Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nature ; 629(8011): 435-442, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38658751

RESUMEN

WRN helicase is a promising target for treatment of cancers with microsatellite instability (MSI) due to its essential role in resolving deleterious non-canonical DNA structures that accumulate in cells with faulty mismatch repair mechanisms1-5. Currently there are no approved drugs directly targeting human DNA or RNA helicases, in part owing to the challenging nature of developing potent and selective compounds to this class of proteins. Here we describe the chemoproteomics-enabled discovery of a clinical-stage, covalent allosteric inhibitor of WRN, VVD-133214. This compound selectively engages a cysteine (C727) located in a region of the helicase domain subject to interdomain movement during DNA unwinding. VVD-133214 binds WRN protein cooperatively with nucleotide and stabilizes compact conformations lacking the dynamic flexibility necessary for proper helicase function, resulting in widespread double-stranded DNA breaks, nuclear swelling and cell death in MSI-high (MSI-H), but not in microsatellite-stable, cells. The compound was well tolerated in mice and led to robust tumour regression in multiple MSI-H colorectal cancer cell lines and patient-derived xenograft models. Our work shows an allosteric approach for inhibition of WRN function that circumvents competition from an endogenous ATP cofactor in cancer cells, and designates VVD-133214 as a promising drug candidate for patients with MSI-H cancers.


Asunto(s)
Helicasa del Síndrome de Werner , Ensayos Antitumor por Modelo de Xenoinjerto , Humanos , Helicasa del Síndrome de Werner/metabolismo , Helicasa del Síndrome de Werner/química , Animales , Ratones , Regulación Alostérica/efectos de los fármacos , Línea Celular Tumoral , Femenino , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/enzimología , Proteómica , Roturas del ADN de Doble Cadena , Inestabilidad de Microsatélites , Descubrimiento de Drogas , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/química , Modelos Moleculares , Masculino , Cisteína/metabolismo , Cisteína/química
2.
Nat Commun ; 12(1): 710, 2021 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-33514714

RESUMEN

Antibody-based therapeutics have experienced a rapid growth in recent years and are now utilized in various modalities spanning from conventional antibodies, antibody-drug conjugates, bispecific antibodies to chimeric antigen receptor (CAR) T cells. Many next generation antibody therapeutics achieve enhanced potency but often increase the risk of adverse events. Antibody scaffolds capable of exhibiting inducible affinities could reduce the risk of adverse events by enabling a transient suspension of antibody activity. To demonstrate this, we develop conditionally activated, single-module CARs, in which tumor antigen recognition is directly modulated by an FDA-approved small molecule drug. The resulting CAR T cells demonstrate specific cytotoxicity of tumor cells comparable to that of traditional CARs, but the cytotoxicity is reversibly attenuated by the addition of the small molecule. The exogenous control of conditional CAR T cell activity allows continual modulation of therapeutic activity to improve the safety profile of CAR T cells across all disease indications.


Asunto(s)
Antígenos de Neoplasias/inmunología , Inmunoterapia Adoptiva/métodos , Metotrexato/administración & dosificación , Neoplasias/terapia , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/efectos de los fármacos , Animales , Línea Celular Tumoral , Terapia Combinada/métodos , Femenino , Células HEK293 , Humanos , Inmunoterapia Adoptiva/efectos adversos , Ratones , Neoplasias/inmunología , Cultivo Primario de Células , Receptores Quiméricos de Antígenos/inmunología , Anticuerpos de Dominio Único/inmunología , Anticuerpos de Dominio Único/metabolismo , Especificidad del Receptor de Antígeno de Linfocitos T/efectos de los fármacos , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/trasplante , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Cell Cycle ; 20(1): 65-80, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33356791

RESUMEN

Palbociclib, a selective CDK4/6 kinase inhibitor, is approved in combination with endocrine therapies for the treatment of advanced estrogen receptor positive (ER+) breast cancer. In pre-clinical cancer models, CDK4/6 inhibitors act primarily as cytostatic agents. In two commonly studied ER+ breast cancer cell lines (MCF7 and T47D), CDK4/6 inhibition drives G1-phase arrest and the acquisition of a senescent-like phenotype, both of which are reversible upon palbociclib withdrawal (incomplete senescence). Here we identify an ER+ breast cancer cell line, CAMA1, in which palbociclib treatment induces irreversible cell cycle arrest and senescence (complete senescence). In stark contrast to T47D and MCF7 cells, mTORC1 activity is not stably suppressed in CAMA1 cells during palbociclib treatment. Importantly, inhibition of mTORC1 signaling either by the mTORC1 inhibitor rapamycin or by knockdown of Raptor, a unique component of mTORC1, during palbociclib treatment of CAMA1 cells blocks the induction of complete senescence. These results indicate that sustained mTORC1 activity promotes complete senescence in ER+ breast cancer cells during CDK4/6 inhibitor-induced cell cycle arrest. Consistent with this mechanism, genetic depletion of TSC2, a negative regulator of mTORC1, in MCF7 cells resulted in sustained mTORC1 activity during palbociclib treatment and evoked a complete senescence response. These findings demonstrate that persistent mTORC1 signaling during palbociclib-induced G1 arrest is a potential liability for ER+ breast cancer cells, and suggest a strategy for novel drug combinations with palbociclib.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Puntos de Control del Ciclo Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Piperazinas/farmacología , Piridinas/farmacología , Receptores de Estrógenos/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Femenino , Humanos , Células MCF-7 , Ratones , Transducción de Señal/efectos de los fármacos
4.
Oncoimmunology ; 9(1): 1800162, 2020 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-32923161

RESUMEN

Macroautophagy (autophagy) is an essential cellular catabolic process required for survival under conditions of starvation. The role of autophagy in cancer is complex, context-dependent and at times contradictory, as it has been shown to inhibit, promote or be dispensable for tumor progression. In this study, we evaluated the contribution of the immune system to the reliance of tumors on autophagy by depleting autophagy-related 7 (ATG7) in murine tumor cells and grafting into immunocompetent versus immunodeficient hosts. Although loss of ATG7 did not affect tumor growth in vitro or in immunodeficient mice, our studies revealed that cancer cell reliance on autophagy was influenced by anti-tumor immune responses, including those mediated by CD8+ T cells. Furthermore, we provide insights into possible mechanisms by which autophagy disruption can enhance anti-tumor immune responses and suggest that autophagy disruption may further benefit patients with immunoreactive tumors.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Animales , Autofagia , Proteína 7 Relacionada con la Autofagia/genética , Humanos , Ratones
5.
Science ; 367(6474): 146-147, 2020 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-31919209
6.
Proc Natl Acad Sci U S A ; 116(19): 9533-9542, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-31019077

RESUMEN

T cell-invigorating cancer immunotherapies have near-curative potential. However, their clinical benefit is currently limited, as only a fraction of patients respond, suggesting that these regimens may benefit from combination with tumor-targeting treatments. As oncogenic progression is accompanied by alterations in metabolic pathways, tumors often become heavily reliant on antioxidant machinery and may be susceptible to increases in oxidative stress. The cystine-glutamate antiporter xCT is frequently overexpressed in cancer and fuels the production of the antioxidant glutathione; thus, tumors prone to redox stress may be selectively vulnerable to xCT disruption. However, systemic inhibition of xCT may compromise antitumor immunity, as xCT is implicated in supporting antigen-induced T cell proliferation. Therefore, we utilized immune-competent murine tumor models to investigate whether cancer cell expression of xCT was required for tumor growth in vivo and if deletion of host xCT impacted antitumor immune responses. Deletion of xCT in tumor cells led to defective cystine uptake, accumulation of reactive oxygen species, and impaired tumor growth, supporting a cancer cell-autonomous role for xCT. In contrast, we observed that, although T cell proliferation in culture was exquisitely dependent on xCT expression, xCT was dispensable for T cell proliferation in vivo and for the generation of primary and memory immune responses to tumors. These findings prompted the combination of tumor cell xCT deletion with the immunotherapeutic agent anti-CTLA-4, which dramatically increased the frequency and durability of antitumor responses. Together, these results identify a metabolic vulnerability specific to tumors and demonstrate that xCT disruption can expand the efficacy of anticancer immunotherapies.


Asunto(s)
Sistema de Transporte de Aminoácidos y+/deficiencia , Células Presentadoras de Antígenos/inmunología , Proliferación Celular , Memoria Inmunológica , Neoplasias Experimentales/inmunología , Linfocitos T/inmunología , Sistema de Transporte de Aminoácidos y+/inmunología , Animales , Células Presentadoras de Antígenos/patología , Línea Celular , Eliminación de Gen , Glutatión/genética , Glutatión/inmunología , Inmunoterapia , Ratones , Ratones Noqueados , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , Linfocitos T/patología
7.
Cell ; 170(4): 605-635, 2017 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-28802037

RESUMEN

Phosphoinositide 3-kinase (PI3K) activity is stimulated by diverse oncogenes and growth factor receptors, and elevated PI3K signaling is considered a hallmark of cancer. Many PI3K pathway-targeted therapies have been tested in oncology trials, resulting in regulatory approval of one isoform-selective inhibitor (idelalisib) for treatment of certain blood cancers and a variety of other agents at different stages of development. In parallel to PI3K research by cancer biologists, investigations in other fields have uncovered exciting and often unpredicted roles for PI3K catalytic and regulatory subunits in normal cell function and in disease. Many of these functions impinge upon oncology by influencing the efficacy and toxicity of PI3K-targeted therapies. Here we provide a perspective on the roles of class I PI3Ks in the regulation of cellular metabolism and in immune system functions, two topics closely intertwined with cancer biology. We also discuss recent progress developing PI3K-targeted therapies for treatment of cancer and other diseases.


Asunto(s)
Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Animales , Fenómenos Fisiológicos Celulares , Humanos , Sistema Inmunológico/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología
8.
Cell Rep ; 19(13): 2665-2680, 2017 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-28658616

RESUMEN

Pharmacologic agents that interfere with nucleotide metabolism constitute an important class of anticancer agents. Recent studies have demonstrated that mTOR complex 1 (mTORC1) inhibitors suppress de novo biosynthesis of pyrimidine and purine nucleotides. Here, we demonstrate that mTORC1 itself is suppressed by drugs that reduce intracellular purine nucleotide pools. Cellular treatment with AG2037, an inhibitor of the purine biosynthetic enzyme GARFT, profoundly inhibits mTORC1 activity via a reduction in the level of GTP-bound Rheb, an obligate upstream activator of mTORC1, because of a reduction in intracellular guanine nucleotides. AG2037 treatment provokes both mTORC1 inhibition and robust tumor growth suppression in mice bearing non-small-cell lung cancer (NSCLC) xenografts. These results indicate that alterations in purine nucleotide availability affect mTORC1 activity and suggest that inhibition of mTORC1 contributes to the therapeutic effects of purine biosynthesis inhibitors.


Asunto(s)
Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Nucleótidos de Purina/metabolismo , Proteína Homóloga de Ras Enriquecida en el Cerebro/metabolismo , Células A549 , Animales , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Línea Celular Tumoral , Femenino , Xenoinjertos , Humanos , Neoplasias Pulmonares/metabolismo , Ratones , Nucleótidos de Purina/biosíntesis
9.
PLoS One ; 12(3): e0173771, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28301521

RESUMEN

Lysosomes are acidic organelles essential for degradation and cellular homoeostasis and recently lysosomes have been shown as signaling hub to respond to the intra and extracellular changes (e.g. amino acid availability). Compounds including pharmaceutical drugs that are basic and lipophilic will become sequestered inside lysosomes (lysosomotropic). How cells respond to the lysosomal stress associated with lysosomotropism is not well characterized. Our goal is to assess the lysosomal changes and identify the signaling pathways that involve in the lysosomal changes. Eight chemically diverse lysosomotropic drugs from different therapeutic areas were subjected to the evaluation using the human adult retinal pigmented epithelium cell line, ARPE-19. All lysosomotropic drugs tested triggered lysosomal activation demonstrated by increased lysosotracker red (LTR) and lysosensor green staining, increased cathepsin activity, and increased LAMP2 staining. However, tested lysosomotropic drugs also prompted lysosomal dysfunction exemplified by intracellular and extracellular substrate accumulation including phospholipid, SQSTM1/p62, GAPDH (Glyceraldehyde 3-phosphate dehydrogenase) and opsin. Lysosomal activation observed was likely attributed to lysosomal dysfunction, leading to compensatory responses including nuclear translocation of transcriptional factors TFEB, TFE3 and MITF. The adaptive changes are protective to the cells under lysosomal stress. Mechanistic studies implicate calcium and mTORC1 modulation involvement in the adaptive changes. These results indicate that lysosomotropic compounds could evoke a compensatory lysosomal biogenic response but with the ultimate consequence of lysosomal functional impairment. This work also highlights a pathway of response to lysosomal stress and evidences the role of TFEB, TFE3 and MITF in the stress response.


Asunto(s)
Adaptación Fisiológica , Lisosomas/efectos de los fármacos , Línea Celular , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Lisosomas/enzimología , Lisosomas/metabolismo , Lisosomas/fisiología , Opsinas/metabolismo , Epitelio Pigmentado de la Retina/citología , Proteína Sequestosoma-1/metabolismo
10.
Mol Cell ; 65(6): 959-960, 2017 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-28306509

RESUMEN

The PARK2 gene encodes an ubiquitin E3 ligase that is involved in mitochondrial homeostasis and linked to Parkinson's disease. In this issue, Gupta et al. (2017) demonstrate that PARK2 expression is frequently reduced in human cancers and that this alteration leads to dysregulated PI3K signaling.


Asunto(s)
Fosfatidilinositol 3-Quinasas , Ubiquitina-Proteína Ligasas/genética , Humanos , Mitocondrias , Neoplasias , Enfermedad de Parkinson/genética , Ubiquitina
11.
Sci Signal ; 9(430): fs10, 2016 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-27245610

RESUMEN

The mTORC1 [mammalian (or mechanistic) target of rapamycin complex 1] inhibitor rapamycin is a potent inhibitor of T and B lymphocyte growth and proliferation. In this issue of Science Signaling, So et al found that the isoform of 4E-BP (4E-BP2) in lymphocytes that regulates messenger RNA translation is more sensitive to the effects of rapamycin than is the 4E-BP isoform found in nonlymphoid cells. Furthermore, inhibition of 4E-BP2 blocks both the growth and proliferation of lymphocytes.


Asunto(s)
Factor 4E Eucariótico de Iniciación , Sirolimus , Animales , Proliferación Celular/efectos de los fármacos , Linfocitos , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos , Serina-Treonina Quinasas TOR
12.
Cell Metab ; 23(3): 397-8, 2016 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-26959180

RESUMEN

The mechanistic target of rapamycin complex 1 (mTORC1) coordinates nutrient availability with cell growth. Recent reports by Sabatini and coworkers (Saxton et al., 2016; Wolfson et al., 2016) characterize a cytoplasmic amino acid receptor that couples the binding of leucine to the activation of mTORC1.

13.
Proc Natl Acad Sci U S A ; 113(1): 182-7, 2016 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-26677873

RESUMEN

Macroautophagy is a key stress-response pathway that can suppress or promote tumorigenesis depending on the cellular context. Notably, Kirsten rat sarcoma (KRAS)-driven tumors have been reported to rely on macroautophagy for growth and survival, suggesting a potential therapeutic approach of using autophagy inhibitors based on genetic stratification. In this study, we evaluated whether KRAS mutation status can predict the efficacy to macroautophagy inhibition. By profiling 47 cell lines with pharmacological and genetic loss-of-function tools, we were unable to confirm that KRAS-driven tumor lines require macroautophagy for growth. Deletion of autophagy-related 7 (ATG7) by genome editing completely blocked macroautophagy in several tumor lines with oncogenic mutations in KRAS but did not inhibit cell proliferation in vitro or tumorigenesis in vivo. Furthermore, ATG7 knockout did not sensitize cells to irradiation or to several anticancer agents tested. Interestingly, ATG7-deficient and -proficient cells were equally sensitive to the antiproliferative effect of chloroquine, a lysosomotropic agent often used as a pharmacological tool to evaluate the response to macroautophagy inhibition. Moreover, both cell types manifested synergistic growth inhibition when treated with chloroquine plus the tyrosine kinase inhibitors erlotinib or sunitinib, suggesting that the antiproliferative effects of chloroquine are independent of its suppressive actions on autophagy.


Asunto(s)
Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Cloroquina/farmacología , Resistencia a Antineoplásicos/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Autofagia/genética , Proteína 7 Relacionada con la Autofagia , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Clorhidrato de Erlotinib/farmacología , Técnicas de Inactivación de Genes , Humanos , Indoles/farmacología , Mutación , Inhibidores de Proteínas Quinasas/farmacología , Pirroles/farmacología , Tolerancia a Radiación/genética , Sunitinib , Enzimas Activadoras de Ubiquitina/genética
14.
Clin Cancer Res ; 21(13): 2905-10, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25941111

RESUMEN

Cancer cells bypass normal controls over mitotic cell-cycle progression to achieve a deregulated state of proliferation. The retinoblastoma tumor suppressor protein (pRb) governs a key cell-cycle checkpoint that normally prevents G1-phase cells from entering S-phase in the absence of appropriate mitogenic signals. Cancer cells frequently overcome pRb-dependent growth suppression via constitutive phosphorylation and inactivation of pRb function by cyclin-dependent kinase (CDK) 4 or CDK6 partnered with D-type cyclins. Three selective CDK4/6 inhibitors, palbociclib (Ibrance; Pfizer), ribociclib (Novartis), and abemaciclib (Lilly), are in various stages of development in a variety of pRb-positive tumor types, including breast cancer, melanoma, liposarcoma, and non-small cell lung cancer. The emerging, positive clinical data obtained to date finally validate the two decades-old hypothesis that the cyclin D-CDK4/6 pathway is a rational target for cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Animales , Antineoplásicos/uso terapéutico , Ciclina D/antagonistas & inhibidores , Ciclina D/metabolismo , Quinasa 4 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 6 Dependiente de la Ciclina/metabolismo , Humanos , Terapia Molecular Dirigida , Neoplasias/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Transducción de Señal
16.
Nat Commun ; 5: 4900, 2014 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-25254627

RESUMEN

The non-essential amino acid, glutamine, exerts pleiotropic effects on cell metabolism, signalling and stress resistance. Here we demonstrate that short-term glutamine restriction triggers an endoplasmic reticulum (ER) stress response that leads to production of the pro-inflammatory chemokine, interleukin-8 (IL-8). Glutamine deprivation-induced ER stress triggers colocalization of autophagosomes, lysosomes and the Golgi into a subcellular structure whose integrity is essential for IL-8 secretion. The stimulatory effect of glutamine restriction on IL-8 production is attributable to depletion of tricarboxylic acid cycle intermediates. The protein kinase, mTOR, is also colocalized with the lysosomal membrane clusters induced by glutamine deprivation, and inhibition of mTORC1 activity abolishes both endomembrane reorganization and IL-8 secretion. Activated mTORC1 elicits IL8 gene expression via the activation of an IRE1-JNK signalling cascade. Treatment of cells with a glutaminase inhibitor phenocopies glutamine restriction, suggesting that these results will be relevant to the clinical development of glutamine metabolism inhibitors as anticancer agents.


Asunto(s)
Interleucina-8/metabolismo , MAP Quinasa Quinasa 4/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Línea Celular Tumoral , Ciclo del Ácido Cítrico , Estrés del Retículo Endoplásmico , Glutamina , Humanos , Lisosomas/metabolismo , MAP Quinasa Quinasa 4/genética , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Serina-Treonina Quinasas TOR/genética
17.
Cell Metab ; 20(1): 4-6, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24988455

RESUMEN

Obesity promotes chronic activation of mTORC1 and is a known risk factor for hepatic injury, inflammation, and carcinogenesis. In this issue, Umemura et al. (2014) demonstrate that a persistent reduction in hepatic mTORC1 activity also promotes cell damage and inflammation and sensitizes the liver to cancer development.


Asunto(s)
Inflamación , Hígado/efectos de los fármacos , Complejos Multiproteicos/metabolismo , Sirolimus/toxicidad , Serina-Treonina Quinasas TOR/metabolismo , Animales , Humanos , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina
18.
Cancer Discov ; 4(5): 513-5, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24795009

RESUMEN

The identification of genetic lesions that affect tumor sensitivity to targeted therapies is a major objective of precision medicine. Two reports in this issue combine tumor genome analyses with functional characterization to uncover activating mutations in MTOR that confer sensitivity to a clinically used mTOR inhibitor.


Asunto(s)
Neoplasias/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Serina-Treonina Quinasas TOR/genética , Resistencia a Antineoplásicos , Humanos , Terapia Molecular Dirigida/métodos , Mutación , Neoplasias/genética , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo
19.
Cell ; 154(6): 1184-6, 2013 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-24034240

RESUMEN

Autophagy is a cell-autonomous, catabolic process that plays context-dependent roles in tumor growth and progression. Wei et al. report that EGFR signaling promotes tumor growth through phosphorylation and functional inactivation of Beclin 1 and the consequent suppression of autophagy.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Autofagia , Resistencia a Antineoplásicos , Receptores ErbB/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Beclina-1 , Humanos
20.
Nat Prod Rep ; 30(5): 625-39, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23525375

RESUMEN

The Antibody Drug Conjugate (ADC) is a therapeutic modality consisting of a monoclonal antibody attached to a cytotoxic, small-molecule payload. The antibody portion of the ADC serves as a transport vehicle that recognizes and binds to a protein antigen expressed in tumor tissues. The localized delivery and release of the payload within or near malignant cells allows for targeted delivery of a potent cytotoxic agent to diseased tissue, while reducing damage to antigen-negative, normal tissues. Recent years have witnessed an explosive increase in ADC-based therapies, due mainly to clinical reports of activity in both hematologic and epithelial cancers. Accompanying this upsurge in ADC development is a renewed interest in natural product cytotoxins, which are typically highly potent cell-killing agents, but suffer from poor drug-like properties and narrow safety margins when systemically administered as conventional chemotherapeutics. In this review, we discuss recent advances related to the construction of ADCs, the optimization of ADC safety and efficacy, and the increasingly pivotal roles of natural product payloads in the current and future landscape of ADC therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Productos Biológicos/uso terapéutico , Inmunoconjugados/uso terapéutico , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/farmacología , Productos Biológicos/farmacología , Humanos , Inmunoconjugados/farmacología , Estructura Molecular , Neoplasias/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...