Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
J Immunol ; 194(3): 878-82, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25548231

RESUMO

CD4(+) regulatory T cells (Tregs) are critical for maintaining self-tolerance and function to prevent autoimmune disease. High densities of intratumoral Tregs are generally associated with poor patient prognosis, a correlation attributed to their broad immune-suppressive features. Two major populations of Tregs have been defined, thymically derived natural Tregs (nTregs) and peripherally induced Tregs (iTregs). However, the relative contribution of nTregs versus iTregs to the intratumoral Treg compartment remains controversial. Demarcating the proportion of nTregs versus iTregs has important implications in the design of therapeutic strategies to overcome their antagonistic effects on antitumor immune responses. We used epigenetic, phenotypic, and functional parameters to evaluate the composition of nTregs versus iTregs isolated from mouse tumor models and primary human tumors. Our findings failed to find evidence for extensive intratumoral iTreg induction. Rather, we identified a population of Foxp3-stable nTregs in tumors from mice and humans.


Assuntos
Epigênese Genética , Fatores de Transcrição Forkhead/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias/genética , Neoplasias/imunologia , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Animais , Antígenos de Superfície/metabolismo , Linhagem Celular Tumoral , Ilhas de CpG , Metilação de DNA , Modelos Animais de Doenças , Fatores de Transcrição Forkhead/metabolismo , Humanos , Imunofenotipagem , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Camundongos , Camundongos Transgênicos , Neoplasias/metabolismo , Fator de Crescimento Transformador beta/metabolismo
2.
Front Oncol ; 12: 1023510, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36419901

RESUMO

NK cells have unique attributes to react towards cells undergoing malignant transformation or viral infection. This reactivity is regulated by activating or inhibitory germline encoded receptors. An impaired NK cell function may result from an aberrant expression of such receptors, a condition often seen in patients with hematological cancers. Acute lymphoblastic leukemia (ALL) is the most common pediatric cancer worldwide and NK cells have emerged as crucial targets for developing immunotherapies. However, there are important gaps concerning the phenotype and behavior of NK cells during emergence of ALL. In this study we analyze the phenotype and function of NK cells from peripheral blood in pediatric patients with ALL at diagnosis. Our results showed that NK cells exhibited an altered phenotype highlighted by a significant reduction in the overall expression and percent representation of activating receptors compared to age-matched controls. No significant differences were found for the expression of inhibitory receptors. Moreover, NK cells with a concurrent reduced expression in various activating receptors, was the dominant phenotype among patients. An alteration in the relative frequencies of NK cells expressing NKG2A and CD57 within the mature NK cell pool was also observed. In addition, NK cells from patients displayed a significant reduction in the ability to sustain antibody-dependent cellular cytotoxicity (ADCC). Finally, an aberrant expression of activating receptors is associated with the phenomenon of leukemia during childhood.

3.
Cancer Immunol Immunother ; 60(2): 197-206, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20976449

RESUMO

We have previously shown that the suppressive function of regulatory T cells (Tregs) from peripheral blood mononuclear cells (PBMCs) is enhanced in patients with prostate cancer when compared with healthy individuals. Two phase II studies using the PSA-TRICOM vaccine in patients with metastatic castration-resistant prostate cancer (mCRPC) showed evidence of patient benefit in terms of enhanced survival. The Halabi nomogram has been used to predict survival (HPS) of patients with mCRPC treated with conventional chemotherapy or second-line hormonal therapy. Tregs from PBMCs of patients (n = 23) with mCRPC were obtained pre- and post-three monthly vaccinations, and analyzed for number, phenotype, and suppressive function. Changes post- versus pre-vaccination in these parameters were compared with 3-year survival and HPS. No differences in Treg numbers were observed post- versus pre-vaccination. Trends (P = 0.029) were observed between overall survival (OS) and a decrease in Treg suppressive function post- versus pre-vaccination. Trends were also observed in analyzing effector:Treg (CD4(+)CD25(+)CD127(-)FoxP3(+)CTLA4(+)) ratio post- versus pre-vaccination with OS versus HPS. These data provide preliminary evidence for a possible association between improved OS and a decrease in Treg function when PBMCs are analyzed after three monthly vaccinations. Patients with an OS > HPS were more likely to have decreased Treg function following vaccine. Larger studies to confirm and extend these findings are warranted.


Assuntos
Vacinas Anticâncer/imunologia , Neoplasias da Próstata/patologia , Neoplasias da Próstata/terapia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia , Antígenos CD/metabolismo , Antígeno CTLA-4 , Vacinas Anticâncer/administração & dosagem , Citometria de Fluxo , Humanos , Tolerância Imunológica/imunologia , Contagem de Linfócitos , Masculino , Metástase Neoplásica/imunologia , Neoplasias da Próstata/imunologia , Análise de Sobrevida , Linfócitos T Reguladores/metabolismo
4.
Clin Cancer Res ; 15(7): 2387-96, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19276249

RESUMO

PURPOSE: IFN-alpha is a pleiotropic cytokine possessing immunomodulatory properties that may improve the efficacy of therapeutic cancer vaccines. The aim of this study was to evaluate the effectiveness and compatibility of combining recombinant IFN-alpha with poxvirus vaccines targeting the human carcinoembryonic antigen (CEA) in murine models of colorectal and pancreatic adenocarcinomas, where CEA is a self-antigen. EXPERIMENTAL DESIGN: The phenotypic and functional effects of IFN-alpha were evaluated in the draining inguinal lymph nodes of tumor-free mice. We studied the effect of the site of IFN-alpha administration (local versus distal) on antigen-specific immune responses to poxvirus vaccination. Mechanistic studies were conducted to assess the efficacy of IFN-alpha and CEA-directed poxvirus vaccines in tumor-bearing CEA transgenic mice. RESULTS: We identified a dose and schedule of IFN-alpha that induced a locoregional expansion of the draining inguinal lymph nodes and improved cellular cytotoxicity (natural killer and CD8(+)) and antigen presentation. Suppression of the vaccinia virus was avoided by administering IFN-alpha distal to the site of vaccination. The combination of IFN-alpha and vaccine inhibited tumor growth, improved survival, and elicited CEA-specific CTL responses in mice with CEA(+) adenocarcinomas. In mice with pancreatic tumors, IFN-alpha slowed tumor growth, induced CTL activity, and increased CD8(+) tumor-infiltrating lymphocytes. CONCLUSIONS: These data suggest that IFN-alpha can be used as a biological response modifier with antigen-directed poxvirus vaccines to yield significant therapeutic antitumor immune responses. This study provides the rationale and mechanistic insights to support a clinical trial of this immunotherapeutic strategy in patients with CEA-expressing carcinomas.


Assuntos
Adenocarcinoma/tratamento farmacológico , Adjuvantes Imunológicos/uso terapêutico , Antineoplásicos/uso terapêutico , Vacinas Anticâncer/uso terapêutico , Vírus da Varíola das Aves Domésticas/genética , Interferon-alfa/uso terapêutico , Vaccinia virus/genética , Adenocarcinoma/imunologia , Animais , Vacinas Anticâncer/genética , Antígeno Carcinoembrionário/genética , Linhagem Celular Tumoral , Terapia Combinada , DNA Recombinante/análise , Feminino , Antígenos de Histocompatibilidade Classe I/metabolismo , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fenótipo
5.
J Immunother Cancer ; 8(1)2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32518090

RESUMO

BACKGROUND: Natural killer and cytotoxic CD8+ T cells are major players during antitumor immunity. They express NKG2D, an activating receptor that promotes tumor elimination through recognition of the MHC class I chain-related proteins A and B (MICA and MICB). Both molecules are overexpressed on a great variety of tumors from different tissues, making them attractive targets for immunotherapy. However, tumors shed MICA and MICB, and the soluble forms of both (sMICA and sMICB) mediate tumor-immune escape. Some reports indicate that anti-MICA antibodies (Ab) can promote the restoration of antitumor immunity through the induction of direct antitumor effects (antibody-dependent cell-mediated cytotoxicity, ADCC) and scavenging of sMICA. Therefore, we reasoned that an active induction of anti-MICA Ab with an immunogenic protein might represent a novel therapeutic and prophylactic alternative to restore antitumor immunity. METHODS: We generated a highly immunogenic chimeric protein (BLS-MICA) consisting of human MICA fused to the lumazine synthase from Brucella spp (BLS) and used it to generate anti-MICA polyclonal Ab (pAb) and to investigate if these anti-MICA Ab can reinstate antitumor immunity in mice using two different mouse tumors engineered to express MICA. We also explored the underlying mechanisms of this expected therapeutic effect. RESULTS: Immunization with BLS-MICA and administration of anti-MICA pAb elicited by BLS-MICA significantly delayed the growth of MICA-expressing mouse tumors but not of control tumors. The therapeutic effect of immunization with BLS-MICA included scavenging of sMICA and the anti-MICA Ab-mediated ADCC, promoting heightened intratumoral M1/proinflammatory macrophage and antigen-experienced CD8+ T cell recruitment. CONCLUSIONS: Immunization with the chimeric protein BLS-MICA constitutes a useful way to actively induce therapeutic anti-MICA pAb that resulted in a reprogramming of the antitumor immune response towards an antitumoral/proinflammatory phenotype. Hence, the BLS-MICA chimeric protein constitutes a novel antitumor vaccine of potential application in patients with MICA-expressing tumors.


Assuntos
Anticorpos Monoclonais/farmacologia , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Linfoma/imunologia , Proteínas Recombinantes de Fusão/imunologia , Neoplasias da Bexiga Urinária/imunologia , Animais , Brucella/enzimologia , Feminino , Antígenos de Histocompatibilidade Classe I/genética , Linfoma/patologia , Linfoma/terapia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/genética , Subfamília K de Receptores Semelhantes a Lectina de Células NK/imunologia , Células Tumorais Cultivadas , Neoplasias da Bexiga Urinária/patologia , Neoplasias da Bexiga Urinária/terapia
6.
J Cell Physiol ; 219(1): 132-42, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19097033

RESUMO

We examined the role of the extracellular signal regulated kinases (ERK) in 1,25-dihydroxyvitamin D (1,25(OH)(2)D(3))-induced gene expression in the differentiated Caco-2 cells. 1,25(OH)(2)D(3)-regulated expression of the 25-hydroxyvitamin D, 24-hydroxylase (CYP24) gene (both natural gene and promoter construct) was strongly modulated by altering ERK activity (i.e., reduced by MEK inhibitors and dominant negative (dn) ERK1 and ERK2, activated by epidermal growth factor) but ERK inhibition had no effect on 1,25(OH)(2)D(3)-regulated expression of the transient receptor potential cation channel, subfamily V, member 6 (TRPV6). ERK5-mediated phosphorylation of the transcription factor Ets-1 enhanced 1,25(OH)(2)D(3)-mediated CYP24 gene transcription in proliferating but not differentiated Caco-2 cells due to reduced levels of ERK5 and Ets-1 (total and phosphoprotein levels) in differentiated cells. MEK inhibition reduced 1,25(OH)(2)D(3)-induced 3X-VDRE promoter activity but had no impact on the association of vitamin D receptor (VDR) with chromatin suggesting a role for co-activator recruitment in ERK-modulation of vitamin D-regulated CYP24 gene activation. Chromatin immunoprecipitation assays revealed that the ERK1/2 target, mediator 1 (MED1), is recruited to the CYP24, but not the TRPV6, promoter following 1,25(OH)(2)D(3) treatment. MED1 phosphorylation was sensitive to activators and inhibitors of the ERK1/2 signaling and MED1 siRNA reduced 1,25(OH)(2)D(3)-regulated human CYP24 promoter activity. This suggests ERK1/2 signaling enhances 1,25(OH)(2)D(3) effects on the CYP24 promoter by MED1-mediated events. Our data show that there are both promoter-specific and cell stage-specific roles for the ERK signaling pathway on 1,25(OH)(2)D(3)-mediated gene induction in enterocyte-like Caco-2 cells.


Assuntos
Calcitriol/metabolismo , Regulação Enzimológica da Expressão Gênica , Sistema de Sinalização das MAP Quinases/fisiologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Esteroide Hidroxilases/genética , Esteroide Hidroxilases/metabolismo , Células CACO-2/fisiologia , Cálcio/metabolismo , Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Inibidores Enzimáticos/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Humanos , Imidazóis/metabolismo , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 1/genética , MAP Quinase Quinase 1/metabolismo , MAP Quinase Quinase 2/antagonistas & inibidores , MAP Quinase Quinase 2/genética , MAP Quinase Quinase 2/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/genética , Regiões Promotoras Genéticas , Proteína Proto-Oncogênica c-ets-1/genética , Proteína Proto-Oncogênica c-ets-1/metabolismo , Interferência de RNA , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Receptor X Retinoide alfa/genética , Receptor X Retinoide alfa/metabolismo , Esteroide Hidroxilases/antagonistas & inibidores , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo , Técnicas do Sistema de Duplo-Híbrido , Vitamina D3 24-Hidroxilase , Vitaminas/metabolismo
7.
Clin Cancer Res ; 14(13): 4316-25, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18594015

RESUMO

PURPOSE: Saccharomyces cerevisiae, a nonpathogenic yeast, has been used previously as a vehicle to elicit immune responses to foreign antigens, and tumor-associated antigens, and has been shown to reduce tumor burden in mice. Studies were designed to determine if vaccination of human carcinoembryonic antigen (CEA)-transgenic (CEA-Tg) mice (where CEA is a self-antigen) with a recombinant S. cerevisiae construct expressing human CEA (yeast-CEA) elicits CEA-specific T-cell responses and antitumor activity. EXPERIMENTAL DESIGN: CEA-Tg mice were vaccinated with yeast-CEA, and CD4(+) and CD8(+) T-cell responses were assessed after one and multiple administrations or vaccinations at multiple sites per administration. Antitumor activity was determined by tumor growth and overall survival in both pulmonary metastasis and s.c. pancreatic tumor models. RESULTS: These studies demonstrate that recombinant yeast can break tolerance and that (a) yeast-CEA constructs elicit both CEA-specific CD4(+) and CD8(+) T-cell responses; (b) repeated yeast-CEA administration causes increased antigen-specific T-cell responses after each vaccination; (c) vaccination with yeast-CEA at multiple sites induces a greater T-cell response than the same dose given at a single site; and (d) tumor-bearing mice vaccinated with yeast-CEA show a reduction in tumor burden and increased overall survival compared to mock-treated or control yeast-vaccinated mice in both pulmonary metastasis and s.c. pancreatic tumor models. CONCLUSIONS: Vaccination with a heat-killed recombinant yeast expressing the tumor-associated antigen CEA induces CEA-specific immune responses, reduces tumor burden, and extends overall survival in CEA-Tg mice. These studies thus form the rationale for the incorporation of recombinant yeast-CEA and other recombinant yeast constructs in cancer immunotherapy protocols.


Assuntos
Antígenos de Neoplasias/química , Antineoplásicos/farmacologia , Antígeno Carcinoembrionário/química , Regulação da Expressão Gênica , Imunoterapia/métodos , Saccharomyces cerevisiae/metabolismo , Vacinas de DNA/química , Animais , Antígenos de Neoplasias/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Vacinas Anticâncer , Antígeno Carcinoembrionário/metabolismo , Proliferação de Células , Feminino , Humanos , Linfócitos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Recombinantes/química , Vacinas de DNA/metabolismo
8.
J Nutr ; 138(1): 115-22, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18156413

RESUMO

The prevalence of obesity, an established risk factor for several chronic diseases, including cancer, has risen dramatically over the past 4 decades. Dietary change and/or increased physical activity are the most commonly recommended lifestyle-based strategies for preventing or reversing obesity. One of several physiological systems that may be enhanced by dietary change and exercise is the immune system. In this study, we examined the effects of energy restriction (ER; 30% reduction relative to control energy intake) and/or exercise (EX; voluntary wheel running) on systemic and mucosal immune function. Female C57BL/6 mice were randomized into 4 treatment conditions: 1) controls consumed ad libitum (AL); 2) AL with access to running wheels (AL + EX); 3) 30% ER; and 4) 30% ER with access to running wheels (ER + EX). Both ER and EX reduced spleen weight and the number of splenic T and B lymphocytes (P < 0.05). ER enhanced natural killer (NK) cell function, but reduced concanavalin A (Con A)-induced T-cell proliferation (P < 0.05). In contrast, EX enhanced Con A-induced proliferation and cytokine production from Peyer's patch cells (P < 0.05). These data suggest that ER and EX enhance some, but not all, components of the immune system and are likely working via different biological mechanisms to regulate NK and T-cell function.


Assuntos
Ingestão de Energia/fisiologia , Imunidade nas Mucosas/imunologia , Atividade Motora/fisiologia , Animais , Composição Corporal , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Tamanho do Órgão , Nódulos Linfáticos Agregados/citologia , Nódulos Linfáticos Agregados/metabolismo , Baço/anatomia & histologia , Linfócitos T
9.
Nat Rev Drug Discov ; 17(7): 509-527, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29904196

RESUMO

Immune cell functions are regulated by co-inhibitory and co-stimulatory receptors. The first two generations of cancer immunotherapy agents consist primarily of antagonist antibodies that block negative immune checkpoints, such as programmed cell death protein 1 (PD1) and cytotoxic T lymphocyte protein 4 (CTLA4). Looking ahead, there is substantial promise in targeting co-stimulatory receptors with agonist antibodies, and a growing number of these agents are making their way through various stages of development. This Review discusses the key considerations and potential pitfalls of immune agonist antibody design and development, their differentiating features from antagonist antibodies and the landscape of agonist antibodies in clinical development for cancer treatment.


Assuntos
Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Neoplasias/imunologia , Neoplasias/terapia , Animais , Antígeno CTLA-4/antagonistas & inibidores , Humanos , Imunoterapia/métodos , Terapia de Alvo Molecular/métodos , Receptor de Morte Celular Programada 1/antagonistas & inibidores
10.
Sci Transl Med ; 10(424)2018 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-29343622

RESUMO

Antibodies targeting immune checkpoints are emerging as potent and viable cancer therapies, but not all patients respond to these as single agents. Concurrently targeting additional immunosuppressive pathways is a promising approach to enhance immune checkpoint blockade, and bifunctional molecules designed to target two pathways simultaneously may provide a strategic advantage over the combination of two single agents. M7824 (MSB0011359C) is a bifunctional fusion protein composed of a monoclonal antibody against programmed death ligand 1 (PD-L1) fused to the extracellular domain of human transforming growth factor-ß (TGF-ß) receptor II, which functions as a "trap" for all three TGF-ß isoforms. We demonstrate that M7824 efficiently, specifically, and simultaneously binds PD-L1 and TGF-ß. In syngeneic mouse models, M7824 suppressed tumor growth and metastasis more effectively than treatment with either an anti-PD-L1 antibody or TGF-ß trap alone; furthermore, M7824 extended survival and conferred long-term protective antitumor immunity. Mechanistically, the dual anti-immunosuppressive function of M7824 resulted in activation of both the innate and adaptive immune systems, which contributed to M7824's antitumor activity. Finally, M7824 was an effective combination partner for radiotherapy or chemotherapy in mouse models. Collectively, our preclinical data demonstrate that simultaneous blockade of the PD-L1 and TGF-ß pathways by M7824 elicits potent and superior antitumor activity relative to monotherapies.


Assuntos
Anticorpos Monoclonais/imunologia , Receptor de Morte Celular Programada 1/imunologia , Fator de Crescimento Transformador beta/imunologia , Animais , Anticorpos Monoclonais/química , Imunoterapia/métodos , Camundongos , Receptor de Morte Celular Programada 1/química , Fator de Crescimento Transformador beta/química
11.
Front Biosci ; 12: 4997-5029, 2007 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-17569626

RESUMO

A number of lifestyle factors that reduce cancer risk in the primary prevention setting may be potential new targets for use in combination with cancer vaccines. This review discusses the modulation of energy balance (physical activity, calorie restriction, and obesity prevention), and the supplementation with natural and synthetic analogs of vitamins A and E, as potential interventions for use in combination with cancer vaccines. Additionally, the pharmacologic manipulation of nutrient metabolism in the tumor microenvironment (e.g., arachidonic acid, arginine, tryptophan, and glucose metabolism) is discussed. This review includes a brief overview of the role of each agent in primary cancer prevention; outlines the effects of these agents on immune function, specifically adaptive and/or anti-tumor immune mechanisms, when known; and discusses the potential use of these interventions in combination with therapeutic cancer vaccines. Modulation of energy balance through exercise and strategies targeting nutrient metabolism in the tumor microenvironment represent the most promising interventions to partner with therapeutic cancer vaccines. Additionally, the use of vitamin E succinate and the retinoid X receptor-directed rexinoids in combination with cancer vaccines offer promise. In summary, a number of energy balance- and nutrition-related interventions are viable candidates for further study in combination with cancer vaccines.


Assuntos
Vacinas Anticâncer , Dieta , Exercício Físico , Neoplasias/prevenção & controle , Terapia Combinada , Ingestão de Energia/imunologia , Humanos , Imunidade Inata , Neoplasias/complicações , Neoplasias/imunologia , Obesidade/complicações , Obesidade/imunologia , Fatores de Risco
12.
Breast Dis ; 22: 9-23, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16735783

RESUMO

Inflammatory breast cancer (IBC) is a rare and aggressive form of breast cancer with unknown etiology and generally poor outcome. It is characterized by diffuse edema (peau d'orange) and redness (erythema), although either the disease itself or case definitions have varied over time and place, confounding temporal trends and geographic variations. In this review, we discuss case definitions for IBC and its clinical characteristics; describe its geographic variation, age and racial distribution, incidence and survival patterns, and summarize the very limited information on its epidemiologic risk factors. We also incorporate emerging data from the National Cancer Institute's (NCI) Surveillance, Epidemiology, and End Results (SEER) Program.


Assuntos
Adenocarcinoma/diagnóstico , Adenocarcinoma/epidemiologia , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/epidemiologia , Adenocarcinoma/terapia , Adulto , Fatores Etários , Idoso , Biópsia por Agulha , Neoplasias da Mama/terapia , Terapia Combinada , Feminino , Humanos , Imuno-Histoquímica , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Prognóstico , Doenças Raras , Programa de SEER , Análise de Sobrevida , Estados Unidos/epidemiologia
13.
Mutat Res ; 576(1-2): 132-54, 2005 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-15888344

RESUMO

In recent years, investigators have carried out several studies designed to evaluate whether human tumor-associated antigens might be exploited as targets for active specific immunotherapy, specifically human cancer vaccines. Not too long ago such an approach would have been met with considerable skepticism because the immune system was believed to be a rigid discriminator between self and non-self which, in turn, protected the host from a variety of pathogens. That viewpoint has been challenged in recent years by a series of studies indicating that antigenic determinants of self have not induced absolute host immune tolerance. Moreover, under specific conditions that evoke danger signals, peptides from self-antigen can be processed by the antigen-presenting cellular machinery, loaded onto the major histocompatibility antigen groove to serve as targets for immune intervention. Those findings provide the rationale to investigate a wide range of tumor-associated antigens, including differentiation antigens, oncogenes, and tumor suppressor genes as possible immune-based targets. One of those tumor-associated antigens is the carcinoembryonic antigen (CEA). Described almost 40 years ago, CEA is a M(r) 180-200,000 oncofetal antigen that is one of the more widely studied human tumor-associated antigens. This review will provide: (i) a brief overview of the CEA gene family, (ii) a summary of early preclinical findings on overcoming immune tolerance to CEA, and (iii) the rationale to develop mouse models which spontaneously develop gastrointestinal tumors and express the CEA transgene. Those models have been used extensively in the study of overcoming host immune tolerance to CEA, a self, tumor-associated antigen, and the experimental findings have served as the rationale for the design of early clinical trials to evaluate CEA-based cancer vaccines.


Assuntos
Vacinas Anticâncer/uso terapêutico , Antígeno Carcinoembrionário/imunologia , Modelos Animais de Doenças , Neoplasias Experimentais/terapia , Animais , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Antígeno Carcinoembrionário/genética , Humanos , Camundongos , Camundongos Transgênicos , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/secundário , Taxa de Sobrevida
14.
Cancer Epidemiol Biomarkers Prev ; 21(2): 337-46, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22144502

RESUMO

BACKGROUND: Macrophage inhibitory cytokine-1 (MIC-1/GDF15) mediates nonsteroidal anti-inflammatory drug (NSAID) protection from colonic polyps in mice and is linked to the development of colorectal carcinoma in humans. Therefore, changes in serum MIC-1/GDF15 levels could predict the presence of premalignant colonic polyposis and assist in population screening strategies. METHODS: Serum MIC-1/GDF15 levels were measured in subjects in the Polyp Prevention Trial, in which NSAID use and colon cancer risk factors were defined. Subjects had an initial adenoma removed, a repeat colonoscopy removing previously unidentified polyps, and serum MIC-1/GDF15 estimation. Three years later recurrent adenomas were identified and serum MIC-1/GDF15 levels reestimated. The relationship between serum MIC-1/GDF15 levels and adenoma presence or recurrence was examined. RESULTS: Serum MIC-1/GDF15 levels differed by adenoma status and were significantly related to colon cancer risk factors. In addition, mean serum MIC-1/GDF15 levels rose with increasing numbers of adenomas present and high-risk adenoma recurrence. NSAID users had higher serum MIC-1/GDF15 concentrations, which were related to protection from adenoma recurrence. Furthermore, adjusted serum MIC-1/GDF15 levels at final follow-up were related to adenoma recurrence (highest quartile MIC-1/GDF15; OR = 14.7, 95% CI: 3.0-73). CONCLUSIONS: These data suggest that MIC-1/GDF15 mediates at least some of the protection afforded by NSAIDs against human colonic polyposis. Furthermore, serum MIC-1/GDF15 levels vary with the development of adnenomatous colonic polyps. IMPACT: Serum MIC-1/GDF15 determination may hold promise as the first serum screening test to assist the detection of premalignant adenomatous colonic polyposis.


Assuntos
Adenoma/prevenção & controle , Neoplasias do Colo/prevenção & controle , Fator 15 de Diferenciação de Crescimento/sangue , Adenoma/sangue , Adenoma/tratamento farmacológico , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Anti-Inflamatórios não Esteroides/uso terapêutico , Biomarcadores Tumorais/sangue , Neoplasias do Colo/sangue , Neoplasias do Colo/tratamento farmacológico , Pólipos do Colo/sangue , Pólipos do Colo/prevenção & controle , Feminino , Humanos , Masculino , Programas de Rastreamento , Camundongos , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/sangue , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/prevenção & controle , Fatores de Risco
15.
J Immunother ; 33(7): 697-705, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20664357

RESUMO

IL-12 is a potent antitumor cytokine that exhibits significant clinical toxicities after systemic administration. We hypothesized that intratumoral (i.t.) administration of IL-12 coformulated with the biodegradable polysaccharide chitosan could enhance the antitumor activity of IL-12 while limiting its systemic toxicity. Noninvasive imaging studies monitored local retention of IL-12, with and without chitosan coformulation, after i.t. injection. Antitumor efficacy of IL-12 alone and IL-12 coformulated with chitosan (chitosan/IL-12) was assessed in mice bearing established colorectal (MC32a) and pancreatic (Panc02) tumors. Additional studies involving depletion of immune cell subsets, tumor rechallenge, and CTL activity were designed to elucidate mechanisms of regression and tumor-specific immunity. Coformulation with chitosan increased local IL-12 retention from 1 to 2 days to 5 to 6 days. Weekly i.t. injections of IL-12 alone eradicated ≤10% of established MC32a and Panc02 tumors, while i.t. chitosan/IL-12 immunotherapy caused complete tumor regression in 80% to 100% of mice. Depletion of CD4(+) or Gr-1(+) cells had no impact on chitosan/IL-12-mediated tumor regression. However, CD8(+) or NK cell depletion completely abrogated antitumor activity. I.t. chitosan/IL-12 immunotherapy generated systemic tumor-specific immunity, as >80% of mice cured with i.t. chitosan/IL-12 immunotherapy were at least partially protected from tumor rechallenge. Furthermore, CTLs from spleens of cured mice lysed MC32a and gp70 peptide-loaded targets. Chitosan/IL-12 immunotherapy increased local retention of IL-12 in the tumor microenvironment, eradicated established, aggressive murine tumors, and generated systemic tumor-specific protective immunity. Chitosan/IL-12 is a well-tolerated, effective immunotherapy with considerable potential for clinical translation.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Quitosana/administração & dosagem , Neoplasias Colorretais/terapia , Imunoterapia , Neoplasias Pancreáticas/terapia , Animais , Antígeno Carcinoembrionário/genética , Células Cultivadas , Neoplasias Colorretais/imunologia , Citotoxicidade Imunológica/efeitos dos fármacos , Memória Imunológica , Interleucina-12/administração & dosagem , Interleucina-12/efeitos adversos , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transplante de Neoplasias , Neoplasias Pancreáticas/imunologia , Indução de Remissão , Carga Tumoral/efeitos dos fármacos
16.
Cancer Epidemiol Biomarkers Prev ; 19(6): 1441-52, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20501764

RESUMO

BACKGROUND: Serum adiponectin, leptin, C-peptide, and homocysteine are indicators for obesity, hyperinsulinemia, and chronic inflammation, which have all been associated with colorectal cancer. AIMS: To determine whether serum adiponectin, leptin, C-peptide, and homocysteine are associated with fat, fiber, fruit and vegetable, flavonol, or dry bean intake and colorectal adenoma recurrence. METHODS: Using logistic regression, we estimated odds ratios (OR) and 95% confidence intervals (95% CI) for adenoma recurrence in 627 participants from the control arm of the Polyp Prevention Trial, a 4-year trial that examined the effectiveness of a low-fat, high-fiber, high-fruit and vegetable diet on adenoma recurrence. RESULTS: Serum concentrations of C-peptide and homocysteine were inversely related to fiber, fruit and vegetable, and flavonol intake and positively related to percentage of calories from fat (all P(trend) < or = 0.01). High homocysteine concentrations were associated with any (4th versus 1st quartile: OR, 2.26; 95% CI, 1.30-3.94) and more than one adenoma recurrence (OR, 2.11; 95% CI, 1.01-4.40). Individuals in the highest, versus lowest, tertile of serum leptin concentration had a decreased risk of advanced adenoma recurrence (OR, 0.22; 95% CI, 0.06-0.79). CONCLUSION: Our results suggest that serum homocysteine may serve as an indicator of dietary exposure, including a low-fat and high-fiber, high-fruit and vegetable, and high-flavonol diet, as well as colorectal adenoma recurrence. IMPACT: Discovering biomarkers that are both modifiable and can predict cancer risk is critical. We identified serum homocysteine as a novel indicator that is modified by diet and predicts risk of adenoma recurrence.


Assuntos
Adenoma/sangue , Neoplasias Colorretais/sangue , Recidiva Local de Neoplasia/sangue , Adenoma/prevenção & controle , Adiponectina/sangue , Adulto , Idoso , Idoso de 80 Anos ou mais , Peptídeo C/sangue , Neoplasias Colorretais/prevenção & controle , Dieta , Feminino , Seguimentos , Homocisteína/sangue , Humanos , Leptina/sangue , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/patologia , Recidiva , Fatores de Risco
17.
Cancer Res ; 69(15): 6192-9, 2009 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-19638573

RESUMO

Intravesical BCG has been used successfully to treat superficial bladder cancer for three decades. However, 20% to 30% of patients will fail initial BCG therapy and 30% to 50% of patients will develop recurrent tumors within 5 years. Alternative or complementary strategies for the management of superficial bladder cancer are needed. Interleukin-12 (IL-12) is a potent T(H)1 cytokine with robust antitumor activity and the ability to potentiate immunologic memory. Unfortunately, intravesical IL-12 did not show antitumor efficacy in a recent clinical study of patients with recurrent superficial bladder cancer. We hypothesized that coformulation of IL-12 with chitosan, a biocompatible, mucoadhesive polysaccharide, could improve intravesical IL-12 delivery and provide an effective and durable alternative for the treatment of superficial bladder cancer. In antitumor studies, 88% to 100% of mice bearing orthotopic bladder tumors were cured after four intravesical treatments with chitosan/IL-12. In contrast, only 38% to 60% of mice treated with IL-12 alone and 0% treated with BCG were cured. Antitumor responses following chitosan/IL-12 treatments were durable and provided complete protection from intravesical tumor rechallenge. Urinary cytokine analysis showed that chitosan/IL-12 induced multiple T(H)1 cytokines at levels significantly higher than either IL-12 alone or BCG. Immunohistochemistry revealed moderate to intense tumor infiltration by T cells and macrophages following chitosan/IL-12 treatments. Bladder submucosa from cured mice contained residual populations of immune cells that returned to baseline levels after several months. Intravesical chitosan/IL-12 is a well-tolerated, effective immunotherapy that deserves further consideration for testing in humans for the management of superficial bladder cancer.


Assuntos
Carcinoma de Células de Transição/terapia , Quitosana/administração & dosagem , Interleucina-12/administração & dosagem , Neoplasias da Bexiga Urinária/terapia , Administração Intravesical , Animais , Vacina BCG/administração & dosagem , Carcinoma de Células de Transição/genética , Carcinoma de Células de Transição/imunologia , Linhagem Celular Tumoral , Feminino , Imuno-Histoquímica , Interferon gama/sangue , Interferon gama/urina , Interleucina-12/sangue , Interleucina-12/urina , Luciferases/biossíntese , Luciferases/genética , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Transfecção , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/imunologia
18.
Vaccine ; 26(42): 5407-15, 2008 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-18706954

RESUMO

Regular moderate exercise has been proposed to enhance immune function, but its effects on immunity and their consequences have not been well studied. Mice without (AL) or with access (AL+EX) to voluntary running wheels were vaccinated with a model antigen (ovalbumin (OVA)) via intranasal or subcutaneous routes to target the mucosal and systemic immune compartments, respectively. EX enhanced OVA-specific CD4(+) T cell cytokine production and proliferation in all lymphoid organs examined without changes in cell distribution in any organ. These results suggest that coupling moderate exercise with vaccination may enhance vaccine efficacy for the prevention and/or therapy of numerous diseases.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Atividade Motora , Vacinação , Animais , Composição Corporal , Linfócitos T CD4-Positivos/metabolismo , Proliferação de Células , Citocinas/biossíntese , Citocinas/imunologia , Feminino , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/imunologia , Especificidade por Substrato
19.
J Cell Biochem ; 100(3): 617-28, 2007 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-16960876

RESUMO

We examined the effects of 1,25 dihydroxyvitamin D(3) (1,25(OH)(2)D(3)) on the distribution and mobility of the vitamin D receptor (VDR) in the enterocyte-like Caco-2 cell. Confocal microscopy showed that a green fluorescent protein-vitamin D receptor (GFP-VDR) fusion protein is predominantly nuclear (58%) and it does not associate with the apical or basolateral membrane of proliferating or polarized, differentiated cells. In contrast to the previously studied cell types, neither endogenous VDR nor GFP-VDR levels accumulate in the nucleus following 1,25(OH)(2)D(3) treatment (100 nM, 30 min). However, in nuclear photobleaching experiments nuclear GFP-VDR import was significantly increased by 1,25(OH)(2)D(3) during both an early (0-5 min) and later (30-35 min) period (20% per 5 min). Compared to the natural ligand, nuclear import of GFP-VDR was 60% lower in cells treated with the 1,25(OH)(2)D(3) analog, 1-alpha-fluoro-16-ene-20-epi-23-ene-26,27-bishomo-25-hydroxyvitamin D(3) (Ro-26-9228, 5 min, 100 nM). Downstream events like ligand-induced association of VDR with chromatin at 1 h and the accumulation of CYP24 mRNA were significantly lower in Ro-26-9228 treated cells compared to 1,25(OH)(2)D(3) (60 and 95% lower, respectively). Collectively our data are consistent with a role for ligand-induced nuclear VDR import in receptor activation. In addition, ligand-dependent VDR nuclear import appears to be balanced by export, thus accounting for the lack of nuclear VDR accumulation even when VDR import is significantly elevated.


Assuntos
Colo/metabolismo , Receptores de Calcitriol/metabolismo , Sequência de Bases , Células CACO-2 , Calcitriol/farmacologia , Colo/citologia , Colo/efeitos dos fármacos , Primers do DNA , Proteínas de Fluorescência Verde/metabolismo , Humanos , Transporte Proteico , Transcrição Gênica
20.
Vaccine ; 25(11): 2085-94, 2007 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-17258843

RESUMO

The development of safe, novel adjuvants is necessary to maximize the efficacy of new and/or available vaccines. Chitosan is a non-toxic, biocompatible, biodegradable, natural polysaccharide derived from the exoskeletons of crustaceans and insects. Chitosan's biodegradability, immunological activity and high viscosity make it an excellent candidate as a depot/adjuvant for parenteral vaccination. To this end, we explored chitosan solution as an adjuvant for subcutaneous vaccination of mice with a model protein antigen. We found that chitosan enhanced antigen-specific antibody titers over five-fold and antigen-specific splenic CD4+ proliferation over six-fold. Strong increases in antibody titers together with robust delayed-type hypersensitivity (DTH) responses revealed that chitosan induced both humoral and cell-mediated immune responses. When compared with traditional vaccine adjuvants, chitosan was equipotent to incomplete Freund's adjuvant (IFA) and superior to aluminum hydroxide. Mechanistic studies revealed that chitosan exhibited at least two characteristics that may allow it to function as an immune adjuvant. First, the viscous chitosan solution created an antigen depot. More specifically, less than 9% of a protein antigen, when delivered in saline, remained at the injection site after 8 h. However, more than 60% of a protein antigen delivered in chitosan remained at the injection site for 7 days. Second, chitosan induced a transient 67% cellular expansion in draining lymph nodes. The expansion peaked between 14 and 21 days after chitosan injection and diminished as the polysaccharide was degraded. These mechanistic studies, taken together with the enhancement of a vaccine response, demonstrate that chitosan is a promising and safe platform for parenteral vaccine delivery.


Assuntos
Adjuvantes Imunológicos , Anticorpos/sangue , Quitosana/imunologia , Vacinação , Vacinas/imunologia , Hidróxido de Alumínio/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Quitosana/farmacocinética , Ensaio de Imunoadsorção Enzimática , Feminino , Adjuvante de Freund , Histocitoquímica , Hipersensibilidade Tardia , Injeções Subcutâneas , Lipídeos , Linfonodos/citologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Pele/patologia , Vacinas/administração & dosagem , beta-Galactosidase/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA