Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Immunother Cancer ; 9(10)2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34599030

RESUMO

BACKGROUND: The human tumor microenvironment (TME) is a complex and dynamic milieu of diverse acellular and cellular components, creating an immunosuppressive environment, which contributes to tumor progression. We have previously shown that phosphatidylserine (PS) expressed on the surface of exosomes isolated from human TMEs is causally linked to T-cell immunosuppression, representing a potential immunotherapeutic target. In this study, we investigated the effect of ExoBlock, a novel PS-binding molecule, on T-cell responses in the TME. METHODS: We designed and synthesized a new compound, (ZnDPA)6-DP-15K, a multivalent PS binder named ExoBlock. The PS-binding avidity of ExoBlock was tested using an in vitro competition assay. The ability of this molecule to reverse exosome-mediated immunosuppression in vitro was tested using human T-cell activation assays. The in vivo therapeutic efficacy of ExoBlock was then tested in two different human tumor xenograft models, the melanoma-based xenomimetic (X-)mouse model, and the ovarian tumor-based omental tumor xenograft (OTX) model. RESULTS: ExoBlock was able to bind PS with high avidity and was found to consistently and significantly block the immunosuppressive activity of human ovarian tumor and melanoma-associated exosomes in vitro. ExoBlock was also able to significantly enhance T cell-mediated tumor suppression in vivo in both the X-mouse and the OTX model. In the X-mouse model, ExoBlock suppressed tumor recurrence in a T cell-dependent manner. In the OTX model, ExoBlock treatment resulted in an increase in the number as well as function of CD4 and CD8 T cells in the TME, which was associated with a reduction in tumor burden and metastasis, as well as in the number of circulating PS+ exosomes in tumor-bearing mice. CONCLUSION: Our results establish that targeting exosomal PS in TMEs with ExoBlock represents a promising strategy to enhance antitumor T-cell responses.


Assuntos
Exossomos/metabolismo , Neoplasias/imunologia , Neoplasias Ovarianas/genética , Fosfatidilserinas/metabolismo , Linfócitos T/metabolismo , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Camundongos Endogâmicos NOD , Neoplasias Ovarianas/patologia , Microambiente Tumoral
2.
Clin Transl Immunology ; 10(2): e1246, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33552509

RESUMO

OBJECTIVES: With a rapidly growing list of candidate immune-based cancer therapeutics, there is a critical need to generate highly reliable animal models to preclinically evaluate the efficacy of emerging immune-based therapies, facilitating successful clinical translation. Our aim was to design and validate a novel in vivo model (called Xenomimetic or 'X' mouse) that allows monitoring of the ability of human tumor-specific T cells to suppress tumor growth following their entry into the tumor. METHODS: Tumor xenografts are established rapidly in the greater omentum of globally immunodeficient NOD-scid IL2Rγnull (NSG) mice following an intraperitoneal injection of melanoma target cells expressing tumor neoantigen peptides, as well as green fluorescent protein and/or luciferase. Changes in tumor burden, as well as in the number and phenotype of adoptively transferred patient-derived tumor neoantigen-specific T cells in response to immunotherapy, are measured by imaging to detect fluorescence/luminescence and flow cytometry, respectively. RESULTS: The tumors progress rapidly and disseminate in the mice unless patient-derived tumor-specific T cells are introduced. An initial T cell-mediated tumor arrest is later followed by a tumor escape, which correlates with the upregulation of the checkpoint molecules programmed cell death-1 (PD-1) and lymphocyte-activation gene 3 (LAG3) on T cells. Treatment with immune-based therapies that target these checkpoints, such as anti-PD-1 antibody (nivolumab) or interleukin-12 (IL-12), prevented or delayed the tumor escape. Furthermore, IL-12 treatment suppressed PD-1 and LAG3 upregulation on T cells. CONCLUSION: Together, these results validate the X-mouse model and establish its potential to preclinically evaluate the therapeutic efficacy of immune-based therapies.

3.
J Immunol ; 201(12): 3750-3758, 2018 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-30446565

RESUMO

The tumor microenvironment is rendered immunosuppressive by a variety of cellular and acellular factors that represent potential cancer therapeutic targets. Although exosomes isolated from ovarian tumor ascites fluids have been previously reported to induce a rapid and reversible T cell arrest, the factors present on or within exosomes that contribute to immunosuppression have not been fully defined. In this study, we establish that GD3, a ganglioside expressed on the surface of exosomes isolated from human ovarian tumor ascites fluids, is causally linked to the functional arrest of T cells activated through their TCR. This arrest is inhibited by Ab blockade of exosomal GD3 or by the removal of GD3+ exosomes. Empty liposomes expressing GD3 on the surface also inhibit the activation of T cells, establishing that GD3 contributes to the functional arrest of T cells independent of factors present in exosomes. Finally, we demonstrate that the GD3-mediated arrest of the TCR activation is dependent upon sialic acid groups, because their enzymatic removal from exosomes or liposomes results in a loss of inhibitory capacity. Collectively, these data define GD3 as a potential immunotherapeutic target.


Assuntos
Líquido Ascítico/metabolismo , Exossomos/metabolismo , Gangliosídeos/metabolismo , Imunoterapia/métodos , Ácido N-Acetilneuramínico/metabolismo , Neoplasias Ovarianas/metabolismo , Linfócitos T/imunologia , Ascite , Células Cultivadas , Feminino , Humanos , Tolerância Imunológica , NF-kappa B/metabolismo , Estadiamento de Neoplasias , Neoplasias Ovarianas/imunologia , Microambiente Tumoral
4.
Cancer Immunol Res ; 6(2): 236-247, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29301753

RESUMO

Nano-sized membrane-encapsulated extracellular vesicles isolated from the ascites fluids of ovarian cancer patients are identified as exosomes based on their biophysical and compositional characteristics. We report here that T cells pulsed with these tumor-associated exosomes during TCR-dependent activation inhibit various activation endpoints including translocation of NFκB and NFAT into the nucleus, upregulation of CD69 and CD107a, production of cytokines, and cell proliferation. In addition, the activation of virus-specific CD8+ T cells that are stimulated with the cognate viral peptides presented in the context of class I MHC is also suppressed by the exosomes. The inhibition occurs without loss of cell viability and coincidentally with the binding and internalization of these exosomes. This exosome-mediated inhibition of T cells was transient and reversible: T cells exposed to exosomes can be reactivated once exosomes are removed. We conclude that tumor-associated exosomes are immunosuppressive and represent a therapeutic target, blockade of which would enhance the antitumor response of quiescent tumor-associated T cells and prevent the functional arrest of adoptively transferred tumor-specific T cells or chimeric antigen receptor T cells. Cancer Immunol Res; 6(2); 236-47. ©2018 AACR.


Assuntos
Microscopia Eletrônica de Transmissão/métodos , Linfócitos T/metabolismo , Proliferação de Células , Exossomos/imunologia , Feminino , Humanos , Neoplasias Ovarianas/imunologia
5.
J Pharm Sci ; 104(11): 3691-3702, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26228094

RESUMO

The role of aggregates in the immunogenicity of biologics is a major concern. A recent US FDA guidance on the issue suggests that a gap in knowledge exists regarding the type and size of aggregates involved in the immunogenicity of biologics. Furthermore, the guidance suggests that current techniques cannot capture the crucial stages of protein aggregation. Using a protein unfolding model developed earlier, we generated and classified aggregates of two therapeutic antibodies based on size and conformation. The immunogenic potential of these aggregates were then tested in a murine model. Our findings show that small native-like oligomeric aggregates (<100 nm) are more immunogenic toward the native protein than monomer and large non-native aggregates in the micron-size range, irrespective of route of administration [intravenous (i.v.) vs. subcutaneous (s.c.)]. Those smaller oligomeric aggregates represented 5%-20% of the total protein concentration in the test formulations. Furthermore, in vitro data suggest that TNF-α production by bone marrow-derived dendritic cells could serve as a predictive marker for increased immunogenic risk of aggregates after s.c. administration. The use of orthogonal techniques such as fluorescence anisotropy and quasielastic light scattering may be useful to detect these oligomeric aggregates.


Assuntos
Anticorpos Biespecíficos/imunologia , Anticorpos Monoclonais/imunologia , Formação de Anticorpos , Agregados Proteicos , Animais , Anticorpos Biespecíficos/administração & dosagem , Anticorpos Biespecíficos/química , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais/química , Células Dendríticas/imunologia , Humanos , Imunização , Camundongos , Conformação Proteica , Desdobramento de Proteína , Fator de Necrose Tumoral alfa/imunologia
6.
J Pharm Sci ; 104(8): 2451-6, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26038127

RESUMO

A major complication of replacement therapy with Factor VIII (FVIII) for hemophilia A (HA) is the development of unwanted immune responses. Previous studies showed that administration of FVIII in the presence of phosphatidyl serine (PS) reduced the development of anti-FVIII antibodies in HA mice. However, the impact of PS-mediated effects on immunological memory, such as generation of memory B-cells, is not clear. The effect of PS on memory B-cells was therefore investigated using adoptive transfer approach in FVIII(-/-) HA mice. Adoptive transfer of memory B-cells from a PS-FVIII-treated group to naïve mice followed by challenge of the recipient mice with FVIII showed a significantly reduced anti-FVIII antibody response in the recipient mice, compared with animals that received memory B-cells from free FVIII and FVIII-charge matched phosphatidyl glycerol (PG) group. The decrease in memory B-cell response is accompanied by an increase in FoxP3 expressing regulatory T-cells (Tregs). Flow cytometry studies showed that the generation of Tregs is higher in PS-treated animals as compared with FVIII and FVIII-PG treated animals. The PS-mediated hyporesponsiveness was found to be antigen-specific. The PS-FVIII immunization showed hyporesponsiveness toward FVIII rechallenge but not against ovalbumin (OVA) rechallenge, an unrelated antigen. This demonstrates that PS reduces immunologic memory of FVIII and induces antigen-specific peripheral tolerance in HA mice.


Assuntos
Fator VIII/administração & dosagem , Hematínicos/administração & dosagem , Hemofilia A/tratamento farmacológico , Tolerância Imunológica/efeitos dos fármacos , Memória Imunológica/efeitos dos fármacos , Veículos Farmacêuticos/química , Fosfatidilserinas/química , Animais , Anticorpos Neutralizantes/análise , Anticorpos Neutralizantes/biossíntese , Especificidade de Anticorpos , Linfócitos B/efeitos dos fármacos , Linfócitos B/imunologia , Hipersensibilidade a Drogas/prevenção & controle , Fator VIII/genética , Fator VIII/metabolismo , Fator VIII/uso terapêutico , Fatores de Transcrição Forkhead/sangue , Fatores de Transcrição Forkhead/metabolismo , Hematínicos/efeitos adversos , Hematínicos/metabolismo , Hematínicos/uso terapêutico , Hemofilia A/sangue , Hemofilia A/imunologia , Hemofilia A/metabolismo , Humanos , Lipossomos , Camundongos Knockout , Camundongos Transgênicos , Fosfatidilgliceróis/química , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/uso terapêutico , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo
7.
J Pharm Sci ; 103(11): 3457-3463, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25266204

RESUMO

The clinical use of therapeutic proteins can be complicated by the development of anti-product antibodies. We have previously observed that O-phospho-l-serine (OPLS) reduced antibody response to FVIII in Hemophilia-A (HA) mice. However, the mechanism underlying this observation is not clear. We hypothesize that OPLS reduces immunogenicity by inducing tolerogenic properties in dendritic cells (DCs). We tested this hypothesis using in vivo, in vitro, and ex vivo methods. Naive HA mice that were pre-exposed to FVIII in the presence of OPLS showed substantially lower antibody response following rechallenge with OPLS free FVIII as compared with dexamethasone-pretreated mice. Exposure of OPLS to bone-marrow-derived dendritic cells (BMDCs) in culturing conditions resulted in an increase in the regulatory cytokine TGF-ß and a decrease in proinflammatory cytokines TNF-α and IL12p70. This was accompanied by a significant reduction in upregulation of costimulatory marker CD40, as measured by flow cytometry. Furthermore, ex vivo matured BMDCs in the presence of FVIII and OPLS failed to elicit a robust immune response in HA mice compared with FVIII-treated BMDCs. Our data suggest that OPLS modulates the immune response by altering the function and maturation of DCs, resulting in the induction of tolerogenic properties. © 2014 Wiley Periodicals, Inc. and the American Pharmacists Association J Pharm Sci 103:3457-3463, 2014.


Assuntos
Adjuvantes Imunológicos/farmacologia , Coagulantes/farmacologia , Células Dendríticas/efeitos dos fármacos , Fator VIII/farmacologia , Hemofilia A/tratamento farmacológico , Tolerância Imunológica/efeitos dos fármacos , Fosfosserina/farmacologia , Animais , Anticorpos/sangue , Antígenos CD40/metabolismo , Células Cultivadas , Coagulantes/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Modelos Animais de Doenças , Fator VIII/imunologia , Hemofilia A/sangue , Hemofilia A/genética , Hemofilia A/imunologia , Interleucina-12/metabolismo , Camundongos Transgênicos , Fatores de Tempo , Fator de Crescimento Transformador beta/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
8.
AAPS J ; 15(4): 897-900, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23856740

RESUMO

The administration of therapeutic proteins via the subcutaneous route (sc) is desired for compliance and convenience, but could be challenging due to perceived immunogenic potential or unwanted immune responses. There are clinical and preclinical data supporting as well as refuting the generalized notion that sc is more immunogenic. We provide a mechanistic perspective of immunogenicity of therapeutic proteins administered via the sc route and discuss strategies and opportunities for novel therapeutic approaches to mitigate immunogenicity.


Assuntos
Apresentação de Antígeno/fisiologia , Fenômenos Imunogenéticos/fisiologia , Pele/imunologia , Pele/metabolismo , Animais , Apresentação de Antígeno/efeitos dos fármacos , Humanos , Fenômenos Imunogenéticos/efeitos dos fármacos , Imunossupressores/administração & dosagem , Imunossupressores/imunologia , Imunossupressores/metabolismo , Injeções Subcutâneas , Proteínas Proto-Oncogênicas/administração & dosagem , Proteínas Proto-Oncogênicas/imunologia , Proteínas Proto-Oncogênicas/metabolismo , Pele/efeitos dos fármacos
9.
Cancer Immun ; 13: 11, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23885217

RESUMO

Despite an initial response to chemotherapy, most patients with ovarian cancer eventually progress and succumb to their disease. Understanding why effector T cells that are known to infiltrate the tumor do not eradicate the disease after cytoreduction is critically important to the development of novel therapeutic strategies to augment tumor immunity and improve patient outcomes. Such studies have been hampered by the lack of a suitable in vivo model. We report here a simple and reliable model system in which ovarian tumor cell aggregates implanted intraperitoneally into severely immunodeficient NSG mice establish tumor microenvironments within the omentum. The rapid establishment of tumor xenografts within this small anatomically well-defined site enables the recovery, characterization, and quantification of tumor and tumor-associated T cells. We validate here the ability of the omental tumor xenograft (OTX) model to quantify changes in tumor cell number in response to therapy, to quantify changes in the tumor vasculature, and to demonstrate and study the immunosuppressive effects of the tumor microenvironment. Using the OTX model, we show that the tumor-associated T cells originally present within the tumor tissues are anergic and that fully functional autologous T cells injected into tumor-bearing mice localize within the tumor xenograft. The transferred T cells remain functional for up to 3 days within the tumor microenvironment but become unresponsive to activation after 7 days. The OTX model provides for the first time the opportunity to study in vivo the cellular and molecular events contributing to the arrest in T cell function in human ovarian tumors.


Assuntos
Neoplasias Ovarianas/irrigação sanguínea , Neoplasias Ovarianas/imunologia , Linfócitos T/imunologia , Animais , Modelos Animais de Doenças , Feminino , Xenoenxertos , Humanos , Imunoquímica , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/patologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Neovascularização Patológica/imunologia , Neovascularização Patológica/patologia , Neoplasias Ovarianas/patologia , Neoplasias Ovarianas/terapia , Linfócitos T/patologia , Microambiente Tumoral
10.
J Pharm Sci ; 101(8): 2763-76, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22581704

RESUMO

Inhibitors of epidermal growth factor (EGF) receptor (EGFR) tyrosine kinases show efficacy in cancers that are highly addicted to nonmutated EGF signaling, but off-target effects limit therapy. Carrier-based formulations could reduce drug deposition in normal tissues, enhance tumor deposition, and reduce free drug concentrations, thereby reducing the side effects. Therefore, the feasibility of developing nanoliposomal formulations of EGFR inhibitors was investigated. Gefitinib and erlotinib fluorescence was characterized as a tool for formulation development. Peak excitation was 345 nm and peak emission was 385-465 nm, depending upon the environment polarity. Emission was negligible in water but intense in nonpolar solvents, membranes, or bound to serum proteins. Cellular uptake and distribution could also be imaged by fluorescence in drug-resistant tumor spheroids. Gefitinib fluorescence characteristics enabled facile optimization of formulations. Although 4-6 mol % gefitinib could be incorporated in the liposome bilayer, 40-60 mol % could be encapsulated in stable, remote-loaded liposomes consisting of distearoylphosphatidylcholine-polyethylene glycol-distereoylphosphatidylethanolamine-cholesterol (9:1:5 mol:mol:mol). Drug leakage in serum, monitored by fluorescence, was minimal over 24 h at 37°C. The results provide both promising lead formulations as well as novel tools for evaluating new formulations of structurally similar receptor tyrosine kinase inhibitors and their cellular uptake and tissue biodistribution.


Assuntos
Antineoplásicos/administração & dosagem , Receptores ErbB/antagonistas & inibidores , Lipossomos/química , Inibidores de Proteínas Quinases/administração & dosagem , Quinazolinas/administração & dosagem , Animais , Antineoplásicos/química , Antineoplásicos/farmacocinética , Linhagem Celular , Linhagem Celular Tumoral , Cloridrato de Erlotinib , Gefitinibe , Humanos , Lipossomos/ultraestrutura , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacocinética , Quinazolinas/química , Quinazolinas/farmacocinética , Ratos , Espectrometria de Fluorescência
11.
J Pharm Sci ; 101(1): 48-55, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21953409

RESUMO

Factor VIII (FVIII) is an important coagulation cofactor and its deficiency causes Hemophilia A, a bleeding disorder. Replacement therapy using recombinant FVIII is currently the first line of therapy for Hemophilia A, but the development of neutralizing antibody is a major clinical complication for this therapy. Recently, it has been shown that FVIII associated with phosphatidylinositol (PI)-containing lipidic nanoparticles reduced development of neutralizing antibodies in Hemophilia A mice (Peng A, Straubinger RM, Balu-Iyer SV. 2010. AAPS J 12(3):473-481). Here, we investigated the underlying mechanism of this reduction in antibody response in culturing conditions. In vitro, PI interfered with the processing of FVIII by cultured dendritic cells (DC), resulting in a reduction in the upregulation of phenotypic costimulatory signal CD40. Furthermore, PI increased secretion of regulatory cytokines Transforming Growth Factor ß1 and Interleukin 10 (IL-10) but reduced the secretion of proinflammatory cytokines IL-6 and IL-17. The data suggest that PI reduces immunogenicity of FVIII by modulating DC maturation and inducing secretion of regulatory cytokines.


Assuntos
Antígenos CD40/metabolismo , Células Dendríticas/imunologia , Fator VIII/imunologia , Fator VIII/farmacologia , Fosfatidilinositóis/imunologia , Fosfatidilinositóis/farmacologia , Fator de Crescimento Transformador beta1/metabolismo , Animais , Anticorpos Neutralizantes/imunologia , Células Apresentadoras de Antígenos/efeitos dos fármacos , Células Apresentadoras de Antígenos/metabolismo , Antígenos CD40/imunologia , Células Cultivadas , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/imunologia , Fator VIII/metabolismo , Humanos , Interleucinas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Nanopartículas/administração & dosagem , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/farmacologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/metabolismo , Fator de Crescimento Transformador beta1/imunologia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/imunologia
12.
Pharm Res ; 29(2): 490-9, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21887597

RESUMO

PURPOSE: To determine the effect of dose, the anatomical site of injection, and the injection volume on subcutaneous absorption of rituximab in rats and to explore absorption mechanisms using pharmacokinetic modeling. METHODS: Rituximab serum concentrations were measured following intravenous and subcutaneous administration at the back, abdomen, and foot of rats. Several pharmacokinetic models were developed that included linear and saturable absorption, and degradation and/or protective binding at the injection site. RESULTS: Rituximab exhibited linear kinetics following intravenous administration; however, bioavailability following subcutaneous injection was inversely related to the dose level. For the 1 mg/kg dose, bioavailability was approximately 70% at all tested injection sites, with faster absorption from the foot (T(max) = 12 h for foot vs. 4.6 days for back). Bioavailability for the 10 mg/kg dose was 44 and 31% for the abdomen and back sites and 18% for 40 mg/kg injected at the back. A pharmacokinetic model that included binding as part of the absorption mechanism successfully captured the nonlinearities in rituximab absorption. CONCLUSION: The anatomical site of subcutaneous injection influences the rate of absorption and bioavailability of rituximab in rats. Saturable binding may be a major determinant of the nonlinear absorptive transport of monoclonal antibodies.


Assuntos
Anticorpos Monoclonais Murinos/administração & dosagem , Anticorpos Monoclonais Murinos/farmacocinética , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Animais , Injeções Subcutâneas , Masculino , Modelos Biológicos , Ratos , Ratos Wistar , Rituximab
13.
PLoS One ; 6(9): e24420, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21935406

RESUMO

Ovarian cancer is the most common cause of death from gynecological cancer. Understanding the biology of this disease, particularly how tumor-associated lymphocytes and fibroblasts contribute to the progression and metastasis of the tumor, has been impeded by the lack of a suitable tumor xenograft model. We report a simple and reproducible system in which the tumor and tumor stroma are successfully engrafted into NOD-scid IL2Rγ(null) (NSG) mice. This is achieved by injecting tumor cell aggregates derived from fresh ovarian tumor biopsy tissues (including tumor cells, and tumor-associated lymphocytes and fibroblasts) i.p. into NSG mice. Tumor progression in these mice closely parallels many of the events that are observed in ovarian cancer patients. Tumors establish in the omentum, ovaries, liver, spleen, uterus, and pancreas. Tumor growth is initially very slow and progressive within the peritoneal cavity with an ultimate development of tumor ascites, spontaneous metastasis to the lung, increasing serum and ascites levels of CA125, and the retention of tumor-associated human fibroblasts and lymphocytes that remain functional and responsive to cytokines for prolonged periods. With this model one will be able to determine how fibroblasts and lymphocytes within the tumor microenvironment may contribute to tumor growth and metastasis, and will make it possible to evaluate the efficacy of therapies that are designed to target these cells in the tumor stroma.


Assuntos
Modelos Animais de Doenças , Neoplasias Ovarianas/patologia , Animais , Ascite/metabolismo , Antígeno Ca-125/sangue , Antígeno Ca-125/metabolismo , Progressão da Doença , Ensaio de Imunoadsorção Enzimática , Feminino , Humanos , Imunoensaio , Imuno-Histoquímica , Interferon gama/metabolismo , Interleucina-12/metabolismo , Camundongos , Metástase Neoplásica/patologia , Neoplasias Ovarianas/sangue , Neoplasias Ovarianas/metabolismo
14.
Clin Immunol ; 138(2): 135-45, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21094627

RESUMO

A major clinical complication in the treatment of Hemophilia A using exogenously administered recombinant Factor VIII (FVIII) is the development of neutralizing antibodies. It has been shown previously that FVIII complexed with phosphatidylserine (PS) reduces the development of total and neutralizing antibody titers in hemophilic mice. The effect of complexation of FVIII with PS upon dendritic cell (DC) uptake, maturation and processing, T-cell proliferation and cytokine secretion profiles was investigated. Flow cytometric studies of DC showed that PS inhibited the up-regulation of cell surface co-stimulatory markers (CD86 and CD40). PS reduced T-cell proliferation and significantly increased levels of TGF-ß and IL-10 but reduced secretion of IL-6 and IL-17 compared to controls. The data suggest that PS reduces immunogenicity of FVIII by regulating dendritic cell maturation and subsequent T-lymphocyte activity through modulation of cytokine secretion. A possible mechanism for PS-mediated induction of FVIII tolerance is discussed.


Assuntos
Anticorpos Neutralizantes/imunologia , Células Dendríticas/efeitos dos fármacos , Fator VIII/imunologia , Hemofilia A/imunologia , Tolerância Imunológica/efeitos dos fármacos , Fosfatidilserinas/farmacologia , Proteínas Recombinantes/imunologia , Animais , Anticorpos Neutralizantes/biossíntese , Antígeno B7-2/imunologia , Linfócitos T CD4-Positivos/imunologia , Antígenos CD40/imunologia , Diferenciação Celular/imunologia , Células Cultivadas , Citocinas/imunologia , Citocinas/metabolismo , Células Dendríticas/imunologia , Fator VIII/uso terapêutico , Hemofilia A/tratamento farmacológico , Humanos , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Camundongos , Proteínas Recombinantes/uso terapêutico , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/imunologia
15.
J Pharm Sci ; 99(4): 1697-706, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19798762

RESUMO

Aggregation is a critical issue that hampers the development of monoclonal antibody therapeutics (Mabs). Traditionally, aggregation is considered a process in which native forms of proteins are transformed into an unstable highly associated form through an intermediate formation step. Here we describe the unfolding of an antiCD40 antibody using a folding model based on Lumry-Eyring nucleated polymerization (LENP) model. This model captures several experimental features of the thermal unfolding of this protein as studied by common in situ biophysical techniques such as circular dichroism, fluorescence spectroscopy, and turbidity measurements. According to this model, the unfolding and aggregation of the antiCD40 antibody is determined by several distinct steps that include conformational change(s) to generate aggregation prone states, reversible oligomer formation, nucleation and growth as well as their kinetics, and the formation of higher order assemblies/aggregates. Furthermore, the loss of monomer is controlled by both thermodynamic (equilibrium unfolding) and kinetic determinants of the unfolding process. This approach captures both of these rate-limiting steps. It can be concluded that this approach is sensitive to formulation conditions such as protein concentration, changes in buffer conditions, and temperature stress. The potential use of this approach in formulation development and stability testing of Mabs is discussed.


Assuntos
Anticorpos Monoclonais/química , Antígenos CD40/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Dicroísmo Circular , Estabilidade de Medicamentos , Modelos Químicos , Murinae , Conformação Proteica , Dobramento de Proteína , Estabilidade Proteica , Espectrometria de Fluorescência , Espectrofotometria Ultravioleta , Temperatura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA