Proteína de Retinoblastoma/genética
6.
Cancer Lett ; 245(1-2): 205-17, 2007 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-16574317

RESUMEN

Schwann cells play a critical role in peripheral nerve function. Regulated proliferation of Schwann cells is an important facet of the response to nerve injury; however, aberrant proliferation can give rise to Schwann cell tumors such as malignant peripheral nerve sheath tumors (MPNST). These tumors exhibit a range of genetic lesions that include loss of the retinoblastoma tumor suppressor (RB) pathway. RB plays a critical role in the regulation of cellular proliferation and its loss is a common event in human cancers. Here, the specific action of RB loss on Schwann cell proliferation and response to therapeutic intervention was explored. In primary mouse Schwann cells, conditional RB loss led to increased levels of critical cell cycle regulatory gene products, yet provided only a modest influence on proliferation. However, RB-deficient Schwann cells efficiently bypassed the cell cycle inhibitory response to the chemotherapeutic agent cisplatin, which is used in the treatment of MPNST and other glial tumors. Surprisingly, RB loss did not facilitate Schwann cell immortalization; and RB-deficient cells actually were less prone to immortalization than cells containing RB. Furthermore, RB-deficient cells that ultimately re-entered the cell cycle had lost both Schwann cell morphology and markers. Since, RB loss is likely a late event in Schwann cell tumor progression, the action of acute RB loss in immortalized Schwann cells was investigated. In this context, loss of RB had a profound effect on expression of target genes and the response to cisplatin. Thus, the loss of RB in both primary and immortal Schwann cells disrupted the response to anti-mitogenic signals and has implications for therapeutic intervention.


Asunto(s)
Ciclo Celular/fisiología , Transformación Celular Neoplásica , Cisplatino/farmacología , Proteína de Retinoblastoma/fisiología , Células de Schwann/metabolismo , Animales , Antineoplásicos/farmacología , Ciclo Celular/efectos de los fármacos , Proliferación Celular , Células Cultivadas , Ganglios Espinales/citología , Ganglios Espinales/metabolismo , Eliminación de Gen , Ratones , Ratones Endogámicos , Microscopía Fluorescente , Antígeno Nuclear de Célula en Proliferación/análisis , Receptores de Factor de Crecimiento Nervioso/análisis , Proteína de Retinoblastoma/deficiencia , Proteína de Retinoblastoma/genética , Células de Schwann/efectos de los fármacos , Células de Schwann/patología
7.
J Clin Invest ; 117(1): 218-28, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17160137

RESUMEN

The retinoblastoma tumor suppressor (RB) protein is functionally inactivated in the majority of human cancers and is aberrant in one-third of all breast cancers. RB regulates G(1)/S-phase cell-cycle progression and is a critical mediator of antiproliferative signaling. Here the specific impact of RB deficiency on E2F-regulated gene expression, tumorigenic proliferation, and the response to 2 distinct lines of therapy was investigated in breast cancer cells. RB knockdown resulted in RB/E2F target gene deregulation and accelerated tumorigenic proliferation, thereby demonstrating that even in the context of a complex tumor cell genome, RB status exerts significant control over proliferation. Furthermore, the RB deficiency compromised the short-term cell-cycle inhibition following cisplatin, ionizing radiation, and antiestrogen therapy. In the context of DNA-damaging agents, this bypass resulted in increased sensitivity to these agents in cell culture and xenograft models. In contrast, the bypass of antiestrogen signaling resulted in continued proliferation and xenograft tumor growth in the presence of tamoxifen. These effects of aberrations in RB function were recapitulated by ectopic E2F expression, indicating that control of downstream target genes was an important determinant of the observed responses. Specific analyses of an RB gene expression signature in 60 human patients indicated that deregulation of this pathway was associated with early recurrence following tamoxifen monotherapy. Thus, because the RB pathway is a critical determinant of tumorigenic proliferation and differential therapeutic response, it may represent a critical basis for directing therapy in the treatment of breast cancer.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Proteína de Retinoblastoma/fisiología , Secuencia de Bases , Línea Celular Tumoral , Cartilla de ADN , Femenino , Fase G1 , Eliminación de Gen , Humanos , Immunoblotting , Proteína de Retinoblastoma/deficiencia , Proteína de Retinoblastoma/genética , Fase S
8.
Cancer Res ; 65(11): 4568-77, 2005 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15930274

RESUMEN

The liver exhibits an exquisitely controlled cell cycle, wherein hepatocytes are maintained in quiescence until stimulated to proliferate. The retinoblastoma tumor suppressor, pRB, plays a central role in proliferative control by inhibiting inappropriate cell cycle entry. In many cases, liver cancer arises due to aberrant cycles of proliferation, and correspondingly, pRB is functionally inactivated in the majority of hepatocellular carcinomas. Therefore, to determine how pRB loss may provide conditions permissive for deregulated hepatocyte proliferation, we investigated the consequence of somatic pRB inactivation in murine liver. We show that liver-specific pRB loss results in E2F target gene deregulation and elevated cell cycle progression during post-natal growth. However, in adult livers, E2F targets are repressed and hepatocytes become quiescent independent of pRB, suggesting that other factors may compensate for pRB loss. Therefore, to probe the consequences of acute pRB inactivation in livers of adult mice, we gave adenoviral-Cre by i.v. injection. We show that acute pRB loss is sufficient to elicit E2F target gene expression and cell cycle entry in adult liver, demonstrating a critical role for pRB in maintaining hepatocyte quiescence. Finally, we show that liver-specific pRB loss results in the development of nuclear pleomorphism associated with elevated ploidy that is evident in adult mice harboring both acute and chronic pRB loss. Together, these results show the crucial role played by pRB in maintaining hepatocyte quiescence and ploidy in adult liver in vivo and underscore the critical importance of delineating the consequences of acute pRB loss in adult animals.


Asunto(s)
Hígado/fisiología , Ploidias , Proteína de Retinoblastoma/deficiencia , Animales , Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Procesos de Crecimiento Celular/genética , Proteínas de Unión al ADN/genética , Factores de Transcripción E2F , Silenciador del Gen , Genes de Retinoblastoma/genética , Hepatocitos/citología , Hepatocitos/fisiología , Hígado/citología , Hígado/metabolismo , Ratones , Ratones Noqueados , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/fisiología , Factores de Transcripción/genética
9.
Nucleic Acids Res ; 33(5): 1581-92, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15767283

RESUMEN

The retinoblastoma tumor suppressor (RB) is functionally inactivated in the majority of cancers and is a critical mediator of DNA damage checkpoints. Despite the critical importance of RB function in tumor suppression, the coordinate impact of RB loss on the response to environmental and therapeutic sources of damage has remained largely unexplored. Here, we utilized a conditional knockout system to ablate RB in adult fibroblasts. This model system enabled us to investigate the temporal role of RB loss on cell cycle checkpoints and DNA damage repair following ultraviolet (UV) and ionizing radiation (IR) damage. We demonstrate that RB loss compromises rapid cell cycle arrest following UV and IR exposure in adult primary cells. Detailed kinetic analysis of the checkpoint response revealed that disruption of the checkpoint is concomitant with RB target gene deregulation, and is not simply a manifestation of chronic RB loss. RB loss had a differential effect upon repair of the major DNA lesions induced by IR and UV. Whereas RB did not affect resolution of DNA double-strand breaks, RB-deficient cells exhibited accelerated repair of pyrimidine pyrimidone photoproducts (6-4 PP). In parallel, this repair was coupled with enhanced expression of specific factors and the behavior of proliferating cell nuclear antigen (PCNA) recruitment to replication and repair foci. Thus, RB loss and target gene deregulation hastens the repair of specific lesions distinct from its ubiquitous role in checkpoint abrogation.


Asunto(s)
Daño del ADN , Reparación del ADN , Radiación Ionizante , Proteína de Retinoblastoma/fisiología , Rayos Ultravioleta , Animales , Ciclo Celular , Células Cultivadas , Regulación hacia Abajo , Regulación de la Expresión Génica , Cinética , Ratones
10.
Methods Mol Biol ; 281: 3-16, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15220518

RESUMEN

Cell cycle checkpoints play a key role in maintaining genome stability by monitoring the order and integrity of cell division events. Checkpoints induced by DNA damage function to limit the propagation of potentially deleterious mutations. The retinoblastoma tumor suppressor (RB) is a critical effector of DNA damage checkpoint function by eliciting G1-phase cell cycle arrest following genotoxic stress. Here, we describe methodologies for evaluation of three facets of RB action in the DNA damage checkpoint response: (1) transcriptional repression of E2F-regulated genes (cyclin A reporter assay); (2) induction of cell cycle arrest (Brd-U incorporation assay); and (3) inhibition of DNA double-strand break accumulation (phosphorylated-histone H2A.X detection). Together, this combination of techniques allows the evaluation of RB action in the coordinated checkpoint response to DNA damage.


Asunto(s)
Proteínas de Ciclo Celular , Ciclo Celular , Daño del ADN/genética , Fibroblastos/metabolismo , Genes cdc/fisiología , Proteína de Retinoblastoma/fisiología , Animales , Antineoplásicos/farmacología , Bromodesoxiuridina , Células Cultivadas , Cisplatino/farmacología , Replicación del ADN/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción E2F , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Fibroblastos/citología , Genes Reporteros , Histonas/metabolismo , Ratones , Ratones Noqueados , Proteína de Retinoblastoma/deficiencia , Factores de Transcripción/metabolismo
11.
Nucleic Acids Res ; 32(1): 25-34, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-14704340

RESUMEN

Cell cycle checkpoints induced by DNA damage play an integral role in preservation of genomic stability by allowing cells to limit the propagation of deleterious mutations. The retinoblastoma tumor suppressor (RB) is crucial for the maintenance of the DNA damage checkpoint function because it elicits cell cycle arrest in response to a variety of genotoxic stresses. Although sporadic loss of RB is characteristic of most cancers and results in the bypass of the DNA damage checkpoint, the consequence of RB loss upon chemotherapeutic responsiveness has been largely uninvestigated. Here, we employed a conditional knockout approach to ablate RB in adult fibroblasts. This system enabled us to examine the DNA damage response of adult cells following acute RB deletion. Using this system, we demonstrated that loss of RB disrupted the DNA damage checkpoint elicited by either cisplatin or camptothecin exposure. Strikingly, this bypass was not associated with enhanced repair, but rather the accumulation of phosphorylated H2AX (gammaH2AX) foci, which indicate DNA double-strand breaks. The formation of gammaH2AX foci was due to ongoing replication following chemotherapeutic treatment in the RB-deficient cells. Additionally, peak gammaH2AX accumulation occurred in S-phase cells undergoing DNA replication in the presence of damage, and these gammaH2AX foci co-localized with replication foci. These results demonstrate that acute RB loss abrogates DNA damage-induced cell cycle arrest to induce gammaH2AX foci formation. Thus, secondary genetic lesions induced by RB loss have implications for the chemotherapeutic response and the development of genetic instability.


Asunto(s)
Daño del ADN/efectos de los fármacos , Replicación del ADN , Mutágenos/farmacología , Proteína de Retinoblastoma/metabolismo , Transducción de Señal , Animales , Camptotecina/farmacología , Ciclo Celular/efectos de los fármacos , Células Cultivadas , Cisplatino/farmacología , Replicación del ADN/efectos de los fármacos , Regulación hacia Abajo , Fibroblastos , Ratones , Proteína de Retinoblastoma/deficiencia , Proteína de Retinoblastoma/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...