Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Proc Natl Acad Sci U S A ; 109(40): 16101-6, 2012 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-22988081

RESUMEN

Antibody-drug conjugates (ADCs) allow selective targeting of cytotoxic drugs to cancer cells presenting tumor-associated surface markers, thereby minimizing systemic toxicity. Traditionally, the drug is conjugated nonselectively to cysteine or lysine residues in the antibody. However, these strategies often lead to heterogeneous products, which make optimization of the biological, physical, and pharmacological properties of an ADC challenging. Here we demonstrate the use of genetically encoded unnatural amino acids with orthogonal chemical reactivity to synthesize homogeneous ADCs with precise control of conjugation site and stoichiometry. p-Acetylphenylalanine was site-specifically incorporated into an anti-Her2 antibody Fab fragment and full-length IgG in Escherichia coli and mammalian cells, respectively. The mutant protein was selectively and efficiently conjugated to an auristatin derivative through a stable oxime linkage. The resulting conjugates demonstrated excellent pharmacokinetics, potent in vitro cytotoxic activity against Her2(+) cancer cells, and complete tumor regression in rodent xenograft treatment models. The synthesis and characterization of homogeneous ADCs with medicinal chemistry-like control over macromolecular structure should facilitate the optimization of ADCs for a host of therapeutic uses.


Asunto(s)
Aminoácidos/química , Anticuerpos Monoclonales Humanizados/química , Neoplasias de la Mama/tratamiento farmacológico , Inmunoconjugados/química , Ingeniería de Proteínas/métodos , Aminobenzoatos/química , Animales , Línea Celular Tumoral , Descubrimiento de Drogas/métodos , Ensayo de Inmunoadsorción Enzimática , Escherichia coli , Femenino , Humanos , Inmunoconjugados/farmacocinética , Inmunoconjugados/uso terapéutico , Inmunoglobulina G/química , Ratones , Ratones SCID , Oligopéptidos/química , Receptor ErbB-2/química , Receptor ErbB-2/inmunología , Trastuzumab
2.
Angew Chem Int Ed Engl ; 53(44): 11863-7, 2014 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-25213874

RESUMEN

A chemically defined anti-CXCR4-auristatin antibody-drug conjugate (ADC) was synthesized that selectively eliminates tumor cells overexpressing the CXCR4 receptor. The unnatural amino acid p-acetylphenylalanine (pAcF) was site-specifically incorporated into an anti-CXCR4 immunoglobulin G (IgG) and conjugated to an auristatin through a stable, non-cleavable oxime linkage to afford a chemically homogeneous ADC. The full-length anti-CXCR4 ADC was selectively cytotoxic to CXCR4(+) cancer cells in vitro (half maximal effective concentration (EC50 )≈80-100 pM). Moreover, the anti-CXCR4 ADC eliminated pulmonary lesions from human osteosarcoma cells in a lung-seeding tumor model in mice. No significant overt toxicity was observed but there was a modest decrease in the bone-marrow-derived CXCR4(+) cell population. Because CXCR4 is highly expressed in a majority of metastatic cancers, a CXCR4-auristatin ADC may be useful for the treatment of a variety of metastatic malignancies.


Asunto(s)
Anticuerpos Monoclonales/química , Antineoplásicos/química , Inmunoconjugados/química , Inmunoterapia/métodos , Receptores CXCR4/química , Línea Celular Tumoral , Humanos
3.
Proc Natl Acad Sci U S A ; 106(26): 10666-71, 2009 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-19541645

RESUMEN

The incidence of brain metastasis is rising and poses a severe clinical problem, as we lack effective therapies and knowledge of mechanisms that control metastatic growth in the brain. Here we demonstrate a crucial role for high-affinity tumor cell integrin alpha(v)beta(3) in brain metastatic growth and recruitment of blood vessels. Although alpha(v)beta(3) is frequently up-regulated in primary brain tumors and metastatic lesions of brain homing cancers, we show that it is the alpha(v)beta(3) activation state that is critical for brain lesion growth. Activated, but not non-activated, tumor cell alpha(v)beta(3) supports efficient brain metastatic growth through continuous up-regulation of vascular endothelial growth factor (VEGF) protein under normoxic conditions. In metastatic brain lesions carrying activated alpha(v)beta(3), VEGF expression is controlled at the post-transcriptional level and involves phosphorylation and inhibition of translational respressor 4E-binding protein (4E-BP1). In contrast, tumor cells with non-activated alpha(v)beta(3) depend on hypoxia for VEGF induction, resulting in reduced angiogenesis, tumor cell apoptosis, and inefficient intracranial growth. Importantly, the microenvironment critically influences the effects that activated tumor cell alpha(v)beta(3) exerts on tumor cell growth. Although it strongly promoted intracranial growth, the activation state of the receptor did not influence tumor growth in the mammary fat pad as a primary site. Thus, we identified a mechanism by which metastatic cells thrive in the brain microenvironment and use the high-affinity form of an adhesion receptor to grow and secure host support for proliferation. Targeting this molecular mechanism could prove valuable for the inhibition of brain metastasis.


Asunto(s)
Neoplasias Encefálicas/secundario , Integrina alfaVbeta3/metabolismo , Neoplasias Mamarias Experimentales/patología , Neovascularización Patológica/patología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Apoptosis , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Hipoxia , Etiquetado Corte-Fin in Situ , Integrina alfaVbeta3/genética , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones SCID , Mutación , Trasplante de Neoplasias , Fosfoproteínas/metabolismo , Fosforilación , Procesamiento Proteico-Postraduccional , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trasplante Heterólogo , Carga Tumoral , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
4.
Am J Pathol ; 176(6): 2958-71, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20382702

RESUMEN

Brain metastases are difficult to treat and mostly develop late during progressive metastatic disease. Patients at risk would benefit from the development of prevention and improved treatments. This requires knowledge of the initial events that lead to brain metastasis. The present study reveals cellular events during the initiation of brain metastasis by breast cancer cells and documents the earliest host responses to incoming cancer cells after carotid artery injection in immunodeficient and immunocompetent mouse models. Our findings capture and characterize heterogeneous astrocytic and microglial reactions to the arrest and extravasation of cancer cells in the brain, showing immediate and drastic changes in the brain microenvironment on arrival of individual cancer cells. We identified reactive astrocytes as the most active host cell population that immediately localizes to individual invading tumor cells and continuously associates with growing metastatic lesions. Up-regulation of matrix metalloproteinase-9 associated with astrocyte activation in the immediate vicinity of extravasating cancer cells might support their progression. Early involvement of different host cell types indicates environmental clues that might codetermine whether a single cancer cell progresses to macrometastasis or remains dormant. Thus, information on the initial interplay between brain homing tumor cells and reactive host cells may help develop strategies for prevention and treatment of symptomatic breast cancer brain metastases.


Asunto(s)
Neoplasias Encefálicas/patología , Encéfalo/metabolismo , Encéfalo/patología , Neoplasias de la Mama/secundario , Animales , Astrocitos/metabolismo , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patología , Encéfalo/irrigación sanguínea , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/metabolismo , Neoplasias de la Mama/irrigación sanguínea , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Circulación Cerebrovascular , Progresión de la Enfermedad , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Microglía/metabolismo , Metástasis de la Neoplasia
5.
Bioconjug Chem ; 22(8): 1535-44, 2011 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-21774545

RESUMEN

Integrins αvß3 and αvß6 are highly expressed on tumor cells and/or by the tumor vasculature of many human cancers, and represent promising targets for anticancer therapy. Novel chemically programmed antibodies (cpAbs) targeting these integrins were prepared using the catalytic aldolase Antibody (Ab) programming strategy. The effects of the cpAbs on cellular functions related to tumor progression were examined in vitro using tumor cell lines and their cognate integrin ligands, fibronectin and osteopontin. The inhibitory functions of the conjugates and their specificity were examined based on interference with cell-cell and cell-ligand interactions related to tumor progression. Cell binding analyses of the anti-integrin cpAbs revealed high affinity for tumor cells that overexpressed αvß3 and αvß6 integrins, and weak interactions with αvß1 and αvß8 integrins, in vitro. Functional analyses demonstrated that the cpAbs strongly inhibited cell-cell interactions through osteopontin binding, and they had little or no immediate effects on cell viability and proliferation. On the basis of these characteristics, the cpAbs are likely to have a broad range of activities in vivo, as they can target and antagonize one or multiple αv integrins expressed on tumors and tumor vasculatures. Presumably, these conjugates may inhibit the establishment of metastastatic tumors in distant organs through interfering with cell adhesion more effectively than antibodies or compounds targeting one integrin only. These anti-integrin cpAbs may also provide useful reagents to study combined effect of multiple αv integrins on cellular functions in vitro, on pathologies, including tumor angiogenesis, fibrosis, and epithelial cancers, in vivo.


Asunto(s)
Anticuerpos/uso terapéutico , Antígenos de Neoplasias/inmunología , Antineoplásicos , Integrina alfaV/inmunología , Neoplasias/inmunología , Anticuerpos/inmunología , Comunicación Celular , Línea Celular Tumoral , Fructosa-Bifosfato Aldolasa , Humanos , Inmunoconjugados/uso terapéutico , Integrina alfaVbeta3/inmunología , Integrinas/inmunología , Neoplasias/tratamiento farmacológico
6.
Bioorg Med Chem Lett ; 19(14): 3821-4, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19428247

RESUMEN

Chemical programming of nine murine antibodies with catalytic aldolase activity was examined using compounds, equipped with diketone or pro-vinyl ketone linkers that inhibit integrin adhesion receptor functions. The results showed that most Abs were programmed using the diketone compounds in a manner similar to previously reported catalytic antibody 38C2. On the other hand, only those antibodies, which catalyzed the retro aldol reaction of the pro-vinyl ketone linkers efficiently, were programmed. Conjugated to integrin targeting compounds, at least three new antibodies, including 84G3, 85H6, and 90G8, exhibited high specific binding to human tumor cells expressing integrin alpha(v)beta(3.).


Asunto(s)
Anticuerpos Catalíticos/química , Fructosa-Bifosfato Aldolasa/química , Fragmentos Fab de Inmunoglobulinas/química , Anticuerpos Catalíticos/inmunología , Anticuerpos Catalíticos/metabolismo , Línea Celular Tumoral , Fructosa-Bifosfato Aldolasa/inmunología , Fructosa-Bifosfato Aldolasa/metabolismo , Humanos , Fragmentos Fab de Inmunoglobulinas/inmunología , Fragmentos Fab de Inmunoglobulinas/metabolismo , Integrina alfaVbeta3/metabolismo , Cetonas/química
7.
Methods Mol Biol ; 568: 249-59, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19582432

RESUMEN

Despite advances for the treatment of cancer, the prognosis for patients suffering from malignant brain tumors remains dismal. High-grade neoplasms, such as gliomas, are highly invasive and spawn widely disseminated microsatellites that have limited the efficacy of surgical and adjunctive therapies. The cancer stem cell hypothesis suggests that conventional chemotherapeutic treatments kill differentiated and differentiating cells which often form the bulk of the tumor. One major concern is that the cells which give rise to the tumor, the cancer stem cells, remain untouched and may be responsible for a relapse of the disease. Therefore, an adjunctive therapy to current cancer treatment is critical for the survivability of patients suffering from brain tumors. We have successfully engineered tumor-tropic neural stem cells to deliver antineoplastic gene products directly to the tumor-producing cells. This potential therapeutic strategy may safely eradicate tumor-producing cells in the brain while minimizing damage to normal, healthy cells.


Asunto(s)
Neoplasias Encefálicas/terapia , Técnicas de Cultivo de Célula/métodos , Terapia Genética/métodos , Neuronas/citología , Células Madre/citología , Células Madre/metabolismo , Línea Celular , Congelación , Humanos , Inyecciones , Lentivirus/genética , Trasplante de Células Madre , Transducción Genética
8.
Cancer Res ; 67(4): 1472-86, 2007 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17308085

RESUMEN

Brain metastases are among the most feared complications in breast cancer, as no therapy exists that prevents or eliminates breast cancer spreading to the brain. New therapeutic strategies depend on specific knowledge of tumor cell properties that allow breast cancer cell growth within the brain tissue. To provide information in this direction, we established a human breast cancer cell model for brain metastasis based on circulating tumor cells from a breast cancer patient and variants of these cells derived from bone or brain lesions in immunodeficient mice. The brain-derived cells showed an increased potential for brain metastasis in vivo and exhibited a unique protein expression profile identified by large-scale proteomic analysis. This protein profile is consistent with either a selection of predisposed cells or bioenergetic adaptation of the tumor cells to the unique energy metabolism of the brain. Increased expression of enzymes involved in glycolysis, tricarboxylic acid cycle, and oxidative phosphorylation pathways suggests that the brain metastatic cells derive energy from glucose oxidation. The cells further showed enhanced activation of the pentose phosphate pathway and the glutathione system, which can minimize production of reactive oxygen species resulting from an enhanced oxidative metabolism. These changes promoted resistance of brain metastatic cells to drugs that affect the cellular redox balance. Importantly, the metabolic alterations are associated with strongly enhanced tumor cell survival and proliferation in the brain microenvironment. Thus, our data support the hypothesis that predisposition or adaptation of the tumor cell energy metabolism is a key element in breast cancer brain metastasis, and raise the possibility of targeting the functional differentiation in breast cancer brain lesions as a novel therapeutic strategy.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/secundario , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Animales , Procesos de Crecimiento Celular/fisiología , Ciclo del Ácido Cítrico , Modelos Animales de Enfermedad , Metabolismo Energético , Femenino , Glutatión/metabolismo , Glucólisis , Humanos , Ratones , Ratones SCID , Mitocondrias/metabolismo , Oxidación-Reducción , Consumo de Oxígeno , Vía de Pentosa Fosfato , Proteómica
9.
Clin Cancer Res ; 13(6): 1656-62, 2007 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-17363518

RESUMEN

Metastasis to the brain is prevalent in solid tumors and lymphomas, and is associated with shortened survival. The brain is regarded as a sanctuary site for metastatic tumor cells where they exist partially protected from drugs by the blood-tumor barrier. Model systems for brain metastasis have been developed and are now yielding mechanistic insights into the roles of angiogenesis, energy metabolism, the Her-2 and Stat3 signaling pathways, and dormancy. Specific, new approaches to combat brain metastatic disease are needed.


Asunto(s)
Neoplasias Encefálicas/etiología , Neoplasias Encefálicas/secundario , Animales , Encéfalo/irrigación sanguínea , Neoplasias Encefálicas/irrigación sanguínea , Permeabilidad de la Membrana Celular/fisiología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Metabolismo Energético/fisiología , Humanos , Melanoma Experimental/patología , Modelos Biológicos , Neovascularización Patológica/patología , Receptor ErbB-2/fisiología , Factor de Transcripción STAT3/fisiología
10.
FASEB J ; 19(9): 1123-4, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15857880

RESUMEN

Maspin, a member of the serine protease inhibitor (serpin) family, is a tumor suppressor in breast and prostate cancer. To address molecular mechanisms underlying maspin's activity, we restored its expression in invasive carcinoma cells and analyzed the resulting changes by shotgun proteomics. Using a mass spectrometry-based multidimensional proteomic method, we observed changes to the expression of approximately 27% of the detectable proteome. In particular, we noted changes to the expression of proteins that regulate cytoskeletal architecture, cell death, and protein turnover. In each case, changes in protein expression were accompanied by measurable changes in tumor cell phenotype. Thus, maspin-expressing cells exhibit a more prominent actin cytoskeleton, a reduced invasive capacity, an increased rate of spontaneous apoptosis, and an altered proteasome function. These observations reveal for the first time the far reaching effects of maspin on multiple protein networks and a new hypothesis of maspin function based on the regulation of proteasome function.


Asunto(s)
Neoplasias de la Mama/patología , Genes Supresores de Tumor/fisiología , Metástasis de la Neoplasia/genética , Proteoma , Serpinas/fisiología , Apoptosis , Neoplasias de la Mama/genética , Línea Celular Tumoral , Femenino , Humanos , Metástasis de la Neoplasia/prevención & control , Complejo de la Endopetidasa Proteasomal/fisiología , Serpinas/genética , Transfección , Ubiquitina/metabolismo
11.
Chem Biol ; 11(7): 897-906, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15271348

RESUMEN

Selective antitumor chemotherapy can be achieved by using antibody-drug conjugates that recognize surface proteins upregulated in cancer cells. One such receptor is integrin alpha3beta1, which is overexpressed on malignant melanoma, prostate carcinoma, and glioma cells. We previously identified a human single-chain Fv antibody (scFv), denoted Pan10, specific for integrin alpha3beta1 that is internalized by human pancreatic cancer cells. Herein, we describe the chemical introduction of reactive thiol groups onto Pan10, the specific conjugation of the modified scFv to maleimide-derivatized analogs of the potent cytotoxic agent duocarmycin SA, and the properties of the resultant conjugates. Our findings provide evidence that Pan10-drug conjugates maintain the internalizing capacity of the parent scFv and are cytotoxic at nanomolar concentrations. Our Pan10-drug conjugates may be promising candidates for targeted chemotherapy of malignant diseases associated with overexpression of integrin alpha3beta1.


Asunto(s)
Anticuerpos/inmunología , Antineoplásicos/administración & dosificación , Endocitosis , Integrina alfa3beta1/inmunología , Antineoplásicos/farmacocinética , Secuencia de Bases , Línea Celular , Cartilla de ADN , Citometría de Flujo , Humanos , Microscopía Confocal/métodos , Proteínas Recombinantes/inmunología , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
12.
Clin Exp Metastasis ; 20(3): 203-13, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-12741679

RESUMEN

Integrins are a family of cell adhesion receptors that support and modulate a variety of cellular functions that are required for tumor metastasis. Integrins expressed by tumor cells and host cells can directly contribute to the control and progress of metastatic dissemination. During tumor development, changes in integrin expression, intracellular control of integrin functions and signals perceived from integrin ligand binding impact upon the ability of tumor cells to interact with their environment and enable metastatic cells to convert from a sessile, stationary to a migratory and invasive phenotype. Integrins are involved in each step of the metastatic cascade and affect tumor cell survival and interaction with changing environments in transit from the primary tumor to distant target organs.


Asunto(s)
Moléculas de Adhesión Celular/fisiología , Integrinas/fisiología , Metástasis de la Neoplasia , Animales , Humanos
13.
Clin Exp Metastasis ; 19(5): 427-36, 2002.
Artículo en Inglés | MEDLINE | ID: mdl-12198771

RESUMEN

Early metastasis is the primary cause of death in melanoma patients. The adhesion receptor integrin alpha v beta 3 contributes to tumor cell functions that are potentially involved in melanoma growth and metastasis. We tested whether integrin alpha v beta 3 supports metastasis of human melanoma cells when injected into the bloodstream of immune deficient mice. Comparing variants of the same melanoma cell type that expressed either alpha v beta 3, alpha IIb beta 3 or no beta 3 integrin, we found that only alpha v beta 3 strongly supported metastasis. Inhibition of tumor cell alpha v beta 3 function reduced melanoma metastasis significantly and prolonged animal survival. To understand mechanisms that allow alpha v beta 3, but not alpha IIb beta 3 to support melanoma metastasis, we analyzed proteolytic and migratory activities of the melanoma cell variants. Melanoma cells expressing alpha v beta 3, but not those expressing alpha IIb beta 3 or no beta 3 integrin, produced the active form of metalloproteinase MMP-2 and expressed elevated mRNA levels of MT1-MMP and TIMP-2. This indicates an association between alpha v beta 3 expression and protease processing. Furthermore, alpha v beta 3 expression was required for efficient melanoma cell migration toward the matrix proteins fibronectin and vitronectin. The results suggest that expression of integrin alpha v beta 3 promotes the metastatic phenotype in human melanoma by supporting specific adhesive, invasive and migratory properties of the tumor cells and that the related integrin alpha IIb beta 3 cannot substitute for alpha v beta 3 in this respect.


Asunto(s)
Neoplasias Pulmonares/secundario , Melanoma Experimental/secundario , Melanoma/patología , Proteínas de Neoplasias/fisiología , Células Neoplásicas Circulantes , Receptores de Vitronectina/fisiología , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Matriz Extracelular/metabolismo , Femenino , Fibronectinas/metabolismo , Humanos , Inyecciones Intravenosas , Metaloproteinasa 14 de la Matriz , Metaloproteinasa 2 de la Matriz/biosíntesis , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasas de la Matriz Asociadas a la Membrana , Melanoma/metabolismo , Melanoma Experimental/terapia , Metaloendopeptidasas/genética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/fisiología , ARN Mensajero/análisis , ARN Neoplásico/análisis , Receptores de Vitronectina/inmunología , Inhibidor Tisular de Metaloproteinasa-2/genética , Células Tumorales Cultivadas/trasplante , Vitronectina/metabolismo
14.
J Clin Invest ; 123(3): 1068-81, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23426180

RESUMEN

Despite advances in clinical therapy, metastasis remains the leading cause of death in breast cancer patients. Mutations in mitochondrial DNA, including those affecting complex I and oxidative phosphorylation, are found in breast tumors and could facilitate metastasis. This study identifies mitochondrial complex I as critical for defining an aggressive phenotype in breast cancer cells. Specific enhancement of mitochondrial complex I activity inhibited tumor growth and metastasis through regulation of the tumor cell NAD+/NADH redox balance, mTORC1 activity, and autophagy. Conversely, nonlethal reduction of NAD+ levels by interfering with nicotinamide phosphoribosyltransferase expression rendered tumor cells more aggressive and increased metastasis. The results translate into a new therapeutic strategy: enhancement of the NAD+/NADH balance through treatment with NAD+ precursors inhibited metastasis in xenograft models, increased animal survival, and strongly interfered with oncogene-driven breast cancer progression in the MMTV-PyMT mouse model. Thus, aberration in mitochondrial complex I NADH dehydrogenase activity can profoundly enhance the aggressiveness of human breast cancer cells, while therapeutic normalization of the NAD+/NADH balance can inhibit metastasis and prevent disease progression.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Complejo I de Transporte de Electrón/fisiología , Neoplasias Pulmonares/metabolismo , Neoplasias Mamarias Experimentales/metabolismo , NAD/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiología , Acrilamidas/farmacología , Animales , Autofagia , Proteína 5 Relacionada con la Autofagia , Neoplasias Encefálicas/secundario , Línea Celular Tumoral , Proliferación Celular , Citocinas/antagonistas & inhibidores , Citocinas/metabolismo , Progresión de la Enfermedad , Complejo I de Transporte de Electrón/biosíntesis , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/patología , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias/metabolismo , Complejos Multiproteicos , NAD/fisiología , Trasplante de Neoplasias , Niacina/farmacología , Niacinamida/farmacología , Nicotinamida Fosforribosiltransferasa/antagonistas & inhibidores , Nicotinamida Fosforribosiltransferasa/metabolismo , Piperidinas/farmacología , Transporte de Proteínas , Proteínas/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas de Saccharomyces cerevisiae/biosíntesis , Serina-Treonina Quinasas TOR
15.
Cancer Cell ; 20(5): 553-4, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22094248

RESUMEN

Metastasis of epithelial tumors critically depends on acquisition of a disseminating phenotype that allows tumor cells to colonize distant organs. In this issue of Cancer Cell, Labelle et al. demonstrate that an epithelial-mesenchymal-like transition can be induced by interaction between platelets and tumor cells.

16.
Nat Rev Cancer ; 11(2): 123-34, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21258396

RESUMEN

Extensive experimental evidence shows that platelets support tumour metastasis. The activation of platelets and the coagulation system have a crucial role in the progression of cancer. Within the circulatory system, platelets guard tumour cells from immune elimination and promote their arrest at the endothelium, supporting the establishment of secondary lesions. These contributions of platelets to tumour cell survival and spread suggest platelets as a new avenue for therapy.


Asunto(s)
Plaquetas/patología , Metástasis de la Neoplasia/fisiopatología , Activación Plaquetaria , Biomarcadores de Tumor/metabolismo , Permeabilidad Capilar , Supervivencia Celular , Humanos , Metástasis de la Neoplasia/prevención & control , Células Neoplásicas Circulantes , Neovascularización Patológica , Inhibidores de Agregación Plaquetaria
17.
J Mol Biol ; 406(4): 595-603, 2011 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-21237172

RESUMEN

Immunoconjugates and multispecific antibodies are rapidly emerging as highly potent experimental therapeutics against cancer. We have developed a method to incorporate an unnatural amino acid, p-acetylphenylalanine (pAcPhe) into an antibody antigen binding fragment (Fab) targeting HER2 (human epidermal growth factor receptor 2), allowing site-specific labeling without disrupting antigen binding. Expression levels of the pAcPhe-containing proteins were comparable to that of wild-type protein in shake-flask and fermentation preparations. The pAcPhe-Fabs were labeled by reaction with hydroxylamine dye and biotin species to produce well-defined, singly conjugated Fabs. We then coupled a hydroxylamine biotin to the pAcPhe-Fab and demonstrated controlled assembly of Fabs in the presence of the tetrameric biotin-binding protein, NeutrAvidin. The position of Fab biotinylation dictates the geometry of multimer assembly, producing unique multimeric Fab structures. These assembled Fab multimers differentially attenuate Her2 phosphorylation in breast cancer cells that overexpress the Her2 receptor. Thus, an encoded unnatural amino acid produces a chemical "handle" by which immunoconjugates and multimers can be engineered.


Asunto(s)
Aminoácidos/metabolismo , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/metabolismo , Fenilalanina/análogos & derivados , Multimerización de Proteína , Humanos , Fragmentos Fab de Inmunoglobulinas/genética , Cinética , Modelos Químicos , Modelos Moleculares , Fenilalanina/metabolismo , Unión Proteica , Estructura Cuaternaria de Proteína , Receptor ErbB-2/inmunología
18.
Chem Biol ; 18(3): 299-303, 2011 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-21439474

RESUMEN

We report a strategy for the generation of heterodimeric protein conjugates using an unnatural amino acid with orthogonal reactivity. This paper addresses the challenges of site-specificity and homogeneity with respect to the synthesis of bivalent proteins and antibody-drug conjugates. There are numerous antibody-drug conjugates in preclinical and clinical development, yet these are based either on nonspecific lysine coupling chemistry or on disulfide modification made difficult by the large number of cysteines in antibodies. Here, we describe a recombinant approach that can be used to rapidly generate a variety of constructs with defined conjugation sites. Moreover, this methodology results in homogeneous antibody conjugates whose biological, physical, and pharmacological properties can be quantitatively assessed and subsequently optimized. As proof of concept, we have generated anti-Her2 Fab-Saporin conjugates that demonstrate excellent potency in vitro.


Asunto(s)
Anticuerpos Monoclonales/química , Antineoplásicos Fitogénicos/química , Inmunotoxinas/química , Fenilalanina/análogos & derivados , Proteínas Inactivadoras de Ribosomas Tipo 1/química , Anticuerpos Monoclonales/genética , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales Humanizados , Antineoplásicos Fitogénicos/inmunología , Antineoplásicos Fitogénicos/toxicidad , Línea Celular Tumoral , Dimerización , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/genética , Fragmentos Fab de Inmunoglobulinas/metabolismo , Inmunotoxinas/inmunología , Inmunotoxinas/toxicidad , Maleimidas/química , Fenilalanina/química , Receptor ErbB-2/inmunología , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Inactivadoras de Ribosomas Tipo 1/genética , Proteínas Inactivadoras de Ribosomas Tipo 1/metabolismo , Saporinas , Trastuzumab
19.
Clin Exp Metastasis ; 27(4): 217-31, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20225083

RESUMEN

Advanced metastatic disease is difficult to manage and specific therapeutic targets are rare. We showed earlier that metastatic breast cancer cells use the activated conformer of adhesion receptor integrin alphavbeta3 for dissemination. We now investigated if targeting this form of the receptor can impact advanced metastatic disease, and we analyzed the mechanisms involved. Treatment of advanced multi-organ metastasis in SCID mice with patient-derived scFv antibodies specific for activated integrin alphavbeta3 caused stagnation and regression of metastatic growth. The antibodies specifically localized to tumor lesions in vivo and inhibited alphavbeta3 ligand binding at nanomolar levels in vitro. At the cellular level, the scFs associated rapidly with high affinity alphavbeta3 and dissociated extremely slowly. Thus, the scFvs occupy the receptor on metastatic tumor cells for prolonged periods of time, allowing for inhibition of established cell interaction with natural alphavbeta3 ligands. Potential apoptosis inducing effects of the antibodies through interaction with caspase-3 were studied as potential additional mechanism of treatment response. However, in contrast to a previous concept, neither the RGD-containing ligand mimetic scFvs nor RGD peptides bound or activated caspase-3 at the cellular or molecular level. This indicates that the treatment effects seen in the animal model are primarily due to antibody interference with alphavbeta3 ligation. Inhibition of advanced metastatic disease by treatment with cancer patient derived single chain antibodies against the activated conformer of integrin alphavbeta3 identifies this form of the receptor as a suitable target for therapy.


Asunto(s)
Integrina alfaVbeta3/antagonistas & inhibidores , Metástasis de la Neoplasia/tratamiento farmacológico , Metástasis de la Neoplasia/patología , Anticuerpos de Cadena Única/uso terapéutico , Animales , Caspasa 3/metabolismo , Modelos Animales de Enfermedad , Humanos , Integrina alfaVbeta3/inmunología , Integrina alfaVbeta3/metabolismo , Ratones , Ratones SCID , Metástasis de la Neoplasia/inmunología , Metástasis de la Neoplasia/prevención & control , Anticuerpos de Cadena Única/inmunología
20.
Cell Stem Cell ; 6(1): 37-47, 2010 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-20085741

RESUMEN

Glioblastoma multiforme (GBM) is a highly aggressive form of brain cancer associated with a very poor prognosis. Recently, the initiation and growth of GBM has been linked to brain tumor-initiating cells (BTICs), which are poorly differentiated and share features with neural stem cells (NSCs). Here we describe a kinome-wide RNA interference screen to identify factors that control the tumorigenicity of BTICs. We identified several genes whose silencing induces differentiation of BTICs derived from multiple GBM patients. In particular, knockdown of the adaptor protein TRRAP significantly increased differentiation of cultured BTICs, sensitized the cells to apoptotic stimuli, and negatively affected cell cycle progression. TRRAP knockdown also significantly suppressed tumor formation upon intracranial BTIC implantation into mice. Together, these findings support a critical role for TRRAP in maintaining a tumorigenic, stem cell-like state.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/análisis , Neoplasias Encefálicas/química , Diferenciación Celular , Transformación Celular Neoplásica/química , Glioblastoma/química , Células Madre Neoplásicas/química , Proteínas Nucleares/análisis , Interferencia de ARN , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Apoptosis , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Glioblastoma/patología , Humanos , Ratones , Ratones SCID , Células Madre Neoplásicas/citología , Proteínas Nucleares/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA