Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cancer Immunol Immunother ; 61(7): 991-1003, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22127364

RESUMO

Breast and ovarian cancer are two of the leading causes of cancer deaths among women in the United States. Overexpression of the HER2/neu oncoprotein has been reported in patients affected with breast and ovarian cancers, and is associated with poor prognosis. To develop a novel targeted therapy for HER2/neu expressing tumors, we have constructed a fully human IgE with the variable regions of the scFv C6MH3-B1 specific for HER2/neu. This antibody was expressed in murine myeloma cells and was properly assembled and secreted. The Fc region of this antibody triggers in vitro degranulation of rat basophilic cells expressing human FcεRI (RBL SX-38) in the presence of murine mammary carcinoma cells that express human HER2/neu (D2F2/E2), but not the shed (soluble) antigen (ECD(HER2)) alone. This IgE is also capable of inducing passive cutaneous anaphylaxis in a human FcεRIα transgenic mouse model, in the presence of a cross-linking antibody, but not in the presence of soluble ECD(HER2). Additionally, IgE enhances antigen presentation in human dendritic cells and facilitates cross-priming, suggesting that the antibody is able to stimulate a secondary T-cell anti-tumor response. Furthermore, we show that this IgE significantly prolongs survival of human FcεRIα transgenic mice bearing D2F2/E2 tumors. We also report that the anti-HER2/neu IgE is well tolerated in a preliminary study conducted in Macaca fascicularis (cynomolgus) monkeys. In summary, our results suggest that this IgE should be further explored as a potential therapeutic against HER2/neu overexpressing tumors, such as breast and ovarian cancers.


Assuntos
Neoplasias da Mama/imunologia , Neoplasias da Mama/terapia , Imunização Passiva/métodos , Imunoglobulina E/farmacologia , Receptor ErbB-2/imunologia , Animais , Especificidade de Anticorpos , Neoplasias da Mama/enzimologia , Células CHO , Linhagem Celular Tumoral , Cricetinae , Feminino , Humanos , Imunoglobulina E/imunologia , Macaca fascicularis , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Ratos , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Biochim Biophys Acta ; 1820(3): 291-317, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21851850

RESUMO

BACKGROUND: Traditional cancer therapy can be successful in destroying tumors, but can also cause dangerous side effects. Therefore, many targeted therapies are in development. The transferrin receptor (TfR) functions in cellular iron uptake through its interaction with transferrin. This receptor is an attractive molecule for the targeted therapy of cancer since it is upregulated on the surface of many cancer types and is efficiently internalized. This receptor can be targeted in two ways: 1) for the delivery of therapeutic molecules into malignant cells or 2) to block the natural function of the receptor leading directly to cancer cell death. SCOPE OF REVIEW: In the present article we discuss the strategies used to target the TfR for the delivery of therapeutic agents into cancer cells. We provide a summary of the vast types of anti-cancer drugs that have been delivered into cancer cells employing a variety of receptor binding molecules including Tf, anti-TfR antibodies, or TfR-binding peptides alone or in combination with carrier molecules including nanoparticles and viruses. MAJOR CONCLUSIONS: Targeting the TfR has been shown to be effective in delivering many different therapeutic agents and causing cytotoxic effects in cancer cells in vitro and in vivo. GENERAL SIGNIFICANCE: The extensive use of TfR for targeted therapy attests to the versatility of targeting this receptor for therapeutic purposes against malignant cells. More advances in this area are expected to further improve the therapeutic potential of targeting the TfR for cancer therapy leading to an increase in the number of clinical trials of molecules targeting this receptor. This article is part of a Special Issue entitled Transferrins: molecular mechanisms of iron transport and disorders.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/administração & dosagem , Terapia de Alvo Molecular/métodos , Neoplasias/tratamento farmacológico , Receptores da Transferrina/metabolismo , Antineoplásicos/metabolismo , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Proteínas de Transporte/administração & dosagem , Proteínas de Transporte/metabolismo , Sistemas de Liberação de Medicamentos , Vetores Genéticos , Humanos , Ferro/metabolismo
3.
Cancer Immunol Immunother ; 61(9): 1535-46, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22193986

RESUMO

Although most monoclonal antibodies developed for cancer therapy are of the IgG class, antibodies of the IgE class have certain properties that make them attractive as cancer therapeutics. These properties include the superior affinity for the Fc epsilon receptors (FcεRs), the low serum level of IgE that minimizes competition of endogenous IgE for FcεR occupancy, and the ability to induce a broad and vigorous immune response through the interaction with multiple cells including mast cells, basophils, monocytes, macrophages, dendritic cells, and eosinophils. Tumor-targeted IgE antibodies are expected to harness the allergic response against tumors and activate a secondary, T-cell-mediated immune response. Importantly, the IgE antibody can be used for passive immunotherapy and as an adjuvant of cancer vaccines. However, there are important limitations in the use of animal models including the fact that human IgE does not interact with rodent FcεRs and that there is a different cellular distribution of FcεRs in humans and rodents. Despite these limitations, different murine models have been used with success to evaluate the in vivo anti-cancer activity of several IgE antibodies. These models include wild-type immunocompetent animals bearing syngeneic tumors, xenograft models using immunocompromised mice bearing human tumors and reconstituted with human effector cells, and human FcεRIα transgenic mice bearing syngeneic tumors. In addition, non-human primates such as cynomolgus monkeys can be potentially used for toxicological and pharmacokinetic studies. This article describes the advantages and disadvantages of these models and their use in evaluating the in vivo properties of IgE antibodies for cancer therapy.


Assuntos
Imunização Passiva/métodos , Imunoglobulina E/imunologia , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/terapia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Transgênicos , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Leuk Lymphoma ; 52(11): 2169-78, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21870996

RESUMO

A number of antibodies have been developed that induce lethal iron deprivation (LID) by targeting the transferrin receptor 1 (TfR1/CD71) and either neutralizing transferrin (Tf) binding, blocking internalization of the receptor and/or inducing its degradation. We have developed recombinant antibodies targeting human TfR1 (ch128.1 and ch128.1Av), which induce receptor degradation and are cytotoxic to certain malignant B-cells. We now show that internalization of TfR1 bound to these antibodies can lead to its sequestration and degradation, as well as reduced Tf uptake, and the induction of a transcriptional response consistent with iron deprivation, which is mediated in part by downstream targets of p53. Cells resistant to these antibodies do not sequester and degrade TfR1 after internalization of the antibody/receptor complex, and accordingly maintain their ability to internalize Tf. These findings are expected to facilitate the rational design and clinical use of therapeutic agents targeting iron import via TfR1 in hematopoietic malignancies.


Assuntos
Anticorpos/metabolismo , Antígenos CD/metabolismo , Linfócitos B/metabolismo , Ferro/metabolismo , Receptores da Transferrina/metabolismo , Animais , Anticorpos/imunologia , Anticorpos/farmacologia , Antígenos CD/genética , Antígenos CD/imunologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/patologia , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Desferroxamina/farmacologia , Endocitose/efeitos dos fármacos , Perfilação da Expressão Gênica , Humanos , Immunoblotting , Camundongos , Camundongos SCID , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Ligação Proteica , Receptores da Transferrina/genética , Receptores da Transferrina/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sideróforos/farmacologia , Transferrina/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
5.
J Immunother ; 34(6): 500-8, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21654517

RESUMO

We previously developed an antibody-avidin fusion protein (ch128.1Av) targeting the human transferrin receptor 1 (TfR1, also known as CD71), which demonstrates direct in vitro cytotoxicity against malignant hematopoietic cells. This cytotoxicity is attributed to its ability to decrease the level of TfR1 leading to lethal iron deprivation. We now report that ch128.1Av shows the ability to bind the Fcγ receptors and the complement component C1q, suggesting that it is capable of eliciting Fc-mediated effector functions such as antibody-dependent cell-mediated cytotoxicity and complement-mediated cytotoxicity. In addition, in 2 disseminated multiple myeloma xenograft mouse models, we show that a single dose of ch128.1Av results in significant antitumor activity, including long-term survival. It is interesting to note that the parental antibody without avidin (ch128.1) also shows remarkable in vivo anticancer activity despite its limited in vitro cytotoxicity. Finally, we demonstrate that ch128.1Av is not toxic to pluripotent hematopoietic progenitor cells using the long-term cell-initiating culture assay suggesting that these important progenitors would be preserved in different therapeutic approaches, including the in vitro purging of cancer cells for autologous transplantation and in vivo passive immunotherapy. Our results suggest that ch128.1Av and ch128.1 may be effective in the therapy of human multiple myeloma and potentially other hematopoietic malignancies.


Assuntos
Anticorpos , Imunoterapia , Mieloma Múltiplo/terapia , Receptores da Transferrina/imunologia , Proteínas Recombinantes de Fusão , Animais , Anticorpos/uso terapêutico , Avidina/imunologia , Linhagem Celular Tumoral , Feminino , Células HL-60 , Células-Tronco Hematopoéticas/imunologia , Humanos , Fragmentos Fc das Imunoglobulinas/imunologia , Camundongos , Camundongos SCID , Mieloma Múltiplo/imunologia , Ligação Proteica/imunologia , Proteínas Recombinantes de Fusão/uso terapêutico , Células U937
6.
Int J Oncol ; 36(5): 1299-307, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20372806

RESUMO

Multiple myeloma (MM) is an incurable disease of malignant plasma cells. Recent therapeutic advancements have resulted in improved response rates, however, there is no improvement in overall survival, therefore, new therapeutics are needed. Since the transferrin receptor is upregulated on the surface of MM cells, we previously developed an antibody fusion protein consisting of an IgG3 specific for the human transferrin receptor 1 (TfR1, CD71) genetically fused to avidin at its carboxy-terminus (ch128.1Av). We have previously shown that ch128.1Av exhibits intrinsic cytotoxicity against certain malignant B-cells by disrupting the cycling of the TfR and decreasing TfR cell surface expression resulting in lethal iron starvation. In addition, ch128.1Av can sensitize malignant cells to apoptosis induced by gambogic acid, a herbal drug used in Chinese medicine. In this study, we hypothesized that ch128.1Av may also sensitize drug-resistant malignant B-cells to chemotherapeutic agents by inhibiting key survival pathways. In this study we show that ch128.1Av sensitizes malignant B-cells to apoptosis induced by cisplatin (CDDP). The sensitization by ch128.1Av resulted in the inhibition of the constitutively activated Akt and NF-kappaB survival/antiapoptotic pathways and downstream decreased expression of antiapoptotic gene products such as BclxL and survivin. The direct role of the inhibition of the Akt and NF-kappaB pathways by ch128.1Av in CDDP-mediated cytotoxicity was demonstrated by the use of specific chemical inhibitors and siRNA which mimicked the effects of ch128.1Av. Overall, this study provides evidence of the therapeutic potential of ch128.1Av as a chemo-sensitizing agent in drug-resistant tumor cells.


Assuntos
Anticorpos/química , Apoptose , Avidina/química , Linfócitos B/patologia , Cisplatino/farmacologia , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Recombinantes de Fusão/química , Humanos , Imunoglobulina G/química , Medicina Tradicional Chinesa , Potencial da Membrana Mitocondrial , Mieloma Múltiplo/metabolismo , Receptores da Transferrina/química , Xantonas/química
7.
Mol Cancer ; 8: 68, 2009 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-19715609

RESUMO

BACKGROUND: Hormone-refractory prostate cancer (HRPC) is characterized by poor response to chemotherapy and high mortality, particularly among African American men when compared to other racial/ethnic groups. It is generally accepted that docetaxel, the standard of care for chemotherapy of HRPC, primarily exerts tumor cell death by inducing mitotic catastrophe and caspase-dependent apoptosis following inhibition of microtubule depolymerization. However, there is a gap in our knowledge of mechanistic events underlying docetaxel-induced caspase-independent cell death, and the genes that antagonize this process. This knowledge is important for circumventing HRPC chemoresistance and reducing disparities in prostate cancer mortality. RESULTS: We investigated mechanistic events associated with docetaxel-induced death in HRPC cell lines using various approaches that distinguish caspase-dependent from caspase-independent cell death. Docetaxel induced both mitotic catastrophe and caspase-dependent apoptosis at various concentrations. However, caspase activity was not essential for docetaxel-induced cytotoxicity since cell death associated with lysosomal membrane permeabilization still occurred in the presence of caspase inhibitors. Partial inhibition of docetaxel-induced cytotoxicity was observed after inhibition of cathepsin B, but not inhibition of cathepsins D and L, suggesting that docetaxel induces caspase-independent, lysosomal cell death. Simultaneous inhibition of caspases and cathepsin B dramatically reduced docetaxel-induced cell death. Ectopic expression of lens epithelium-derived growth factor p75 (LEDGF/p75), a stress survival autoantigen and transcription co-activator, attenuated docetaxel-induced lysosomal destabilization and cell death. Interestingly, LEDGF/p75 overexpression did not protect cells against DTX-induced mitotic catastrophe, and against apoptosis induced by tumor necrosis factor related apoptosis inducing ligand (TRAIL), suggesting selectivity in its pro-survival activity. CONCLUSION: These results underscore the ability of docetaxel to induce concomitantly caspase-dependent and independent death pathways in prostate cancer cells. The results also point to LEDGF/p75 as a potential contributor to cellular resistance to docetaxel-induced lysosomal destabilization and cell death, and an attractive candidate for molecular targeting in HRPC.


Assuntos
Apoptose/efeitos dos fármacos , Caspases/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Lisossomos/efeitos dos fármacos , Taxoides/farmacologia , Clorometilcetonas de Aminoácidos/farmacologia , Antineoplásicos/farmacologia , Caspase 2/metabolismo , Caspase 3/metabolismo , Inibidores de Caspase , Catepsina B/antagonistas & inibidores , Catepsina B/metabolismo , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Inibidores de Cisteína Proteinase/farmacologia , Docetaxel , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Citometria de Fluxo , Humanos , Immunoblotting , Peptídeos e Proteínas de Sinalização Intercelular/genética , Lisossomos/metabolismo , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Fatores de Tempo , Transfecção
8.
J Gene Med ; 11(8): 655-63, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19455593

RESUMO

BACKGROUND: Targeted gene transduction in vivo is the ultimate preferred method for gene delivery. We previously developed targeting lentiviral vectors that specifically recognize cell surface molecules with conjugated antibodies and mediate targeted gene transduction both in vitro and in vivo. Although effective in some experimental settings, the conjugation of virus with antibodies is mediated by the interaction between protein A and the Fc region of antibodies, which is not as stable as covalent conjugation. We have now developed a more stable conjugation strategy utilizing the interaction between avidin and biotin. METHODS: We inserted the biotin-adaptor-peptide, which was biotinylated by secretory biotin ligase at specific sites, into our targeting envelope proteins, enabling conjugation of the pseudotyped virus with avidin, streptavidin or neutravidin. RESULTS: When conjugated with avidin-antibody fusion proteins or the complex of avidin and biotinylated targeting molecules, the vectors could mediate specific transduction to targeted cells recognized by the targeting molecules. When conjugated with streptavidin-coated magnetic beads, transduction by the vectors was targeted to the locations of magnets. CONCLUSIONS: This targeting vector system can be used for broad applications of targeted gene transduction using biotinylated targeting molecules or targeting molecules fused with avidin.


Assuntos
Biotina/metabolismo , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Lentivirus/genética , Peptídeos/metabolismo , Avidina/metabolismo , Biotinilação , Western Blotting , Linhagem Celular , Eletroforese em Gel de Poliacrilamida , Humanos , Magnetismo , Mutagênese Insercional , Receptores da Transferrina/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Sindbis virus/genética , Transdução Genética , Proteínas do Envelope Viral/metabolismo , Vírion/metabolismo
9.
Cancer Immunol Immunother ; 58(6): 915-30, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18941743

RESUMO

Trastuzumab (Herceptin), a humanized IgG1 antibody raised against the human epidermal growth factor receptor 2 (HER2/neu), is the main antibody in clinical use against breast cancer. Pre-clinical evidence and clinical studies indicate that trastuzumab employs several anti-tumour mechanisms that most likely contribute to enhanced survival of patients with HER2/neu-positive breast carcinomas. New strategies are aimed at improving antibody-based therapeutics like trastuzumab, e.g. by enhancing antibody-mediated effector function mechanisms. Based on our previous findings that a chimaeric ovarian tumour antigen-specific IgE antibody showed greater efficacy in tumour cell killing, compared to the corresponding IgG1 antibody, we have produced an IgE homologue of trastuzumab. Trastuzumab IgE was engineered with the same light- and heavy-chain variable-regions as trastuzumab, but with an epsilon in place of the gamma-1 heavy-chain constant region. We describe the physical characterisation and ligand binding properties of the trastuzumab IgE and elucidate its potential anti-tumour activities in functional assays. Both trastuzumab and trastuzumab IgE can activate monocytic cells to kill tumour cells, but they operate by different mechanisms: trastuzumab functions in antibody-dependent cell-mediated phagocytosis (ADCP), whereas trastuzumab IgE functions in antibody-dependent cell-mediated cytotoxicity (ADCC). Trastuzumab IgE, incubated with mast cells and HER2/neu-expressing tumour cells, triggers mast cell degranulation, recruiting against cancer cells a potent immune response, characteristic of allergic reactions. Finally, in viability assays both antibodies mediate comparable levels of tumour cell growth arrest. These functional characteristics of trastuzumab IgE, some distinct from those of trastuzumab, indicate its potential to complement or improve upon the existing clinical benefits of trastuzumab.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Imunoglobulina E/imunologia , Receptor ErbB-2/imunologia , Anticorpos Monoclonais Humanizados , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Neoplasias da Mama/imunologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Feminino , Citometria de Fluxo , Humanos , Monócitos/citologia , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , Fagocitose , Engenharia de Proteínas , Receptores de IgE/imunologia , Receptores de IgE/metabolismo , Trastuzumab
10.
Expert Opin Biol Ther ; 8(5): 609-32, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18407765

RESUMO

BACKGROUND: Antibody-cytokine fusion proteins consist of cytokines fused to an antibody to improve antibody-targeted cancer immunotherapy. These molecules have the capacity to enhance the tumoricidal activity of the antibodies and/or activate a secondary antitumor immune response. OBJECTIVE: To review the strategies used to develop antibody-cytokine fusion proteins and their in vitro and in vivo properties, including preclinical and clinical studies focusing on IL-2, IL-12 and GM-CSF. METHODS: Articles were found by searching databases such as PubMed and Clinical Trials of the US National Institutes of Health. RESULTS/CONCLUSION: Multiple antibody-cytokine fusion proteins have demonstrated significant antitumor activity as direct therapeutics or as adjuvants of cancer vaccines in preclinical studies, paving the way for their clinical evaluation.


Assuntos
Anticorpos/uso terapêutico , Citocinas/uso terapêutico , Neoplasias/terapia , Proteínas Recombinantes de Fusão/uso terapêutico , Animais , Anticorpos/química , Citocinas/química , Humanos , Neoplasias/imunologia
11.
J Immunol ; 180(9): 6199-210, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18424742

RESUMO

TRAIL promotes apoptotic tumor cell death; however, TRAIL-resistant tumors need to be sensitized to reverse resistance. Proteasome inhibitors potentiate TRAIL apoptosis in vitro and in vivo and correlate with up-regulation of death receptor 5 (DR5) via an unknown mechanism. We hypothesized that the proteasome inhibitor NPI-0052 inhibits the transcription repressor Yin Yang 1 (YY1) which regulates TRAIL resistance and negatively regulates DR5 transcription. Treatment of PC-3 and Ramos cells with NPI-0052 (

Assuntos
Apoptose/efeitos dos fármacos , Lactonas/farmacologia , Neoplasias/tratamento farmacológico , Inibidores de Proteases/farmacologia , Pirróis/farmacologia , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/biossíntese , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Regulação para Cima/efeitos dos fármacos , Fator de Transcrição YY1/biossíntese , Apoptose/genética , Ácidos Borônicos/farmacologia , Bortezomib , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Células-Tronco Hematopoéticas/metabolismo , Humanos , Lactonas/efeitos adversos , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Neoplasias/metabolismo , Inibidores de Proteases/efeitos adversos , Pirazinas/farmacologia , Pirróis/efeitos adversos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Transcrição Gênica/efeitos dos fármacos , Fator de Transcrição YY1/antagonistas & inibidores , Fator de Transcrição YY1/genética
12.
Mol Cancer Ther ; 6(11): 2995-3008, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18025284

RESUMO

We have previously developed an antibody fusion protein composed of a mouse/human chimeric IgG3 specific for the human transferrin receptor genetically fused to avidin (anti-hTfR IgG3-Av) as a universal delivery system for cancer therapy. This fusion protein efficiently delivers biotinylated FITC into cancer cells via TfR-mediated endocytosis. In addition, anti-hTfR IgG3-Av alone exhibits intrinsic cytotoxic activity and interferes with hTfR recycling, leading to the rapid degradation of the TfR and lethal iron deprivation in certain malignant B-cell lines. We now report on the cytotoxic effects of a conjugate composed of anti-hTfR IgG3-Av and biotinylated saporin 6 (b-SO6), a toxin derived from the plant Saponaria officinalis that inhibits protein synthesis. Conjugation of anti-hTfR IgG3-Av with b-SO6 enhances the cytotoxic effect of the fusion protein in sensitive cells and also overcomes the resistance of malignant cells that show low sensitivity to the fusion protein alone. Our results show for the first time that loading anti-hTfR IgG3-Av with a biotinylated toxin enhances the cytotoxicity of the fusion protein alone. These results suggest that anti-hTfR IgG3-Av has great potential as a therapeutic agent for a wide range of applications due to its intrinsic cytotoxic activity plus its ability to deliver biotinylated molecules into cancer cells.


Assuntos
Avidina/metabolismo , Biotinilação , Neoplasias Hematológicas/patologia , Imunoglobulina G/farmacologia , Proteínas de Plantas/metabolismo , Receptores da Transferrina/antagonistas & inibidores , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Inativadoras de Ribossomos Tipo 1/metabolismo , Caspases/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Ativação Enzimática/efeitos dos fármacos , Neoplasias Hematológicas/enzimologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Deficiências de Ferro , Biossíntese de Proteínas/efeitos dos fármacos , Receptores da Transferrina/metabolismo , Saporinas
13.
J Control Release ; 124(1-2): 35-42, 2007 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-17884229

RESUMO

The human transferrin receptor (hTfR1) is a membrane-bound protein involved in transferrin (Tf)-mediated iron uptake and is highly expressed on malignant cells. A second version of the receptor (hTfR2) also mediates Tf-dependent iron import. We previously developed a protein composed of avidin fused to a mouse/human chimeric IgG3 specific for hTfR (anti-hTfR IgG3-Av) that was originally designed to deliver biotinylated drugs into cancer cells. We have now found that anti-hTfR IgG3-Av does not cross-react with hTfR2 and binds hTfR1 expressed on the surface of cells, attached to a solid surface, and in solution. We also found that the hemochromatosis protein (HFE), another ligand of the TfR, does not inhibit the binding of anti-hTfR IgG3-Av to the receptor. In addition, using live cell laser scanning confocal microscopy (LCLSCM) we demonstrated that anti-hTfR IgG3-Av and anti-hTfR IgG3 are internalized into cells expressing hTfR1 at a similar rate. Furthermore, our proliferation and morphological studies demonstrated the effective cytotoxicity of a biotinylated toxin delivered by anti-hTfR IgG3-Av only into cells expressing hTfR1. Our results better define the properties of anti-hTfR IgG3-Av and pave the way for the rational design of future in vitro and in vivo studies for the treatment of human malignancies.


Assuntos
Anticorpos Monoclonais/farmacologia , Antineoplásicos/farmacologia , Avidina/farmacologia , Imunoconjugados/farmacologia , Receptores da Transferrina/antagonistas & inibidores , Proteínas Recombinantes de Fusão/farmacologia , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/metabolismo , Antígenos CD/biossíntese , Antígenos CD/imunologia , Antineoplásicos/administração & dosagem , Antineoplásicos/metabolismo , Avidina/administração & dosagem , Avidina/metabolismo , Biotinilação , Células CHO , Sobrevivência Celular/efeitos dos fármacos , Cricetinae , Cricetulus , Sistemas de Liberação de Medicamentos , Ensaio de Imunoadsorção Enzimática , Humanos , Imunoconjugados/administração & dosagem , Imunoconjugados/metabolismo , Imunoglobulina G/administração & dosagem , Imunoglobulina G/metabolismo , Imunoglobulina G/farmacologia , Ferro/metabolismo , Ligantes , Camundongos , Microscopia Confocal , Ligação Proteica , Receptores da Transferrina/biossíntese , Receptores da Transferrina/imunologia , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/metabolismo
14.
Cancer Immunol Immunother ; 56(9): 1507-12, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17310381

RESUMO

We have previously reported that the antibody fusion proteins anti-HER2/neu IgG3 fused to IL-12 [(IL-12)-IgG3] or GM-CSF [IgG3-(GM-CSF)] independently or in combination are effective anti-tumor agents against D2F2/E2 murine mammary cancer cells expressing human HER2/neu in the peritoneum. Importantly, the long-term survivors were immune to the subcutaneous challenge with D2F2/E2 and the parental D2F2 not expressing HER2/neu. We now show that these long-term survivors also exhibit significant protection against subsequent subcutaneous challenge with the murine colon carcinoma CT26-HER2/neu, and later against subcutaneous challenge with the parental CT26. These results suggest that the long-term systemic protection against mammary cancer elicited by treatment with antibody-cytokine fusion proteins can be extended to prevent the growth of a tumor from different origin expressing HER2/neu, and that this protection is not limited to this antigen alone, since it also prevented the growth of the parental tumor cells.


Assuntos
Anticorpos/uso terapêutico , Neoplasias do Colo/terapia , Citocinas/uso terapêutico , Neoplasias Mamárias Animais/terapia , Neoplasias Experimentais/terapia , Proteínas Recombinantes de Fusão/uso terapêutico , Animais , Anticorpos/imunologia , Antineoplásicos/imunologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Neoplasias do Colo/imunologia , Citocinas/imunologia , Feminino , Imunidade , Neoplasias Mamárias Animais/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Experimentais/imunologia , Proteínas Recombinantes de Fusão/imunologia
15.
Blood ; 109(9): 3915-21, 2007 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-17213289

RESUMO

The aurora kinases facilitate transit from G2 through cytokinesis and, thus, are targets in cancer therapy. Multiple myeloma (MM) is a malignancy characterized by genetic instability, suggesting a disruption of checkpoints that arrest cells at G2M when injury to the mitotic machinery occurs. Since deficient checkpoints would prevent cell cycle arrest and may render cells susceptible to apoptosis in mitosis and since aurora kinases are intermediaries in checkpoint pathways, we tested antimyeloma effects of 2 agents that inhibit aurora kinases. Both inhibited growth of MM lines and primary myeloma samples at nanomolar concentrations while having less of an effect on proliferating lymphocytes and hematopoietic cells. MM cells were not protected by IL-6 or activating mutations of Ras. Antimyeloma effects included induction of tetraploidy followed by apoptosis. Apoptosis correlated with inhibition of aurora activity as shown by reduction of histone 3B phosphorylation. Ectopic expression of aurora A protected MM cells against aurora inhibitors but had no effect on apoptosis induced by bortezomib. As expression of RHAMM in MM contributes to genetic instability, we tested effects of RHAMM. RHAMM overexpression enhanced sensitivity to apoptosis and RHAMM silencing decreased sensitivity. These results suggest potential for aurora kinase inhibitors in MM especially in patients in whom RHAMM is overexpressed.


Assuntos
Fase G2/efeitos dos fármacos , Mieloma Múltiplo/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Aurora Quinases , Ácidos Borônicos/farmacologia , Bortezomib , Linhagem Celular Transformada , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Ensaios de Seleção de Medicamentos Antitumorais , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/genética , Proteínas da Matriz Extracelular/biossíntese , Proteínas da Matriz Extracelular/genética , Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Células-Tronco Hematopoéticas/enzimologia , Histonas/metabolismo , Humanos , Receptores de Hialuronatos/biossíntese , Receptores de Hialuronatos/genética , Interleucina-6/metabolismo , Linfócitos/enzimologia , Mieloma Múltiplo/enzimologia , Mieloma Múltiplo/genética , Mutação , Fosforilação/efeitos dos fármacos , Ploidias , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Serina-Treonina Quinases/biossíntese , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Pirazinas/farmacologia
16.
Clin Immunol ; 121(2): 144-58, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16904380

RESUMO

The transferrin receptor (TfR) is a cell membrane-associated glycoprotein involved in the cellular uptake of iron and in the regulation of cell growth. Iron uptake occurs via the internalization of iron-loaded transferrin (Tf) mediated by the interaction with the TfR. In addition, the TfR may also contain other growth regulatory properties in certain normal and malignant cells. The elevated levels of TfR in malignancies, its relevance in cancer, and the extracellular accessibility of this molecule make it an excellent antigen for the treatment of cancer using antibodies. The TfR can be targeted by monoclonal antibodies specific for the extracellular domain of the receptor. In this review, we summarize advancements in the basic physiology of the TfR including structure, function, and expression. We also discuss the efficacy of targeting the TfR using cytotoxic antibodies that inhibit cell growth and/or induce apoptosis in targeted malignant cells.


Assuntos
Neoplasias/terapia , Receptores da Transferrina/fisiologia , Transferrina/química , Animais , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais/uso terapêutico , Apoptose , Proliferação de Células , Citoplasma/metabolismo , Humanos , Ferro/farmacocinética , Células K562 , Camundongos , Modelos Biológicos , Receptores da Transferrina/imunologia , Receptores da Transferrina/metabolismo
17.
Clin Immunol ; 121(2): 159-76, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16920030

RESUMO

Traditional anti-cancer treatments consist of chemotherapeutic drugs that effectively eliminate rapidly dividing tumor cells. However, in many cases chemotherapy fails to eliminate the tumor and even when chemotherapy is successful, its systemic cytotoxicity often results in detrimental side effects. To overcome these problems, many laboratories have focused on the design of novel therapies that exhibit tumor specific toxicity. The transferrin receptor (TfR), a cell membrane-associated glycoprotein involved in iron homeostasis and cell growth, has been explored as a target to deliver therapeutics into cancer cells due to its increased expression on malignant cells, accessibility on the cell surface, and constitutive endocytosis. The TfR can be targeted by direct interaction with conjugates of its ligand transferrin (Tf) or by monoclonal antibodies specific for the TfR. In this review we summarize the strategies of targeting the TfR in order to deliver therapeutic agents into tumor cells by receptor-mediated endocytosis.


Assuntos
Antineoplásicos/administração & dosagem , Portadores de Fármacos/administração & dosagem , Neoplasias/terapia , Receptores da Transferrina/metabolismo , Antibióticos Antineoplásicos/administração & dosagem , Anticorpos Monoclonais/administração & dosagem , Doxorrubicina/administração & dosagem , Sistemas de Liberação de Medicamentos , Endocitose , Vetores Genéticos/administração & dosagem , Humanos , Modelos Biológicos , Polímeros/administração & dosagem , Receptores da Transferrina/imunologia , Ribonucleases/administração & dosagem , Ricina/administração & dosagem , Toxinas Biológicas/administração & dosagem
18.
Blood ; 108(8): 2745-54, 2006 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-16804109

RESUMO

We have previously reported that an anti-human transferrin receptor IgG3-avidin fusion protein (anti-hTfR IgG3-Av) inhibits the proliferation of an erythroleukemia-cell line. We have now found that anti-hTfR IgG3-Av also inhibits the proliferation of additional human malignant B and plasma cells. Anti-hTfR IgG3-Av induces internalization and rapid degradation of the TfR. These events can be reproduced in cells treated with anti-hTfR IgG3 cross-linked with a secondary Ab, suggesting that they result from increased TfR cross-linking. Confocal microscopy of cells treated with anti-hTfR IgG3-Av shows that the TfR is directed to an intracellular compartment expressing the lysosomal marker LAMP-1. The degradation of TfR is partially blocked by cysteine protease inhibitors. Furthermore, cells treated with anti-hTfR IgG3-Av exhibit mitochondrial depolarization and activation of caspases 9, 8, and 3. The mitochondrial damage and cell death can be prevented by iron supplementation, but cannot be fully blocked by a pan-caspase inhibitor. These results suggest that anti-hTfR IgG3-Av induces lethal iron deprivation, but the resulting cell death does not solely depend on caspase activation. This report provides insights into the mechanism of cell death induced by anti-TfR Abs such as anti-hTfR IgG3-Av, a molecule that may be useful in the treatment of B-cell malignancies such as multiple myeloma.


Assuntos
Avidina/farmacologia , Neoplasias Hematológicas/terapia , Imunoglobulina G/farmacologia , Receptores da Transferrina/antagonistas & inibidores , Clorometilcetonas de Aminoácidos/farmacologia , Apoptose/efeitos dos fármacos , Inibidores de Caspase , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Reagentes de Ligações Cruzadas , Desferroxamina/farmacologia , Neoplasias Hematológicas/imunologia , Neoplasias Hematológicas/metabolismo , Neoplasias Hematológicas/patologia , Humanos , Ferro/farmacologia , Leucemia Plasmocitária/metabolismo , Leucemia Plasmocitária/patologia , Leucemia Plasmocitária/terapia , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Mieloma Múltiplo/terapia , Receptores da Transferrina/imunologia , Receptores da Transferrina/metabolismo , Proteínas Recombinantes de Fusão/farmacologia , Sideróforos/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA