Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Angew Chem Int Ed Engl ; 53(44): 11863-7, 2014 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-25213874

RESUMO

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.


Assuntos
Anticorpos Monoclonais/química , Antineoplásicos/química , Imunoconjugados/química , Imunoterapia/métodos , Receptores CXCR4/química , Linhagem Celular Tumoral , Humanos
2.
J Clin Invest ; 123(3): 1068-81, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23426180

RESUMO

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.


Assuntos
Neoplasias Encefálicas/metabolismo , Complexo I de Transporte de Elétrons/fisiologia , Neoplasias Pulmonares/metabolismo , Neoplasias Mamárias Experimentais/metabolismo , NAD/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiologia , Acrilamidas/farmacologia , Animais , Autofagia , Proteína 5 Relacionada à Autofagia , Neoplasias Encefálicas/secundário , Linhagem Celular Tumoral , Proliferação de Células , Citocinas/antagonistas & inibidores , Citocinas/metabolismo , Progressão da Doença , Complexo I de Transporte de Elétrons/biossíntese , Feminino , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Mitocôndrias/metabolismo , Complexos Multiproteicos , NAD/fisiologia , Transplante de Neoplasias , Niacina/farmacologia , Niacinamida/farmacologia , Nicotinamida Fosforribosiltransferase/antagonistas & inibidores , Nicotinamida Fosforribosiltransferase/metabolismo , Piperidinas/farmacologia , Transporte Proteico , Proteínas/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas de Saccharomyces cerevisiae/biossíntese , Serina-Treonina Quinases TOR
3.
Proc Natl Acad Sci U S A ; 109(40): 16101-6, 2012 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-22988081

RESUMO

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.


Assuntos
Aminoácidos/química , Anticorpos Monoclonais Humanizados/química , Neoplasias da Mama/tratamento farmacológico , Imunoconjugados/química , Engenharia de Proteínas/métodos , Aminobenzoatos/química , Animais , Linhagem Celular Tumoral , Descoberta de Drogas/métodos , Ensaio de Imunoadsorção Enzimática , Escherichia coli , Feminino , Humanos , Imunoconjugados/farmacocinética , Imunoconjugados/uso terapêutico , Imunoglobulina G/química , Camundongos , Camundongos SCID , Oligopeptídeos/química , Receptor ErbB-2/química , Receptor ErbB-2/imunologia , Trastuzumab
4.
Cancer Cell ; 20(5): 553-4, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22094248

RESUMO

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.

5.
Bioconjug Chem ; 22(8): 1535-44, 2011 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-21774545

RESUMO

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.


Assuntos
Anticorpos/uso terapêutico , Antígenos de Neoplasias/imunologia , Antineoplásicos , Integrina alfaV/imunologia , Neoplasias/imunologia , Anticorpos/imunologia , Comunicação Celular , Linhagem Celular Tumoral , Frutose-Bifosfato Aldolase , Humanos , Imunoconjugados/uso terapêutico , Integrina alfaVbeta3/imunologia , Integrinas/imunologia , Neoplasias/tratamento farmacológico
6.
Chem Biol ; 18(3): 299-303, 2011 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-21439474

RESUMO

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.


Assuntos
Anticorpos Monoclonais/química , Antineoplásicos Fitogênicos/química , Imunotoxinas/química , Fenilalanina/análogos & derivados , Proteínas Inativadoras de Ribossomos Tipo 1/química , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais Humanizados , Antineoplásicos Fitogênicos/imunologia , Antineoplásicos Fitogênicos/toxicidade , Linhagem Celular Tumoral , Dimerização , Humanos , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/genética , Fragmentos Fab das Imunoglobulinas/metabolismo , Imunotoxinas/imunologia , Imunotoxinas/toxicidade , Maleimidas/química , Fenilalanina/química , Receptor ErbB-2/imunologia , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Inativadoras de Ribossomos Tipo 1/genética , Proteínas Inativadoras de Ribossomos Tipo 1/metabolismo , Saporinas , Trastuzumab
7.
Nat Rev Cancer ; 11(2): 123-34, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21258396

RESUMO

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.


Assuntos
Plaquetas/patologia , Metástase Neoplásica/fisiopatologia , Ativação Plaquetária , Biomarcadores Tumorais/metabolismo , Permeabilidade Capilar , Sobrevivência Celular , Humanos , Metástase Neoplásica/prevenção & controle , Células Neoplásicas Circulantes , Neovascularização Patológica , Inibidores da Agregação Plaquetária
8.
J Mol Biol ; 406(4): 595-603, 2011 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-21237172

RESUMO

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.


Assuntos
Aminoácidos/metabolismo , Fragmentos Fab das Imunoglobulinas/química , Fragmentos Fab das Imunoglobulinas/metabolismo , Fenilalanina/análogos & derivados , Multimerização Proteica , Humanos , Fragmentos Fab das Imunoglobulinas/genética , Cinética , Modelos Químicos , Modelos Moleculares , Fenilalanina/metabolismo , Ligação Proteica , Estrutura Quaternária de Proteína , Receptor ErbB-2/imunologia
9.
Am J Pathol ; 176(6): 2958-71, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20382702

RESUMO

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.


Assuntos
Neoplasias Encefálicas/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Neoplasias da Mama/secundário , Animais , Astrócitos/metabolismo , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Encéfalo/irrigação sanguínea , Neoplasias Encefálicas/irrigação sanguínea , Neoplasias Encefálicas/metabolismo , Neoplasias da Mama/irrigação sanguínea , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Circulação Cerebrovascular , Progressão da Doença , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Microglia/metabolismo , Metástase Neoplásica
10.
Clin Exp Metastasis ; 27(4): 217-31, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20225083

RESUMO

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.


Assuntos
Integrina alfaVbeta3/antagonistas & inibidores , Metástase Neoplásica/tratamento farmacológico , Metástase Neoplásica/patologia , Anticorpos de Cadeia Única/uso terapêutico , Animais , Caspase 3/metabolismo , Modelos Animais de Doenças , Humanos , Integrina alfaVbeta3/imunologia , Integrina alfaVbeta3/metabolismo , Camundongos , Camundongos SCID , Metástase Neoplásica/imunologia , Metástase Neoplásica/prevenção & controle , Anticorpos de Cadeia Única/imunologia
11.
Cell Stem Cell ; 6(1): 37-47, 2010 Jan 08.
Artigo em Inglês | MEDLINE | ID: mdl-20085741

RESUMO

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.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/análise , Neoplasias Encefálicas/química , Diferenciação Celular , Transformação Celular Neoplásica/química , Glioblastoma/química , Células-Tronco Neoplásicas/química , Proteínas Nucleares/análise , Interferência de RNA , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Apoptose , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Feminino , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Glioblastoma/patologia , Humanos , Camundongos , Camundongos SCID , Células-Tronco Neoplásicas/citologia , Proteínas Nucleares/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Methods Mol Biol ; 568: 249-59, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19582432

RESUMO

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.


Assuntos
Neoplasias Encefálicas/terapia , Técnicas de Cultura de Células/métodos , Terapia Genética/métodos , Neurônios/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Linhagem Celular , Congelamento , Humanos , Injeções , Lentivirus/genética , Transplante de Células-Tronco , Transdução Genética
13.
Proc Natl Acad Sci U S A ; 106(26): 10666-71, 2009 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-19541645

RESUMO

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.


Assuntos
Neoplasias Encefálicas/secundário , Integrina alfaVbeta3/metabolismo , Neoplasias Mamárias Experimentais/patologia , Neovascularização Patológica/patologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Apoptose , Western Blotting , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Hipóxia , Marcação In Situ das Extremidades Cortadas , Integrina alfaVbeta3/genética , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Camundongos , Camundongos SCID , Mutação , Transplante de Neoplasias , Fosfoproteínas/metabolismo , Fosforilação , Processamento de Proteína Pós-Traducional , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante Heterólogo , Carga Tumoral , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
14.
Bioorg Med Chem Lett ; 19(14): 3821-4, 2009 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-19428247

RESUMO

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.).


Assuntos
Anticorpos Catalíticos/química , Frutose-Bifosfato Aldolase/química , Fragmentos Fab das Imunoglobulinas/química , Anticorpos Catalíticos/imunologia , Anticorpos Catalíticos/metabolismo , Linhagem Celular Tumoral , Frutose-Bifosfato Aldolase/imunologia , Frutose-Bifosfato Aldolase/metabolismo , Humanos , Fragmentos Fab das Imunoglobulinas/imunologia , Fragmentos Fab das Imunoglobulinas/metabolismo , Integrina alfaVbeta3/metabolismo , Cetonas/química
15.
Blood ; 111(1): 190-9, 2008 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-17901245

RESUMO

Coagulation activation by tissue factor (TF) is implicated in cancer progression, cancer-associated thrombosis and metastasis. The role of direct TF signaling pathways in cancer, however, remains incompletely understood. Here we address how TF contributes to primary tumor growth by using a unique pair of isotype-matched antibodies that inhibit either coagulation (monoclonal antibody [Mab]-5G9) or direct signaling (Mab-10H10). We demonstrate that the inhibitory antibody of direct TF-VIIa signaling not only blocks TF-VIIa mediated activation of PAR2, but also disrupts the interaction of TF with integrins. In epithelial and TF-expressing endothelial cells, association of TF with beta1 integrins is regulated by TF extracellular ligand binding and independent of PAR2 signaling or proteolytic activity of VIIa. In contrast, alpha3beta1 integrin association of TF is constitutive in breast cancer cells and blocked by Mab-10H10 but not by Mab-5G9. Mab-5G9 has antitumor activity in vivo, but we show here that Mab-10H10 is at least as effective in suppressing human xenograft tumors in 2 different models. Breast tumor growth was also attenuated by blocking PAR2 signaling. These results show that tumor cell TF-PAR2 signaling is crucial for tumor growth and suggest that anti-TF strategies can be applied in cancer therapy with minor impairment of TF-dependent hemostatic pathways.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/secundário , Transdução de Sinais/fisiologia , Tromboplastina/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Divisão Celular , Linhagem Celular Transformada , Endotélio Vascular/citologia , Fator VIIa/metabolismo , Fator VIIa/farmacologia , Humanos , Integrina beta1/metabolismo , Queratinócitos/citologia , Queratinócitos/metabolismo , Camundongos , Camundongos SCID , Receptor PAR-1/metabolismo , Receptor PAR-2/metabolismo , Transdução de Sinais/imunologia , Tromboplastina/imunologia , Veias Umbilicais/citologia
16.
Proc Natl Acad Sci U S A ; 104(21): 9024-8, 2007 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-17494758

RESUMO

The platelet paradigm in hemostasis and thrombosis involves an initiation step that depends on platelet membrane receptors binding to ligands on a damaged or inflamed vascular surface. Once bound to the surface, platelets provide a unique microenvironment supporting the accumulation of more platelets and the elaboration of a fibrin-rich network produced by coagulation factors. The platelet-specific receptor glycoprotein (GP) Ib-IX, is critical in this process and initiates the formation of a platelet-rich thrombus by tethering the platelet to a thrombogenic surface. A role for platelets beyond the hemostasis/thrombosis paradigm is emerging with significant platelet contributions in both tumorigenesis and inflammation. We have established congenic (N10) mouse colonies (C57BL/6J) with dysfunctional GP Ib-IX receptors in our laboratory that allow us an opportunity to examine the relevance of platelet GP Ib-IX in syngeneic mouse models of experimental metastasis. Our results demonstrate platelet GP Ib-IX contributes to experimental metastasis because a functional absence of GP Ib-IX correlates with a 15-fold reduction in the number of lung metastatic foci using B16F10.1 melanoma cells. The results demonstrate that the extracellular domain of the alpha-subunit of GP Ib is the structurally relevant component of the GP Ib-IX complex contributing to metastasis. Our results support the hypothesis that platelet GP Ib-IX functions that support normal hemostasis or pathologic thrombosis also contribute to tumor malignancy.


Assuntos
Plaquetas/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Complexo Glicoproteico GPIb-IX de Plaquetas/metabolismo , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Deleção de Genes , Humanos , Neoplasias Pulmonares/secundário , Melanoma/genética , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexo Glicoproteico GPIb-IX de Plaquetas/genética , Ligação Proteica , Subunidades Proteicas/deficiência , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo
17.
Clin Cancer Res ; 13(6): 1656-62, 2007 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-17363518

RESUMO

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.


Assuntos
Neoplasias Encefálicas/etiologia , Neoplasias Encefálicas/secundário , Animais , Encéfalo/irrigação sanguínea , Neoplasias Encefálicas/irrigação sanguínea , Permeabilidade da Membrana Celular/fisiologia , Modelos Animais de Doenças , Progressão da Doença , Metabolismo Energético/fisiologia , Humanos , Melanoma Experimental/patologia , Modelos Biológicos , Neovascularização Patológica/patologia , Receptor ErbB-2/fisiologia , Fator de Transcrição STAT3/fisiologia
18.
Mol Pharm ; 4(3): 435-47, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17373820

RESUMO

Major obstacles in the development of new therapeutic anticancer drugs are the low bioavailability of hydrophilic substances and the nonspecific toxicity toward healthy tissues. As such, cell-targeting oligopeptides have emerged as attractive drug delivery vehicles for a variety of different types of cargo. The recently identified peptide Pep42 binds to the glucose-regulated protein 78 (GRP78), which is overexpressed on the cell surface of human cancer cells and internalizes into these cells. Herein, we demonstrate how Pep42 can be utilized as a carrier for different types of cytotoxic drugs to specifically target human cancer cell lines in vitro in a strictly GRP78-dependent manner. Furthermore, the mechanism of internalization of Pep42 was elucidated and found to involve clathrin-mediated endocytosis. Pep42 subsequently colocalizes within the lysosomal compartment. Importantly, we also provide evidence that Pep42-conjugated quantum dots have the ability to selectively enrich in tumor tissue in a xenograft mouse model. Our results suggest that the highly specific GRP78-Pep42 interaction can be utilized for the generation of Pep42-drug conjugates as a powerful anticancer drug delivery system that could substantially enhance the efficacy of chemotherapy by increasing the drug-tumor specificity, thus minimizing the adverse side effects associated with conventional cancer therapeutics.


Assuntos
Sistemas de Liberação de Medicamentos , Proteínas de Choque Térmico/metabolismo , Chaperonas Moleculares/metabolismo , Peptídeos Cíclicos/metabolismo , Sequência de Aminoácidos , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Apoptose , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Clatrina/metabolismo , Endocitose , Chaperona BiP do Retículo Endoplasmático , Proteínas de Choque Térmico/antagonistas & inibidores , Proteínas de Choque Térmico/genética , Humanos , Ligantes , Lisossomos/metabolismo , Camundongos , Chaperonas Moleculares/antagonistas & inibidores , Chaperonas Moleculares/genética , Transplante de Neoplasias , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/metabolismo , Peptídeos Cíclicos/química , Fotoquimioterapia , Pontos Quânticos , RNA Interferente Pequeno/genética , Transplante Heterólogo
19.
Cancer Res ; 67(4): 1472-86, 2007 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-17308085

RESUMO

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.


Assuntos
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/secundário , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Animais , Processos de Crescimento Celular/fisiologia , Ciclo do Ácido Cítrico , Modelos Animais de Doenças , Metabolismo Energético , Feminino , Glutationa/metabolismo , Glicólise , Humanos , Camundongos , Camundongos SCID , Mitocôndrias/metabolismo , Oxirredução , Consumo de Oxigênio , Via de Pentose Fosfato , Proteômica
20.
Biochemistry ; 45(31): 9434-44, 2006 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-16878978

RESUMO

Peptidic ligands can be used for specific cell targeting and the delivery of payloads into the target cell. Here we describe the screening of a pool of cyclic peptide phage display libraries using whole-cell panning against human melanoma cell line Me6652/4. This strategy resulted in the selection of the cyclic 13-mer Pep42, CTVALPGGYVRVC, which showed preferential internalization into melanoma cell line Me6652/4 versus the reference cell line Me6652/56. This translocation is a receptor-mediated process that does not require electrostatic interactions nor does it involve transfer to the lysosomal compartment. The cellular receptor for Pep42 was identified as the surface membrane form of glucose-regulated protein 78 (GRP78), a member of the heat shock protein family and a marker on malignant cancer cells. The cellular uptake and intracellular trafficking of Pep42-Quantum Dot conjugates was monitored by confocal laser microscopy, and colocalization within the endoplasmic reticulum was observed. The uptake of Pep42 could be blocked by a monoclonal antibody against the identified receptor. Furthermore, Pep42 was shown to target specifically GRP78-expressing cancer cells. The in vitro cytotoxicity of a Pep42-Taxol conjugate was evaluated by flow cytometry wherein the conjugate was shown to induce apoptosis and was more effective in promoting programmed cell death in Me6652/4 cells. In summary, the data presented suggest that cyclic peptide Pep42 might be a powerful tool in the construction of drug conjugates designed to selectively kill malignant cancer cells.


Assuntos
Proteínas de Choque Térmico/metabolismo , Melanoma/metabolismo , Chaperonas Moleculares/metabolismo , Peptídeos Cíclicos/metabolismo , Sequência de Aminoácidos , Anticorpos Monoclonais/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Linhagem Celular Tumoral , Chaperona BiP do Retículo Endoplasmático , Proteínas de Choque Térmico/efeitos dos fármacos , Proteínas de Choque Térmico/genética , Humanos , Ligantes , Melanoma/química , Chaperonas Moleculares/efeitos dos fármacos , Chaperonas Moleculares/genética , Dados de Sequência Molecular , Paclitaxel/farmacologia , Biblioteca de Peptídeos , Peptídeos Cíclicos/efeitos dos fármacos , Peptídeos Cíclicos/genética , Transporte Proteico/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA